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:

Immunotherapies for hepatocellular carcinoma

Abstract

Liver cancer, more specifically hepatocellular carcinoma (HCC), is the second leading cause of cancer-related death and its incidence is increasing globally. Around 50% of patients with HCC receive systemic therapies, traditionally sorafenib or lenvatinib in the first line and regorafenib, cabozantinib or ramucirumab in the second line. In the past 5 years, immune-checkpoint inhibitors have revolutionized the management of HCC. The combination of atezolizumab and bevacizumab has been shown to improve overall survival relative to sorafenib, resulting in FDA approval of this regimen. More recently, durvalumab plus tremelimumab yielded superior overall survival versus sorafenib and atezolizumab plus cabozantinib yielded superior progression-free survival. In addition, pembrolizumab monotherapy and the combination of nivolumab plus ipilimumab have received FDA Accelerated Approval in the second-line setting based on early efficacy data. Despite these major advances, the molecular underpinnings governing immune responses and evasion remain unclear. The immune microenvironment has crucial roles in the development and progression of HCC and distinct aetiology-dependent immune features have been defined. Inflamed and non-inflamed classes of HCC and genomic signatures have been associated with response to immune-checkpoint inhibitors, yet no validated biomarker is available to guide clinical decision-making. This Review provides information on the immune microenvironments underlying the response or resistance of HCC to immunotherapies. In addition, current evidence from phase III trials on the efficacy, immune-related adverse events and aetiology-dependent mechanisms of response are described. Finally, we discuss emerging trials assessing immunotherapies across all stages of HCC that might change the management of this disease in the near future.

Key points

  • The composition of the hepatocellular carcinoma (HCC) immune microenvironment is the result of an interplay between immunosuppressive cells, immune effector cells, the cytokine milieu and tumour cell-intrinsic signalling pathways.

  • The aetiology of HCC influences the immune response and leads to unique microenvironmental features.

  • Combining anti-angiogenic drugs with immune-checkpoint inhibitors (ICIs) alters the tumour endothelium, thereby improving drug delivery and increasing the infiltration of effector immune cells.

  • Inhibiting vascular endothelial growth factor signalling also synergizes with ICIs by enhancing antitumour immune cell responses and inhibiting key immunosuppressive pathways.

  • A combination consisting of the ICI atezolizumab and the anti-angiogenic agent bevacizumab is the first treatment regimen that has been shown to improve overall survival in patients with advanced-stage HCC when compared with sorafenib.

  • Across all stages of HCC, almost 30 phase III trials testing immunotherapies (either alone or in combination) are currently ongoing and are likely to reshape the treatment landscape.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Overview of the roles of major immune cell types in the HCC tumour microenvironment.
Fig. 2: Context-dependent immune response in HCC.
Fig. 3: Cellular and molecular traits associated with distinct immune classes and subclasses of HCC.
Fig. 4: Mechanistic insights into the synergistic therapeutic activity of anti-angiogenic agents and ICIs.
Fig. 5: Natural history, current treatment algorithm and future prospects for immunotherapy in the management of HCC.

Similar content being viewed by others

References

  1. Sung, H. et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Ca. Cancer J. Clin. 71, 209–249 (2021).

    PubMed  Google Scholar 

  2. Llovet, J. M. et al. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 7, 6 (2021). This paper provides a comprehensive overview of the epidemiology, pathogenesis and management of HCC.

    PubMed  Google Scholar 

  3. Villanueva, A. Hepatocellular Carcinoma. N. Engl. J. Med. 380, 1450–1462 (2019).

    CAS  PubMed  Google Scholar 

  4. Llovet, J. M. et al. Locoregional therapies in the era of molecular and immune treatments for hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 18, 293–313 (2021).

    CAS  PubMed  Google Scholar 

  5. Llovet, J. M., Montal, R., Sia, D. & Finn, R. S. Molecular therapies and precision medicine for hepatocellular carcinoma. Nat. Rev. Clin. Oncol. 15, 599–616 (2018).

    PubMed  Google Scholar 

  6. Qin, S. et al. Donafenib versus sorafenib in first-line treatment of unresectable or metastatic hepatocellular carcinoma: a randomized, open-label, parallel-controlled phase II-III trial. J. Clin. Oncol. 39, 3002–3011 (2021).

    CAS  PubMed  Google Scholar 

  7. Finn, R. S. et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 382, 1894–1905 (2020). This trial is the first to demonstrate the superiority of an immunotherapy combination, atezolizumab–bevacizumab, over frontline sorafenib for advanced-stage HCC.

    CAS  PubMed  Google Scholar 

  8. Ren, Z. et al. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2–3 study. Lancet Oncol. 22, 977–990 (2021).

    CAS  PubMed  Google Scholar 

  9. Bruni, D., Angell, H. K. & Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Rev. Cancer 20, 662–680 (2020).

    CAS  PubMed  Google Scholar 

  10. Kalbasi, A. & Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 20, 25–39 (2020).

    CAS  PubMed  Google Scholar 

  11. Huinen, Z. R., Huijbers, E. J. M., van Beijnum, J. R., Nowak-Sliwinska, P. & Griffioen, A. W. Anti-angiogenic agents — overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat. Rev. Clin. Oncol. 18, 527–540 (2021). This Review discusses the main mechanisms supporting the synergism of anti-angiogenic drugs and immune-checkpoint inhibitors.

    PubMed  Google Scholar 

  12. Havel, J. J., Chowell, D. & Chan, T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 19, 133–150 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhu, A. X. et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): a non-randomised, open-label phase 2 trial. Lancet Oncol. 19, 940–952 (2018).

    PubMed  Google Scholar 

  14. Yau, T. et al. Efficacy and safety of nivolumab plus ipilimumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib. JAMA Oncol. 6, e204564 (2020). This study was the first to provide evidence of the efficacy of combining two immune-checkpoint inhibitors targeting CTLA4 (ipilimumab) and PD-1 (nivolumab) in patients with HCC.

    PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Hou, J., Zhang, H., Sun, B. & Karin, M. The immunobiology of hepatocellular carcinoma in humans and mice: basic concepts and therapeutic implications. J. Hepatol. 72, 167–182 (2020).

    CAS  PubMed  Google Scholar 

  17. Sia, D. et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology 153, 812–826 (2017). This study was the first to identify an immune-specific class of HCC with unique biological traits and potential implications for immunotherapy.

    CAS  PubMed  Google Scholar 

  18. Sangro, B., Sarobe, P., Hervás-Stubbs, S. & Melero, I. Advances in immunotherapy for hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2, 1–19 (2021).

    Google Scholar 

  19. Llovet, J. M. et al. Trial design and endpoints in hepatocellular carcinoma: AASLD consensus conference. Hepatology 73, 158–191 (2021).

    PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  21. Cancer Genome Atlas Research Network.Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell 169, 1327–1341.e23 (2017).

    Google Scholar 

  22. Zheng, C. et al. Landscape of Infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356.e16 (2017). This study was one of the first in-depth characterizations of the immune microenvironment of HCC at single-cell resolution.

    CAS  PubMed  Google Scholar 

  23. Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845.e20 (2019).

    CAS  PubMed  Google Scholar 

  24. Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, 450–456 (2021). This study demonstrates the existence of a CD8+PD-1+ subset of protumorigenic cells in non-alcoholic steatohepatitis that favour the development of HCC and hamper response to immune-checkpoint inhibitors.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hoechst, B. et al. A new population of myeloid-derived suppressor cells in hepatocellular carcinoma patients induces CD4+CD25+Foxp3+ T cells. Gastroenterology 135, 234–243 (2008).

    CAS  PubMed  Google Scholar 

  26. Kalathil, S., Lugade, A. A., Miller, A., Iyer, R. & Thanavala, Y. Higher frequencies of GARP+CTLA-4+Foxp3+ T regulatory cells and myeloid-derived suppressor cells in hepatocellular carcinoma patients are associated with impaired T-cell functionality. Cancer Res. 73, 2435–2444 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Han, Y. et al. Human CD14+CTLA-4+ regulatory dendritic cells suppress T-cell response by cytotoxic T-lymphocyte antigen-4-dependent IL-10 and indoleamine-2,3-dioxygenase production in hepatocellular carcinoma. Hepatology 59, 567–579 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Calderaro, J. et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma. J. Hepatol. 70, 58–65 (2019).

    PubMed  Google Scholar 

  30. Bruno, T. C. B cells to the forefront of immunotherapy. Nature 577, 474–476 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Dong, L. et al. Heterogeneous immunogenomic features and distinct escape mechanisms in multifocal hepatocellular carcinoma. J. Hepatol. 72, 896–908 (2020).

    CAS  PubMed  Google Scholar 

  32. Jhunjhunwala, S., Hammer, C. & Delamarre, L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021).

    CAS  PubMed  Google Scholar 

  33. Amsen, D., van Gisbergen, K. P. J. M., Hombrink, P. & van Lier, R. A. W. Tissue-resident memory T cells at the center of immunity to solid tumors. Nat. Immunol. 19, 538–546 (2018).

    CAS  PubMed  Google Scholar 

  34. Pallett, L. J. et al. Longevity and replenishment of human liver-resident memory T cells and mononuclear phagocytes. J. Exp. Med. 217, e20200050 (2020).

    PubMed  PubMed Central  Google Scholar 

  35. Tauber, C. et al. Inefficient induction of circulating TAA-specific CD8+ T-cell responses in hepatocellular carcinoma. Oncotarget 10, 5194–5206 (2019).

    PubMed  PubMed Central  Google Scholar 

  36. Payne, K. K. et al. BTN3A1 governs antitumor responses by coordinating αβ and γδ T cells. Science 369, 942–949 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Sun, Y. et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 184, 404–421.e16 (2021).

    CAS  PubMed  Google Scholar 

  38. Song, G. et al. Global immune characterization of HBV/HCV-related hepatocellular carcinoma identifies macrophage and T-cell subsets associated with disease progression. Cell Discov. 6, 90 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Zecca, A. et al. Energy metabolism and cell motility defect in NK-cells from patients with hepatocellular carcinoma. Cancer Immunol. Immunother. 69, 1589–1603 (2020).

    CAS  PubMed  Google Scholar 

  40. 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).

    CAS  PubMed  Google Scholar 

  41. Keenan, T. E., Burke, K. P. & Van Allen, E. M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 25, 389–402 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Craig, A. J. et al. Transcriptomic characterization of cancer-testis antigens identifies MAGEA3 as a driver of tumor progression in hepatocellular carcinoma. PLoS Genet. 17, e1009589 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Michler, T. et al. Knockdown of virus antigen expression increases therapeutic vaccine efficacy in high-titer hepatitis B virus carrier mice. Gastroenterology 158, 1762–1775.e9 (2020).

    CAS  PubMed  Google Scholar 

  44. Liu, D. et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 25, 1916–1927 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51, 202–206 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Rosenthal, R. et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567, 479–485 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Haber, P. K. et al. Molecular markers of response to anti-PD1 therapy in advanced hepatocellular carcinoma. J. Clin. Oncol. 39, 4110 (2021).

    Google Scholar 

  48. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Sci 355, eaaf8399 (2017).

    Google Scholar 

  49. Montironi, C. et al. Immune Classification of Hepatocellular Carcinoma Based on New Molecular Features: The Inflamed Class (EASL, 2020).

  50. Bassaganyas, L. et al. Copy-number alteration burden differentially impacts immune profiles and molecular features of hepatocellular carcinoma. Clin. Cancer Res. 26, 6350–6361 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhou, S.-L. et al. Overexpression of CXCL5 mediates neutrophil infiltration and indicates poor prognosis for hepatocellular carcinoma. Hepatology 56, 2242–2254 (2012).

    CAS  PubMed  Google Scholar 

  52. Liu, L.-Z. et al. CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology 69, 143–159 (2019).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Sangro, B. et al. Association of inflammatory biomarkers with clinical outcomes in nivolumab-treated patients with advanced hepatocellular carcinoma. J. Hepatol. 73, 1460–1469 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614.e14 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ruiz de Galarreta, M. et al. β-Catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 9, 1124–1141 (2019). This study demonstrates that β-catenin activation leads to immune evasion by interfering with dendritic cell chemotaxis.

    CAS  PubMed  Google Scholar 

  57. Cadoux, M. et al. Expression of NKG2D ligands is downregulated by β-catenin signalling and associates with HCC aggressiveness. J. Hepatol. 74, 1386–1397 (2021).

    CAS  PubMed  Google Scholar 

  58. Moeini, A. et al. An immune gene expression signature associated with development of human hepatocellular carcinoma identifies mice that respond to chemopreventive agents. Gastroenterology 157, 1383–1397.e11 (2019).

    CAS  PubMed  Google Scholar 

  59. Xu, Y. et al. Translation control of the immune checkpoint in cancer and its therapeutic targeting. Nat. Med. 25, 301–311 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Shen, J. et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 24, 556–562 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, J. et al. Epigenetic driver mutations in ARID1A shape cancer immune phenotype and immunotherapy. J. Clin. Invest. 130, 2712–2726 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhou, J. et al. Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut 67, 931–944 (2018).

    CAS  PubMed  Google Scholar 

  63. Chiang, D. Y. et al. Focal Gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res. 68, 6779–6788 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Maini, M. K. & Pallett, L. J. Defective T-cell immunity in hepatitis B virus infection: why therapeutic vaccination needs a helping hand. Lancet Gastroenterol. Hepatol. 3, 192–202 (2018).

    PubMed  Google Scholar 

  65. Kassel, R. et al. Chronically inflamed livers up-regulate expression of inhibitory B7 family members. Hepatology 50, 1625–1637 (2009).

    PubMed  Google Scholar 

  66. Wieland, D. et al. TCF1+ hepatitis C virus-specific CD8+ T cells are maintained after cessation of chronic antigen stimulation. Nat. Commun. 8, 15050 (2017).

    PubMed  PubMed Central  Google Scholar 

  67. Dudek, M. et al. Auto-aggressive CXCR6+CD8 T cells cause liver immune pathology in NASH. Nature 592, 444–449 (2021).

    CAS  PubMed  Google Scholar 

  68. 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 

  69. Mason, W. S. et al. HBV DNA integration and clonal hepatocyte expansion in chronic hepatitis B patients considered immune tolerant. Gastroenterology 151, 986–998.e4 (2016).

    CAS  PubMed  Google Scholar 

  70. Goto, K., Roca Suarez, A. A., Wrensch, F., Baumert, T. F. & Lupberger, J. Hepatitis C virus and hepatocellular carcinoma: when the host loses its grip. Int. J. Mol. Sci. 21, 3057 (2020).

    CAS  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Sitia, G. et al. Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc. Natl Acad. Sci. USA 109, E2165–E2172 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Tan, A. T. et al. Use of expression profiles of HBV-DNA integrated into genomes of hepatocellular carcinoma cells to select T cells for immunotherapy. Gastroenterology 156, 1862–1876.e9 (2019).

    CAS  PubMed  Google Scholar 

  74. Dunn, C. et al. Temporal analysis of early immune responses in patients with acute hepatitis B virus infection. Gastroenterology 137, 1289–1300 (2009).

    CAS  PubMed  Google Scholar 

  75. Das, A. et al. IL-10–producing regulatory B cells in the pathogenesis of chronic hepatitis B virus infection. J. Immunol. 189, 3925–3935 (2012).

    CAS  PubMed  Google Scholar 

  76. Xue, H. et al. Overrepresentation of IL-10-expressing B cells suppresses cytotoxic CD4+ T cell activity in HBV-induced hepatocellular carcinoma. PLoS ONE 11, e0154815 (2016).

    PubMed  PubMed Central  Google Scholar 

  77. Lim, C. J. et al. Multidimensional analyses reveal distinct immune microenvironment in hepatitis B virus-related hepatocellular carcinoma. Gut 68, 916–927 (2019).

    CAS  PubMed  Google Scholar 

  78. Heim, M. H. & Thimme, R. Innate and adaptive immune responses in HCV infections. J. Hepatol. 61, S14–S25 (2014).

    CAS  PubMed  Google Scholar 

  79. Semmo, N. et al. Preferential loss of IL-2-secreting CD4+ T helper cells in chronic HCV infection. Hepatology 41, 1019–1028 (2005).

    CAS  PubMed  Google Scholar 

  80. Cabrera, R. et al. An immunomodulatory role for CD4+CD25+ regulatory T lymphocytes in hepatitis C virus infection. Hepatology 40, 1062–1071 (2004).

    CAS  PubMed  Google Scholar 

  81. Abel, M. et al. Intrahepatic virus-specific IL-10-producing CD8 T cells prevent liver damage during chronic hepatitis C virus infection. Hepatology 44, 1607–1616 (2006).

    CAS  PubMed  Google Scholar 

  82. Hirano, J. et al. Hepatitis C virus modulates signal peptide peptidase to alter host protein processing. Proc. Natl Acad. Sci. USA 118, e2026184118 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Pallett, L. J. et al. Metabolic regulation of hepatitis B immunopathology by myeloid-derived suppressor cells. Nat. Med. 21, 591–600 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Jiang, Y. et al. Proteomics identifies new therapeutic targets of early-stage hepatocellular carcinoma. Nature 567, 257–261 (2019).

    CAS  PubMed  Google Scholar 

  85. Schmidt, N. M. et al. Targeting human Acyl-CoA:cholesterol acyltransferase as a dual viral and T cell metabolic checkpoint. Nat. Commun. 12, 2814 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Pikarsky, E. & Heikenwalder, M. Focal and local: ectopic lymphoid structures and aggregates of myeloid and other immune cells in liver. Gastroenterology 151, 780–783 (2016).

    PubMed  Google Scholar 

  87. 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).

    PubMed  Google Scholar 

  88. 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).

    CAS  PubMed  Google Scholar 

  89. Simon, T. G. et al. Association between aspirin use and risk of hepatocellular carcinoma. JAMA Oncol. 4, 1683 (2018).

    PubMed  PubMed Central  Google Scholar 

  90. 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 

  91. Ma, C. et al. NAFLD causes selective CD4+ T lymphocyte loss and promotes hepatocarcinogenesis. Nature 531, 253–257 (2016). This study shows that non-alcoholic fatty liver disease causes dysregulation of lipid metabolism, which leads to CD4+ T cell depletion and promotes hepatocarcinogenesis.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Ganne-Carrié, N. & Nahon, P. Hepatocellular carcinoma in the setting of alcohol-related liver disease. J. Hepatol. 70, 284–293 (2019).

    PubMed  Google Scholar 

  93. Parlesak, A., Schäfer, C., Schütz, T., Bode, J. C. & Bode, C. Increased intestinal permeability to macromolecules and endotoxemia in patients with chronic alcohol abuse in different stages of alcohol-induced liver disease. J. Hepatol. 32, 742–747 (2000).

    CAS  PubMed  Google Scholar 

  94. Gao, M. et al. Granulocytic myeloid-derived suppressor cell population increases with the severity of alcoholic liver disease. J. Cell. Mol. Med. 23, 2032–2041 (2019).

    CAS  PubMed  Google Scholar 

  95. Yan, G. et al. Chronic alcohol consumption promotes diethylnitrosamine-induced hepatocarcinogenesis via immune disturbances. Sci. Rep. 7, 2567 (2017).

    PubMed  PubMed Central  Google Scholar 

  96. Gao, B. & Bataller, R. Alcoholic liver disease: pathogenesis and new therapeutic targets. Gastroenterology 141, 1572–1585 (2011).

    CAS  PubMed  Google Scholar 

  97. Tilg, H., Moschen, A. R. & Szabo, G. Interleukin-1 and inflammasomes in alcoholic liver disease/acute alcoholic hepatitis and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Hepatology 64, 955–965 (2016).

    CAS  PubMed  Google Scholar 

  98. Ma, H.-Y. et al. IL-17 signaling in steatotic hepatocytes and macrophages promotes hepatocellular carcinoma in alcohol-related liver disease. J. Hepatol. 72, 946–959 (2020).

    CAS  PubMed  Google Scholar 

  99. Ponziani, F. R. et al. Hepatocellular carcinoma is associated with gut microbiota profile and inflammation in nonalcoholic fatty liver disease. Hepatology 69, 107–120 (2019).

    CAS  PubMed  Google Scholar 

  100. Schwabe, R. F. & Greten, T. F. Gut microbiome in HCC – mechanisms, diagnosis and therapy. J. Hepatol. 72, 230–238 (2020).

    CAS  PubMed  Google Scholar 

  101. Dapito, D. H. et al. Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4. Cancer Cell 21, 504–516 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Ma, C. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Sci 360, eaan5931 (2018).

    Google Scholar 

  103. Yoshimoto, S. et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499, 97–101 (2013).

    CAS  PubMed  Google Scholar 

  104. Thorsson, V. V. et al. The immune landscape of cancer. Immunity 48, 812–830.e14 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Montal, R. et al. Molecular classification and therapeutic targets in extrahepatic cholangiocarcinoma. J. Hepatol. 73, 315–327 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Job, S. et al. Identification of four immune subtypes characterized by distinct composition and functions of tumor microenvironment in intrahepatic cholangiocarcinoma. Hepatology 72, 965–981 (2020).

    CAS  PubMed  Google Scholar 

  107. Gay, C. M. et al. Patterns of transcription factor programs and immune pathway activation define four major subtypes of SCLC with distinct therapeutic vulnerabilities. Cancer Cell 39, 346–360.e7 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Nsengimana, J. et al. β-Catenin–mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J. Clin. Invest. 128, 2048–2063 (2018).

    PubMed  PubMed Central  Google Scholar 

  109. Chen, Y.-P. et al. Identification and validation of novel microenvironment-based immune molecular subgroups of head and neck squamous cell carcinoma: implications for immunotherapy. Ann. Oncol. 30, 68–75 (2019).

    PubMed  Google Scholar 

  110. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    PubMed  Google Scholar 

  111. Yokosuka, T. et al. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 209, 1201–1217 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Ahn, E. et al. Role of PD-1 during effector CD8 T cell differentiation. Proc. Natl Acad. Sci. USA 115, 4749–4754 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Maker, A. V., Attia, P. & Rosenberg, S. A. Analysis of the cellular mechanism of antitumor responses and autoimmunity in patients treated with CTLA-4 blockade. J. Immunol. 175, 7746–7754 (2005).

    CAS  PubMed  Google Scholar 

  114. Peggs, K. S., Quezada, S. A., Chambers, C. A., Korman, A. J. & Allison, J. P. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J. Exp. Med. 206, 1717–1725 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Agdashian, D. et al. The effect of anti-CTLA4 treatment on peripheral and intra-tumoral T cells in patients with hepatocellular carcinoma. Cancer Immunol. Immunother. 68, 599–608 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Calderaro, J. et al. Programmed death ligand 1 expression in hepatocellular carcinoma: relationship with clinical and pathological features. Hepatology 64, 2038–2046 (2016).

    CAS  PubMed  Google Scholar 

  117. Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350–1355 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Duan, J. et al. Use of immunotherapy with programmed cell death 1 vs programmed cell death ligand 1 inhibitors in patients with cancer. JAMA Oncol. 6, 375 (2020).

    PubMed  Google Scholar 

  119. Kurino, T. et al. Poor outcome with anti-programmed death-ligand 1 (PD-L1) antibody due to poor pharmacokinetic properties in PD-1/PD-L1 blockade-sensitive mouse models. J. Immunother. Cancer 8, e000400 (2020).

    PubMed  PubMed Central  Google Scholar 

  120. Yearley, J. H. et al. PD-L2 expression in human tumors: relevance to Anti-PD-1 therapy in cancer. Clin. Cancer Res. 23, 3158–3167 (2017).

    CAS  PubMed  Google Scholar 

  121. Butterfield, L. H. Immunotherapeutic strategies for hepatocellular carcinoma. Gastroenterology 127, S232–S241 (2004).

    CAS  PubMed  Google Scholar 

  122. Palmer, D. H. et al. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology 49, 124–132 (2009).

    PubMed  Google Scholar 

  123. Meng, W. S. et al.α-Fetoprotein-specific tumor immunity induced by plasmid prime-adenovirus boost genetic vaccination. Cancer Res. 61, 8782–8786 (2001).

    CAS  PubMed  Google Scholar 

  124. Butterfield, L. H., Ribas, A., Potter, D. M. & Economou, J. S. Spontaneous and vaccine induced AFP-specific T cell phenotypes in subjects with AFP-positive hepatocellular cancer. Cancer Immunol. Immunother. 56, 1931–1943 (2007).

    CAS  PubMed  Google Scholar 

  125. Greten, T. F. et al. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer 10, 209 (2010).

    PubMed  PubMed Central  Google Scholar 

  126. Goyal, L. et al. Abstract 3183: Initial safety of AFP SPEAR T-cells in patients with advanced hepatocellular carcinoma. Cancer Res. https://doi.org/10.1158/1538-7445.AM2019-3183 (2019).

    Article  Google Scholar 

  127. Dai, H. et al. Efficacy and biomarker analysis of CD133-directed CAR T cells in advanced hepatocellular carcinoma: a single-arm, open-label, phase II trial. Oncoimmunology 9, 1846926 (2020).

    PubMed  PubMed Central  Google Scholar 

  128. Shi, D. et al. Chimeric antigen receptor-glypican-3 T-cell therapy for advanced hepatocellular carcinoma: results of phase I trials. Clin. Cancer Res. 26, 3979–3989 (2020).

    CAS  PubMed  Google Scholar 

  129. Finn, R. S. et al. Phase Ib study of lenvatinib plus pembrolizumab in patients with unresectable hepatocellular carcinoma. J. Clin. Oncol. 38, 2960–2970 (2020).

    PubMed  PubMed Central  Google Scholar 

  130. Xu, J. et al. Camrelizumab in combination with apatinib in patients with advanced hepatocellular carcinoma (RESCUE): a nonrandomized, open-label, phase II trial. Clin. Cancer Res. 27, 1003–1011 (2021).

    CAS  PubMed  Google Scholar 

  131. Fukumura, D., Kloepper, J., Amoozgar, Z., Duda, D. G. & Jain, R. K. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat. Rev. Clin. Oncol. 15, 325–340 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Torrens, L. et al. Immunomodulatory effects of lenvatinib plus anti‐PD1 in mice and rationale for patient enrichment in hepatocellular carcinoma. Hepatology https://doi.org/10.1002/hep.32023 (2021).

    Article  PubMed  Google Scholar 

  133. Esteban-Fabró, R. et al. Cabozantinib enhances the efficacy and immune modulatory activity of anti-PD1 therapy in a syngeneic mouse model of hepatocellular carcinoma. J. Hepatol. 73, S40 (2020).

    Google Scholar 

  134. Sprinzl, M. F. et al. Sorafenib inhibits macrophage-induced growth of hepatoma cells by interference with insulin-like growth factor-1 secretion. J. Hepatol. 62, 863–870 (2015).

    CAS  PubMed  Google Scholar 

  135. Shigeta, K. et al. Regorafenib combined with PD1 blockade increases CD8 T-cell infiltration by inducing CXCL10 expression in hepatocellular carcinoma. J. Immunother. Cancer 8, e001435 (2020).

    PubMed  PubMed Central  Google Scholar 

  136. Zhuang, P.-Y. et al. Prognostic roles of cross-talk between peritumoral hepatocytes and stromal cells in hepatocellular carcinoma involving peritumoral VEGF-C, VEGFR-1 and VEGFR-3. PLoS ONE 8, e64598 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Ma, L. et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell 36, 418–430.e6 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Horwitz, E. et al. Human and Mouse VEGFA -amplified hepatocellular carcinomas are highly sensitive to sorafenib treatment. Cancer Discov. 4, 730–743 (2014).

    CAS  PubMed  Google Scholar 

  139. Zerbini, A. et al. Radiofrequency thermal ablation for hepatocellular carcinoma stimulates autologous NK-cell response. Gastroenterology 138, 1931–1942.e2 (2010).

    CAS  PubMed  Google Scholar 

  140. Lee, M. S. et al. Atezolizumab with or without bevacizumab in unresectable hepatocellular carcinoma (GO30140): an open-label, multicentre, phase 1b study. Lancet Oncol. 21, 808–820 (2020).

    CAS  PubMed  Google Scholar 

  141. Qin, S. et al. Camrelizumab in patients with previously treated advanced hepatocellular carcinoma: a multicentre, open-label, parallel-group, randomised, phase 2 trial. Lancet Oncol. 21, 571–580 (2020).

    CAS  PubMed  Google Scholar 

  142. Kelley, R. K. et al. Safety, efficacy, and pharmacodynamics of tremelimumab plus durvalumab for patients with unresectable hepatocellular carcinoma: randomized expansion of a phase I/II study. J. Clin. Oncol. 39, 2991–3001 (2021).

    CAS  PubMed  Google Scholar 

  143. Yen, C.-J. et al. Preliminary results of a phase 1A/1B study of BGB-A317, an anti-PD-1 monoclonal antibody (mAb), in patients with advanced hepatocellular carcinoma (HCC). Ann. Oncol. 28, iii54 (2017).

    Google Scholar 

  144. Ducreux, M. et al. O-1 Results from a global phase 2 study of tislelizumab, an investigational PD-1 antibody, in patients with unresectable hepatocellular carcinoma. Ann. Oncol. 32, S217 (2021).

    Google Scholar 

  145. Huang, A. C. et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 545, 60–65 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Topalian, S. L. et al. Five-year survival and correlates among patients with advanced melanoma, renal cell carcinoma, or non-small cell lung cancer treated with nivolumab. JAMA Oncol. 5, 1411–1420 (2019).

    PubMed  PubMed Central  Google Scholar 

  147. Takayama, T. et al. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet 356, 802–807 (2000).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    Google Scholar 

  150. Heimbach, J. K. et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 67, 358–380 (2018).

    PubMed  Google Scholar 

  151. Haber, P. K. et al. Evidence-based management of HCC: systematic review and meta-analysis of randomized controlled trials (2002-2020). Gastroenterology 161, 879–898 (2021).

    CAS  PubMed  Google Scholar 

  152. Topalian, S. L., Taube, J. M. & Pardoll, D. M. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science 367, eaax0182 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Liu, J. et al. Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov. 6, 1382–1399 (2016).

    CAS  PubMed  Google Scholar 

  154. Pinato, D. J., Fessas, P., Sapisochin, G. & Marron, T. U. Perspectives on the neoadjuvant use of immunotherapy in hepatocellular carcinoma. Hepatology 74, 483–490 (2021).

    PubMed  Google Scholar 

  155. Kaseb, A. O. et al. Immunologic correlates of pathologic complete response to preoperative immunotherapy in hepatocellular carcinoma. Cancer Immunol. Res. 7, 1390–1395 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Marron, T. U. et al. Abstract CT182: Neoadjuvant cemiplimab demonstrates complete pathological responses in hepatocellular carcinoma. Cancer Res. https://doi.org/10.1158/1538-7445.AM2021-CT182 (2021).

    Article  Google Scholar 

  157. Ho, W. J. et al. Neoadjuvant cabozantinib and nivolumab convert locally advanced hepatocellular carcinoma into resectable disease with enhanced antitumor immunity. Nat. Cancer 2, 891–903 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Greten, T. F., Mauda-Havakuk, M., Heinrich, B., Korangy, F. & Wood, B. J. Combined locoregional-immunotherapy for liver cancer. J. Hepatol. 70, 999–1007 (2019).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  160. 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).

    CAS  PubMed  Google Scholar 

  161. 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 

  162. El-Khoueiry, A. B. et al. Impact of antitumor activity on survival outcomes, and nonconventional benefit, with nivolumab (NIVO) in patients with advanced hepatocellular carcinoma (aHCC): subanalyses of CheckMate-040. J. Clin. Oncol. 36, 475 (2018).

    Google Scholar 

  163. Yau, T. et al. CheckMate 459: a randomized, multi-center phase III study of nivolumab (NIVO) vs sorafenib (SOR) as first-line (1L) treatment in patients (pts) with advanced hepatocellular carcinoma (aHCC). Ann. Oncol. 30, v874–v875 (2019).

    Google Scholar 

  164. Finn, R. S. et al. Pembrolizumab as second-line therapy in patients with advanced hepatocellular carcinoma in KEYNOTE-240: a randomized, double-blind, phase III trial. J. Clin. Oncol. 38, 193–202 (2020). This study was the first phase III trial assessing the efficacy of pembrolizumab, an anti-PD-1 antibody, in the second-line treatment of advanced-stage HCC.

    CAS  PubMed  Google Scholar 

  165. Bristol Myers Squibb Statement. Bristol Myers Squibb Statement on Opdivo® (nivolumab) Monotherapy Post-Sorafenib Hepatocellular Carcinoma U.S. Indication. https://news.bms.com/news/corporate-financial/2021/Bristol-Myers-Squibb-Statement-on-Opdivo-nivolumab-Monotherapy-Post-Sorafenib-Hepatocellular-Carcinoma-U.S.-Indication/default.aspx (2021).

  166. Merck. Merck Announces KEYTRUDA® (pembrolizumab) Met Primary Endpoint of Overall Survival (OS) in Patients with Advanced Hepatocellular Carcinoma Previously Treated with Sorafenib. Merck.com https://www.merck.com/news/merck-announces-keytruda-pembrolizumab-met-primary-endpoint-of-overall-survival-os-in-patients-with-advanced-hepatocellular-carcinoma-previously-treated-with-sorafenib/ (27 September 2021).

  167. Sangro, B. et al. LBA-3 CheckMate 459: long-term (minimum follow-up 33.6 months) survival outcomes with nivolumab versus sorafenib as first-line treatment in patients with advanced hepatocellular carcinoma. Ann. Oncol. 31, S241–S242 (2020).

    Google Scholar 

  168. Finn, R. S. et al. IMbrave150: updated overall survival (OS) data from a global, randomized, open-label phase III study of atezolizumab (atezo) + bevacizumab (bev) versus sorafenib (sor) in patients (pts) with unresectable hepatocellular carcinoma (HCC). J. Clin. Oncol. 39, 267 (2021).

    Google Scholar 

  169. Exelixis. Exelixis and Ipsen Announce Cabozantinib in Combination with an Immune Checkpoint Inhibitor Significantly Improved Progression-Free Survival in Phase 3 COSMIC-312 Pivotal Trial in Patients with Previously Untreated Advanced Liver Cancer. https://ir.exelixis.com/news-releases/news-release-details/exelixis-and-ipsen-announce-cabozantinib-combination-immune (2021).

  170. 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).

    PubMed  Google Scholar 

  171. Hu, J. et al. Image-guided percutaneous microwave ablation versus cryoablation for hepatocellular carcinoma in high-risk locations: intermediate-term results. Cancer Manag. Res. 11, 9801–9811 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Yu, J. et al. Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat. Med. 27, 152–164 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Ducreux, M. et al. IMbrave150: exploratory analysis to examine the association between treatment response and overall survival (OS) in patients (pts) with unresectable hepatocellular carcinoma (HCC) treated with atezolizumab (atezo) + bevacizumab (bev) versus sorafenib (sor). J. Clin. Oncol. 39 (Suppl. 15), 4071 (2021).

    Google Scholar 

  174. Enrico, D., Paci, A., Chaput, N., Karamouza, E. & Besse, B. Antidrug antibodies against immune checkpoint blockers: impairment of drug efficacy or indication of immune activation? Clin. Cancer Res. 26, 787–792 (2020).

    CAS  PubMed  Google Scholar 

  175. FDA. Highlights of Prescribing Information: TECENTRIQ. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761034s025lbl.pdf (2020).

  176. Pinato, D. J. et al. Clinical implications of heterogeneity in PD-L1 immunohistochemical detection in hepatocellular carcinoma: the Blueprint-HCC study. Br. J. Cancer 120, 1033–1036 (2019).

    PubMed  PubMed Central  Google Scholar 

  177. Hause, R. J., Pritchard, C. C., Shendure, J. & Salipante, S. J. Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 22, 1342–1350 (2016).

    CAS  PubMed  Google Scholar 

  178. Ang, C. et al. Prevalence of established and emerging biomarkers of immune checkpoint inhibitor response in advanced hepatocellular carcinoma. Oncotarget 10, 4018–4025 (2019).

    PubMed  PubMed Central  Google Scholar 

  179. Wong, C. N. et al. Qualification of tumour mutational burden by targeted next‐generation sequencing as a biomarker in hepatocellular carcinoma. Liver Int. 41, 192–203 (2021).

    CAS  PubMed  Google Scholar 

  180. 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).

    CAS  PubMed  Google Scholar 

  181. von Felden, J. et al. Mutations in circulating tumor DNA predict primary resistance to systemic therapies in advanced hepatocellular carcinoma. Oncogene 40, 140–151 (2021).

    Google Scholar 

  182. Zheng, Y. et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 7, 193 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Fessas, P. et al. Immunotoxicity from checkpoint inhibitor therapy: clinical features and underlying mechanisms. Immunology 159, 167–177 (2020).

    CAS  PubMed  Google Scholar 

  184. Puzanov, I. et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 5, 95 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Nogueira, E., Newsom-Davis, T. & Morganstein, D. L. Immunotherapy-induced endocrinopathies: assessment, management and monitoring. Ther. Adv. Endocrinol. Metab. 10, 204201881989618 (2019).

    Google Scholar 

  186. Brahmer, J. R. et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: american society of clinical oncology clinical practice guideline. J. Clin. Oncol. 36, 1714–1768 (2018). These guidelines provide detailed recommendations on the management of the most common immune-related adverse events.

    CAS  PubMed  Google Scholar 

  187. Pinato, D. J. et al. Treatment-related toxicity and improved outcome from immunotherapy in hepatocellular cancer: evidence from an FDA pooled analysis of landmark clinical trials with validation from routine practice. Eur. J. Cancer 157, 140–152 (2021).

    CAS  PubMed  Google Scholar 

  188. Dolladille, C. et al. Immune checkpoint inhibitor rechallenge after immune-related adverse events in patients with cancer. JAMA Oncol. 6, 865 (2020).

    PubMed  Google Scholar 

  189. Pinato, D. J. et al. Impact of corticosteroid therapy on the outcomes of hepatocellular carcinoma treated with immune checkpoint inhibitor therapy. J. Immunother. Cancer 8, e000726 (2020).

    PubMed  PubMed Central  Google Scholar 

  190. Gudd, C. L. C. et al. Activation and transcriptional profile of monocytes and CD8+ T cells are altered in checkpoint inhibitor-related hepatitis. J. Hepatol. 75, 177–189 (2021).

    CAS  PubMed  Google Scholar 

  191. Fessas, P. et al. Post-registration experience of nivolumab in advanced hepatocellular carcinoma: an international study. J. Immunother. Cancer 8, e001033 (2020).

    PubMed  PubMed Central  Google Scholar 

  192. Pinato, D. J. et al. Immunotherapy in hepatocellular cancer patients with mild to severe liver dysfunction: adjunctive role of the ALBI Grade. Cancers 12, 1862 (2020).

    CAS  PubMed Central  Google Scholar 

  193. Sangro, B. et al. Diagnosis and management of toxicities of immune checkpoint inhibitors in hepatocellular carcinoma. J. Hepatol. 72, 320–341 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  194. d’Izarny‐Gargas, T., Durrbach, A. & Zaidan, M. Efficacy and tolerance of immune checkpoint inhibitors in transplant patients with cancer: a systematic review. Am. J. Transplant. 20, 2457–2465 (2020).

    PubMed  Google Scholar 

  195. Gordan, J. D. et al. Systemic therapy for advanced hepatocellular carcinoma: ASCO guideline. J. Clin. Oncol. 38, 4317–4345 (2020).

    CAS  PubMed  Google Scholar 

  196. Bruix, J., Chan, S. L., Galle, P. R., Rimassa, L. & Sangro, B. Systemic treatment of hepatocellular carcinoma: An EASL position paper. J. Hepatol. 75, 960–974 (2021).

    CAS  PubMed  Google Scholar 

  197. Vogel, A. et al. Updated treatment recommendations for hepatocellular carcinoma (HCC) from the ESMO clinical practice guidelines. Ann. Oncol. 32, 801–805 (2021).

    CAS  PubMed  Google Scholar 

  198. Kudo, M. et al. CheckMate 040 cohort 5: a phase I/II study of nivolumab in patients with advanced hepatocellular carcinoma and Child-Pugh B cirrhosis. J. Hepatol. 75, 600–609 (2021).

    CAS  PubMed  Google Scholar 

  199. Siu, L. L. et al. Challenges and opportunities in adapting clinical trial design for immunotherapies. Clin. Cancer Res. 23, 4950–4958 (2017).

    PubMed  PubMed Central  Google Scholar 

  200. Ritchie, G. et al. Defining the most appropriate primary end point in phase 2 trials of immune checkpoint inhibitors for advanced solid cancers. JAMA Oncol. 4, 522 (2018).

    PubMed  PubMed Central  Google Scholar 

  201. Rimola, J. et al. Radiological response to nivolumab in patients with hepatocellular carcinoma: A multicenter analysis of real-life practice. Eur. J. Radiol. 135, 109484 (2021).

    PubMed  Google Scholar 

  202. Kim, C. G. et al. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J. Hepatol. 74, 350–359 (2021).

    CAS  PubMed  Google Scholar 

  203. Hodi, F. S. et al. Immune-modified response evaluation criteria in solid tumors (imRECIST): refining guidelines to assess the clinical benefit of cancer immunotherapy. J. Clin. Oncol. 36, 850–858 (2018).

    PubMed  Google Scholar 

  204. Anagnostou, V. et al. Immuno-oncology trial endpoints: capturing clinically meaningful activity. Clin. Cancer Res. 23, 4959–4969 (2017).

    PubMed  PubMed Central  Google Scholar 

  205. André, T. et al. Pembrolizumab in microsatellite-instability–high advanced colorectal cancer. N. Engl. J. Med. 383, 2207–2218 (2020).

    PubMed  Google Scholar 

  206. Martins, F. et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 16, 563–580 (2019).

    CAS  PubMed  Google Scholar 

  207. Pinato, D. J. et al. PRIME-HCC: phase Ib study of neoadjuvant ipilimumab and nivolumab prior to liver resection for hepatocellular carcinoma. BMC Cancer 21, 301 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Nordness, M. F. et al. Fatal hepatic necrosis after nivolumab as a bridge to liver transplant for HCC: Are checkpoint inhibitors safe for the pretransplant patient? Am. J. Transplant. 20, 879–883 (2020).

    PubMed  Google Scholar 

  209. Tabrizian, P. et al. A US multicenter analysis of 2529 HCC patients undergoing liver transplantation: 10‐year outcome assessing the role of down‐staging to within Milan criteria. Hepatology 70, 10A–11A (2019).

    Google Scholar 

  210. Mazzaferro, V. et al. Liver transplantation in hepatocellular carcinoma after tumour downstaging (XXL): a randomised, controlled, phase 2b/3 trial. Lancet Oncol. 21, 947–956 (2020).

    CAS  PubMed  Google Scholar 

  211. Chu, K. F. & Dupuy, D. E. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat. Rev. Cancer 14, 199–208 (2014).

    CAS  PubMed  Google Scholar 

  212. Chew, V. et al. Immune activation underlies a sustained clinical response to yttrium-90 radioembolisation in hepatocellular carcinoma. Gut 68, 335–346 (2019).

    CAS  PubMed  Google Scholar 

  213. Pinato, D. J. et al. A phase Ib study of pembrolizumab following trans-arterial chemoembolization (TACE) in hepatocellular carcinoma (HCC): PETAL. Ann. Oncol. 30, v288 (2019).

    Google Scholar 

  214. AstraZeneca. Imfinzi plus tremelimumab significantly improved overall survival in HIMALAYA Phase III trial in 1st-line unresectable liver cancer. astrazeneca.com, https://www.astrazeneca.com/media-centre/press-releases/2021/imfinzi-and-tremelimumab-improved-os-in-liver-cancer.html (15 October 2021).

  215. Yau, T. et al. Development of Hong Kong liver cancer staging system with treatment stratification for patients with hepatocellular carcinoma. Gastroenterology 146, 1691–1700.e3 (2014).

    PubMed  Google Scholar 

  216. Zhou, J. et al. Guidelines for the diagnosis and treatment of hepatocellular carcinoma (2019 Edition). Liver Cancer 9, 682–720 (2020).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The work of J.M.L. is supported by grants from the European Commission (EC) Horizon 2020 Programme (HEPCAR, proposal number 667273-2), the US NIH (RO1DK56621 and RO1DK128289), the Samuel Waxman Cancer Research Foundation, EIT Health (CRISH2, Ref. 18053) and the Spanish National Health Institute (MICINN, PID2019-105378RB-I00) through a partnership between Cancer Research UK (CRUK), Fondazione AIRC and Fundación Científica de la Asociación Española Contra el Cáncer (HUNTER, ref. C9380/A26813), and by the Generalitat de Catalunya (AGAUR, SGR-1358). The work of M.H. has been supported by a European Research Council (ERC) Consolidator grant (HepatoMetaboPath), the German Research Foundation (Deutsche Forschungsgemeinschaft; grants SFB/TR 209 Project-ID 314905040 and SFBTR1335 Project-ID 360372040), the Rainer Hoenig Foundation, a Horizon 2020 Programme grant (HEPCAR) and German Cancer Aid (Deutsche Krebshilfe; projects 70113166 and 70113167). M.K.M. acknowledges research support from the CRUK Immunology Project (grant 26603) and the CRUK Accelerator award HUNTER. D.P. acknowledges infrastructural support from the National Institute for Health Research (NIHR) Imperial Biomedical Research Centre and the Imperial Experimental Cancer Medicine Centre (ECMC), and grant funding from the Wellcome Trust Strategic Fund (PS3416), the ASCO Conquer Cancer Foundation Global Oncology Young Investigator Award 2019 (14704), CRUK (C57701/A26137), and by CW+ and the Westminster Medical School Research Trust (JRC SG 009 2018-19).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to all aspects of the preparation of this manuscript.

Corresponding author

Correspondence to Josep M. Llovet.

Ethics declarations

Competing interests

J.M.L. has received research support from Bayer HealthCare, Boehringer Ingelheim, Bristol Myers Squibb (BMS), Eisai and Ipsen, and consulting fees from AstraZeneca, Bayer HealthCare, BMS, Eisai, Eli Lilly, Exelixis, Genentech, Glycotest, Ipsen, Merck, Mina Alpha, Nucleix and Roche. M.K.M. is named co-inventor on international patent application No.1917498.6, entitled Treatment of Hepatitis B Virus (HBV) Infection, filed by UCL Business Ltd. M.K.M. has received unrestricted funding from Gilead, Immunocore and Roche, and advisory board/consulting fees from Abbvie, Freeline, Galapagos NV, Gilead, GlaxoSmithKline, Hoffmann La Roche, Immunocore and VIR. D.P. has received consulting fees from AstraZeneca, Bayer HealthCare, BMS, DaVolterra, Eisai, H3 Biomedicine, Ipsen, Mina Alpha, Roche and ViiV Healthcare, and research funding (to institution) from BMS and Merck Sharp & Dohme. The other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Clinical Oncology thanks A. Bertoletti, Q. Gao 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.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Llovet, J.M., Castet, F., Heikenwalder, M. et al. Immunotherapies for hepatocellular carcinoma. Nat Rev Clin Oncol 19, 151–172 (2022). https://doi.org/10.1038/s41571-021-00573-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41571-021-00573-2

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