Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Feb 24.
Published in final edited form as: Hepatology. 2020 Nov 7;73(Suppl 1):27–37. doi: 10.1002/hep.31481

Multiple roles for hepatitis B and C viruses and the host in the development of hepatocellular carcinoma

Kirk J Wangensteen 1, Kyong-Mi Chang 2
PMCID: PMC7855312  NIHMSID: NIHMS1633193  PMID: 32737895

Abstract

Chronic hepatitis B and C viral infections are major risk factors for hepatocellular carcinoma (HCC) in the United States and worldwide. Direct and indirect mechanisms of viral infection lead to the development of HCC. Chronic viral infection leads to inflammation and liver damage, culminating in cirrhosis, the penultimate step in the progression toward HCC. Host, viral, and environmental factors likely interact to promote oncogenesis. Clinical considerations include recommendations for screening for HCC in persons at risk, treatment with antivirals, and an emerging role for immunotherapy in HCC. We pose unanswered questions regarding HCC susceptibility and pathogenesis in the setting of chronic hepatitis B and C.

I. Introduction

Hepatitis B virus (HBV) and hepatitis C virus (HCV) cause persistent liver injury and regenerative responses that can progress to liver cirrhosis and hepatocellular carcinoma (HCC). HCC is an aggressive form of cancer that arises from hepatocytes, the main parenchymal cell type of the liver (1, 2). Globally, HCC ranks 6th in cancer incidence but 2nd in estimated number of cancer deaths in 2018, according to the International Agency for Research of the Cancer in the World Health Organization. In the United States, the incidence of HCC has been rising in the past decades, with 5 year survival estimated at only 18% (3). At least 60% of HCC can be attributed to underlying chronic hepatitis B (CHB) or chronic hepatitis C (CHC) (4). Here, we review mechanisms of HCC pathogenesis in the setting of CHB and CHC, and their relevance to clinical management and therapy. Figure 1 provides a schematic overview.

Figure: Multiple factors are involved in hepatocarcinogenesis with chronic HBV and HCV infection.

Figure:

The vast majority of hepatocellular carcinomas (HCCs) develop in the setting of cirrhosis, whereas a fraction of HCCs can develop without cirrhosis in association with HBV or AAV2 genomic viral integration events. Cirrhosis develops after many years of necroinflammatory responses to viral infections, and is accelerated by alcohol, viral co-infection, and fatty liver disease. Cirrhosis is associated with epigenetic changes that drive the development of HCC. The earliest somatic mutations linked to HCC include disruption of p53, activation of β-catenin, and/or promoter mutations that activate TERT. Progression is associated with increasing load of mutations in tumors. Additional factors associated with HCC include male sex, hereditary factors, viral factors, and environmental toxins such as aristolochic acids and aflatoxin.

II. Pathogenesis of HCC in the setting of viral infection

CHB and CHC share common pathways in HCC pathogenesis, related to chronic hepatocellular injury and turnover in the setting of inflammation. However, there are also important differences in mechanisms of infection, co-factors and host genetics.

A. Mechanisms for HCC common to HBV and HCV

Maladaptive immune activation promotes HCC:

Both HBV and HCV are non-cytopathic viruses. As such, hepatocellular injury during HBV or HCV infection is largely mediated by host immune responses (5). While both viruses circumvent innate immune responses in early infection, MHC class I-restricted CD8 T-cells mediate both hepatocellular injury and virus control by directly killing virus-infected hepatocytes and secreting antiviral cytokines such as IFN-gamma and TNF. In addition, B-cells control viral spread by secreting neutralizing antibodies, whereas MHC class II-restricted antiviral CD4 T-cells regulate the overall adaptive immune response that promotes the resolution of acute hepatitis and viremia.

In CHB and CHC, antiviral T-cells are tolerized or “exhausted” by persistent activation upon exposure to cognate viral antigens and ongoing inflammation with the induction of immune inhibitory receptors (e.g. PD-1 and CTLA-4), regulatory T-cells and cytokines (6, 7). CHB is also associated with virus-specific and global B-cell deficits (8, 9). In this setting, innate immune components including dendritic cells, Kupffer cells, and natural killer cells (NK-cells) can contribute to persistent inflammation and hepatocellular injury (10).

The maladaptive immune responses in the inflamed liver microenvironment can compromise effective anti-tumor immunity. For example, anti-tumor T and/or NK-cell responses are tamped down by myeloid-derived suppressor cells, regulatory T-cells, inhibitory receptors, and non-immune cells such as liver sinusoidal endothelial cells and hepatic stellate cells, all leading to tumor evasion of the immune system (11). NK-cells infiltrating human HCC can be metabolically deranged with mitochondrial fragmentation, reduced cytotoxicity and reduced survival (12). Furthermore, a critical role for platelets in hepatic inflammation and injury as well as HCC development was shown in a mouse model of CHB in which anti-platelet therapy reduced hepatic accumulation of HBV-specific CD8 T-cells and hepatocellular injury, prevented HCC and improved survival (13, 14).

Cytidine deaminases:

The cytidine deaminase family of enzymes is induced by interferons and inflammatory cytokines in response to viral infection as an innate defense mechanism (15). Among their members, APOBEC3A and APOBEC3B were shown to degrade HBV covalently closed circular DNA (cccDNA) (16). Cytidine deaminases have been implicated in promoting HBV mutations that enable immune evasion and associated with somatic mutations in human cancers (15). However, APOBEC mutational signature was not significantly correlated with HCC in a recent genetic analysis of different cancer types (17).

Hepatic regeneration and mutagenesis in the setting of chronic viral hepatitis:

Hepatocytes are not highly proliferative under normal circumstances, dividing every 100 days or less (18). However, chronic immune-mediated hepatocellular injury can lead to increased hepatocyte turnover by 3–5 times (19) with accumulation of hepatocytes with DNA damage (20, 21). Along these lines, the risk for HCC development is greater among patients with immune-active CHB with flares of hepatitis and high viremia as compared to inactive carriers without active inflammatory activity or cirrhosis (22), reflecting increased hepatocyte turnover and oxidative stress with the generation of reactive oxygen species (ROS) that promote mutagenesis (23).

Role of ER stress with unfolded protein response:

Viral assembly and maturation for HBV and HCV occur in the endoplasmic reticulum (ER). The production of large amounts of viral proteins can cause ER stress and activate the unfolded protein response (UPR) pathway. For example, HBsAg made in abundant quantities can accumulate in the ER of infected hepatocytes, cause their “ground-glass” appearance on H&E stain and activate the UPR, leading to DNA damage and genomic instability (24, 25). Indeed, overexpression of large HBsAg in the hepatocytes resulted in HCC in an HBV transgenic mouse model, with ER retention of large HBsAg with “ground glass” hepatocytes and increased hepatic inflammation, regenerative hyperplasia, transcriptional deregulation and aneuploidy (26, 27).

Similarly, HCV can activate the UPR due to HCV RNA replication in the ER and anchoring of viral factors on the ER membrane, with increased ROS and TGFβ1 expression that can contribute to liver fibrosis and HCC (28, 29).

Role of cirrhosis in transformation:

Chronic inflammation, necrosis, and liver regenerative responses from CHB and CHC result in cirrhosis—likely a critical step in the development of HCC as it is present in over 90% of incident cases. Cirrhosis is the end stage of hepatic fibrogenesis in which activated hepatic stellate cells transdifferentiate into myofibroblasts and deposit extracellular matrix factors, particularly collagen (30). While cirrhosis is associated with hepatocyte senescence with shortened telomeres and lack of telomerase activity, HCC development is associated with telomerase reactivation (31) and differential methylation patterns of hepatocyte genomic DNA including at TERT and TP53 loci (32). The epigenetic changes may promote tumorigenesis by blocking the expression of tumor suppressors and enabling expression of genes that allow cells to escape senescence (32, 33). Liver stiffness caused by fibrosis may directly impact hepatocyte function and cause a permissive microenvironment for early transformation (34).

B. HBV-specific features associated with HCC

HBV is a hepatotropic, partially double-stranded DNA virus within the Hepadnaviridae family, with overlapping open reading frames that encode HBV envelope, nucleocapsid, polymerase and X proteins. HBV enters hepatocytes through its receptor NTCP, a hepatocyte-specific bile acid transporter, and reaches the nucleus where viral replication and transcription take place with the cccDNA as the template for all HBV gene products (35). In patients with CHB, high HBV DNA levels, sero-positivity for HBV e antigen (HBeAg), and basal core promoter mutations have been associated with increased risk for HCC (36, 37). We provide select highlights regarding HBV-specific mechanisms of HCC development.

Integration into host genome:

HBV DNA sequences can integrate into the hepatocyte genome, becoming a permanent part of the cell and its progenitors (38), with potential contribution to tumorigenesis by inducing genomic instability and disrupting cancer related genes. HBV integration is more frequently detected in tumor tissues (86.4%) as compared to adjacent liver tissues (30.7%), and at multiple loci involving cancer-associated genes such as CCNA2, TERT, MLL4, KMT2B and CCNE1 (39, 40). The role for HBV integration in HCC pathogenesis is becoming clearer with advancements in sequencing technology, which has enabled analysis of integration of HBV in the genome. When HCC tissues were sampled multiple times and sequenced deeply, integration events were found in cancer-associated genes both in early “truncal” events, and in later subclonal events (32), suggesting a mechanistic role for HBV integration both in early tumor formation and its progression. In addition, integration of HBV into the genome has been linked to gene fusion events such as with LINE1, which was found in 23% of HBV-associated HCCs and is suspected to drive HCC (41). HBV integration can occur early in HBV infection, and with minimal associated liver inflammation (37), suggesting that early therapeutic intervention might minimize accumulation of genetic damage (42).

Interestingly, AAV2 viral integrations have also been detected in some of the same driver genes as HBV, suggesting that this virus may also play a mechanistic role in HCC formation (43). However, the absolute contribution to HCC by AAV is unclear.

HBV gene products with oncogenic potential:

There is some evidence for direct oncogenic potential for HBV. First, HBV infection of primary cells upregulates host factors involved in the cell cycle including PPARA, RXRA, and CEBPB, (44). Second, several HBV proteins have been implicated as potential “viral oncoproteins” that can directly drive HCC, including HBsAg, HBcAg, and X protein (HBx). In addition to HBsAg mentioned above, HBcAg variants have been found in populations at high risk of HCC, with increased viral replication and hepatocyte turnover with upregulation of genes associated with fibrosis and cancer, including MYC, when used to infect a mouse model repopulated with human hepatocytes (45). Furthermore, HBV X protein (HBx) is known for its generalized transcriptional activating properties and may drive tumorigenesis directly, although with variable results depending on the genetic background (reviewed in (35)). A truncated form of HBx is also seen in 46% HBV-derived HCC and can directly transform cell lines (46, 47). Further studies are needed to clarify the role of HBV proteins in directly promoting HCC.

C. HCV-specific features that are associated with HCC

Viral and host factors:

HCV is a hepatotropic positive-strand RNA virus with high sequence heterogeneity due to error-prone viral RNA polymerase (48). As an RNA virus, HCV cannot integrate into the genome. However, CHC can promote hepatocarcinogenesis by activating inflammatory and toxin-responsive pathways. The HCV gene products including HCV core protein, Core-E1-E2, NS3 and NS5 proteins have all been shown to generate ROS or alter miRNA expression, leading to hepatic steatohepatitis, injury, and/or turnover (reviewed in (49)). Of interest, miRNA122 is the most abundant miRNA in the liver and is required for the HCV life cycle (diverse roles of miRNA122 reviewed in (50)). An important role for miRNA122 as a tumor suppressor of HCC was shown in a mouse knock-out model deleted for the miRNA122 locus (51). Finally, HCC development was reported in transgenic mice with hepatic expression of HCV NS3 protein (52), in which steatohepatitis precedes adenoma and HCC development consistent with a progression from injury to carcinogenesis.

Iron and CHC:

CHC is associated with increased iron deposition in the liver, which increases ROS and promote HCC (53). Iron absorption into the body is controlled by hepcidin production by hepatocytes, and HCV was shown to down-regulate hepcidin, leading to iron accumulation in the liver (54). Surprisingly, hepatic iron overload and C282Y mutation was associated with increased HCC risk in patients with cirrhosis due to alcohol but not CHC (55).

D. Co-infections and Co-factors in HCC pathogenesis related to HBV and HCV infection:

Viral Coinfections:

Viral co-infections can impact the clinical course of CHB and CHC. Chronic co-infection with HBV and hepatitis delta virus (HDV) is associated with a more rapid progression to cirrhosis and HCC, with HDV large delta antigen proposed to induce an inflammatory response with increased endoplasmic reticulum stress and ROS (Reviewed in (56)). Due to similar routes of transmission, there is a greater prevalence of HCV and HBV coinfection among HIV-infected patients, with accelerated liver disease progression and HCC in the setting of HIV-associated immune dysregulation (57).

Dietary carcinogens and toxins:

Aflatoxin is produced by a mold found on peanuts and other crops, with high levels detected in the diet in Africa and China. A cooperative effect between aflatoxin and HBV has been suggested by epidemiological data in which geographic regions with high rates of HCC also show high rates of aflatoxin and CHB (58) and directly shown in HBV transgenic mice (59). HCCs from these regions have frequent mutations in TP53, particularly at codon 249 (58, 60). Aflatoxin is also linked to a specific mutational signature within HCC (33). Notably, African migrants to Europe with HCC were found to have a strong Aflatoxin mutational signature in their primary HCC tumors that dissipated in HCC subclones that developed in Europe, suggesting that the aflatoxin-specific risk diminishes when the exposure is removed (80).

Additional risk factors associated with worse outcomes in patients with HCC include aristolochic acids (61), alcohol use and smoking (62).

E. Host genetic factors

Germline associations:

A family history of liver cancer is an important risk factor for the development of HCC. Multiple independent groups have found that in patients with CHB or CHC, having a first degree relative with HCC doubles the risk of incident HCC as compared to those without family history of HCC (6365). This suggests that host genetic factors contribute to the risk for HCC, as well as potential shared environmental exposures among family members. Therefore, the American Association for the Study of Liver Diseases (AASLD) HBV guidelines recommend regular screening for HCC based on first-degree relative with HCC (66).

However, it is not clear whether germline variants in genes associated with hereditary cancer syndromes are linked to an increased risk of HCC in patients with CHB or CHC. An analysis of 363 HCCs in The Cancer Genome Atlas (TCGA) database, a collective at the National Cancer Institute, discovered one patient with CHB and a germline TERT promoter activating mutation (67). A few other germline pathogenic variants in cancer-associated genes were found in a separate analysis of the same TCGA cohort (68), but these did not reach statistically significant enrichment in HCC. Genome-wide association studies (GWAS) of HBV-associated HCC in different populations identified various loci associated with immunity and inflammation including a STAT4 variant with known association with autoimmunity, HLA class I/II, and CTLA-4, in addition to tumor suppressor genes and DNA repair genes, but with small odds ratios and inconsistent validation across studies (6972).

Somatic alterations:

HCC tissues from patients with CHB and CHC have been examined for changes in gene expression and for mutations (Reviewed in (33)). These analyses have been especially useful in improving our understanding of the mechanisms of hepatocarcinogenesis and in predicting outcomes. The most commonly mutated gene region in HCC is the promoter of TERT, which encodes telomerase, and is found in up to 90% of HCC (73). TERT mutations have been found in pre-neoplastic lesions in cirrhotic and non-cirrhotic livers (31). The next most common mutations occur in TP53 (inactivating) and CTNNB1 (activating) (33). Interestingly, the prevalent types of mutations have patterns of co-occurrence and mutual exclusivity. For example, mutations in CTNNB1 are usually not linked to mutations in TP53 or AXIN1. In contrast, CTNNB1 and TERT mutations are often found together. TP53 mutation usually coincides with mutations in KEAP1, TSC2, or amplification in CCND1.

Somatic Associations with HBV:

HCC associated with HBV infection is enriched in certain genetic changes that are also seen in HCCs of other etiologies (reviewed in (73)). HBV tends to have the most aggressive of HCC molecular subclasses. Non-cirrhotic HBV integration events are thought to contribute to the etiology for this subclass, as are TP53 mutations and chromosomal instability (74). HCCs arising from HBV tend to have activation of the AKT pathway (75), and to be negative for CTNNB1 mutation (74).

There are also specific epigenetic changes associated with HCCs that arise from HBV. Whole genome bisulfite sequencing demonstrated differential methylation of a number of genes including genes involved in glycolysis, and genes implicated in cancers including MDM2, FGF4, FGF19, HSP90AA1 (76). The HBx protein has been implicated in altering the methylation of genes by directly activating DNA methyltransferases, leading to global changes in DNA methylation (77, 78).

A recent study performed an integrated proteomic and genomic analysis of HCCs associated with CHB (79). This revealed distinct subtypes of HCCs in CHB, with differential mortality that can be identified based on differential expression of proteins PYCR2 and ADH1A, suggesting that these proteins could be used as biomarkers in HCC.

Somatic Associations with HCV:

Mutations in TERT and CTNNB1 are enriched in HCV-associated HCC, though these mutations are present in HCCs of all etiologies (31, 80). In fact, no mutation signature emerging from genomic studies has been found to be specific to HCV (33).

Epigenetic studies related to HCV infection have examined the effect of viral infection on gene transcriptional regulation. HCV induces the expression of PPP2CA, which leads to epigenetic changes stemming from deregulation of histone modifications (81). HCV infection also leads to GaDD45B promoter hypermethylation and silencing, which inhibits DNA repair responses (82).

III. Clinically relevant considerations in viral hepatitis associated HCC.

Anti-viral treatment:

Therapeutic virus suppression can enhance virus-specific T-cell function and improve hepatic inflammation, liver enzyme levels and regulatory pathways (83, 84), albeit without necessarily providing protective immunity. CHC is now readily cured with direct-acting antiviral (DAA) therapy, but patients can be re-infected. HBV therapy reduces the risk of HCC by more than 50% (85). Current options for HBV therapy are well tolerated with a high barrier to resistance, although without providing a “cure” and may therefore require prolonged therapy (reviewed in (66)). Hence, there is strong rationale to improve antiviral therapy to achieve functional cure of CHB (86). Notably, differential effects between tenofovir and entecavir in reducing the risk of HCC have been reported, and further prospective examination is warrented (8789).

Surveillance for HCC:

Patients at risk of HCC are recommended to enter into a surveillance program, typically with an ultrasound of the liver and/or alpha-fetoprotein level every six months (22, 66). The major determinant of risk is the presence of cirrhosis, with the risk of incident HCC in the setting of HBV or HCV-associated cirrhosis approaching levels as high as 3–8% per year. In geographic regions of Asia and Africa HCC occurs at a younger age and independently of cirrhosis, likely due to vertical transmission of HBV and environmental exposure to aflatoxin. Therefore, AASLD guidelines recommend screening all Asian and black men with HBV infection starting at age 40, and women at age 50 (66). Patients with HBV/HDV coinfection are also recommended to undergo regular surveillance for HCC regardless of whether or not they have cirrhosis.

HCC surveillance after achieving sustained virological response (SVR) in HCV:

When highly effective treatment for HCV became available with the arrival of DAAs, early studies reported higher rates of HCC than expected based on historical controls (reviewed in (90)), raising the hypothesis that anti-HCV inflammatory responses were also anti-tumorigenic and that its resolutionl facilitated HCC progression. However, subsequent studies showed reduced risk of all-cause mortality and incident HCC after HCV clearance, although patients with cirrhosis or advanced fibrosis may continue to have increased HCC risk for 10 years or more and need continued surveillance (91). Liver stiffness and harmful antiviral immune responses are reduced with HCV clearance, suggesting at least partial reversal of cirrhosis (91). As more patients who have achieved SVR are followed over time, it will be possible to define if/when HCC screening can stop, or whether any biomarkers can identify patients at persistently high risk for HCC.

Role of aspirin in chemoprevention:

Patients with CHB or CHC infection who take aspirin have been found to have a lower adjusted hazard ratio for the development of HCC (9294). The mechanism of the cancer-protective effect may involve its disruption of pathogenic platelet aggregations in the liver vasculature, as mentioned earlier (13). The use of aspirin as a chemopreventative agent has not yet been studied prospectively and may warrant further study.

Immunotherapy with Checkpoint Inhibitors:

Blockade of immune regulatory pathways (e.g. PD-1, CTLA-4, Tim-3) has been shown to be safe and effective in reactivating immunity against a number of tumor types, with some impressive clinical results (95, 96). PD-1 blockade in patients with HCC has shown efficacy in 15–20%, with similar efficacy across HBV, HCV, and non-viral etiologies, and with a manageable adverse effects profile (97). However, blocking these regulatory pathways could also activate antiviral immune responses and induce hepatocellular injury (98), as evidenced by ALT elevations following PD-1 blockade in several virally suppressed HBeAg-negative CHB patients (including one who achieved HBsAg loss) (99) and a subset of CHC patients (100). A recent phase III study combining anti-PD-L1 with VEGF inhibitor showed significantly better overall and progression-free survival compared to sorafenib, which represents the most promising advance in HCC therapy in over a decade (101). Nevertheless, grade 3 or 4 side effects occurred in over 50% of patients in both treatment arms, indicating a need for careful consideration torwards the underlying hepatic functional reserve and reducing hepatic viral burden with antiviral therapy.

Immunotherapy with Engineered T Cells:

An exciting breakthrough in cancer immunotherapy has been the development of engineered autologous ‘CAR T cells’ that express chimeric antigen receptor (CAR) that target tumor antigens (102). This approach can be extended to chronic viral infections, given the similarities in T-cell ‘exhaustion’ and metabolic derangements (103). In fact, limited adoptive transfer of HBsAg-specific CAR T-cells into a liver transplant recipient with metastatic HBV-associated HCC demonstrated the approach could be done safely, although clinical efficacy was not demonstrated (104). Further studies are needed before considering generalized use in HBV-infected patients given the potential for immune attack of hepatocytes.

IV. Closing statement and unanswered questions:

It is clear that viral and host factors contribute to the development of HCC, but a number of questions remain.

For one, it is generally accepted that multiple genomic mutations are needed to result in HCC. However, it is not clear at which point the genetic changes irreversibly lead to cancer, with no chance of return to quiescence. Furthermore, although cirrhosis is a readily identifiable major risk factor, approaches are needed to prevent HCC. In addition to anti-viral drug treatment, future treatments could target early cancer driver genes, reduce smoldering inflammatory responses, perhaps by aspirin, or reverse fibrosis.

An approach to identify targets genes in early HCC that is being explored using animal models of liver cancer development is to express HCC driver genes such as MYC or MET or HBx, and then to identify factors that are required for cancer progression in this setting. Such “synthetic lethal” factors could be targeted with drugs to prevent cancer progression (105).

Another area for future study is to perform risk prediction models in HCC. Larger GWAS and other types of genomics or proteomics studies are needed to determine whether genetic tests or biomarkers can identify patients at high risk for HCC. Such patients could benefit from more aggressive screening programs.

Financial Support

This work was supported by the National Institutes of Health (K08-DK106478 to K.J.W.) and by the Veterans Health Administration (K.M.C.).

Footnotes

Conflicts of Interest

Nothing to disclose

References

  • 1.Shin S, Wangensteen KJ, Teta-Bissett M, Wang YJ, Mosleh-Shirazi E, Buza EL, Greenbaum LE, et al. Genetic lineage tracing analysis of the cell of origin of hepatotoxin-induced liver tumors in mice. Hepatology 2016;64:1163–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Font-Burgada J, Shalapour S, Ramaswamy S, Hsueh B, Rossell D, Umemura A, Taniguchi K, et al. Hybrid Periportal Hepatocytes Regenerate the Injured Liver without Giving Rise to Cancer. Cell 2015;162:766–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Institute NC. Cancer Stat Facts: Liver and Intrahepatic Bile Duct Cancer. In; 2019.
  • 4.de Martel C, Maucort-Boulch D, Plummer M, Franceschi S. World-wide relative contribution of hepatitis B and C viruses in hepatocellular carcinoma. Hepatology 2015;62:1190–1200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bengsch B, Chang KM. Evolution in Our Understanding of Hepatitis B Virus Virology and Immunology. Clin Liver Dis 2016;20:629–644. [DOI] [PubMed] [Google Scholar]
  • 6.Rehermann B, Thimme R. Insights From Antiviral Therapy Into Immune Responses to Hepatitis B and C Virus Infection. Gastroenterology 2019;156:369–383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Chang KM, Liu M. Chronic hepatitis B: immune pathogenesis and emerging immunotherapeutics. Curr Opin Pharmacol 2016;30:93–105. [DOI] [PubMed] [Google Scholar]
  • 8.Salimzadeh L, Le Bert N, Dutertre CA, Gill US, Newell EW, Frey C, Hung M, et al. PD-1 blockade partially recovers dysfunctional virus-specific B cells in chronic hepatitis B infection. J Clin Invest 2018;128:4573–4587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Burton AR, Pallett LJ, McCoy LE, Suveizdyte K, Amin OE, Swadling L, Alberts E, et al. Circulating and intrahepatic antiviral B cells are defective in hepatitis B. J Clin Invest 2018;128:4588–4603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Maini MK, Gehring AJ. The role of innate immunity in the immunopathology and treatment of HBV infection. J Hepatol 2016;64:S60–S70. [DOI] [PubMed] [Google Scholar]
  • 11.Eggert T, Greten TF. Tumor regulation of the tissue environment in the liver. Pharmacol Ther 2017;173:47–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zheng X, Qian Y, Fu B, Jiao D, Jiang Y, Chen P, Shen Y, et al. Mitochondrial fragmentation limits NK cell-based tumor immunosurveillance. Nat Immunol 2019. [DOI] [PubMed] [Google Scholar]
  • 13.Sitia G, Aiolfi R, Di Lucia P, Mainetti M, Fiocchi A, Mingozzi F, Esposito A, et al. Antiplatelet therapy prevents hepatocellular carcinoma and improves survival in a mouse model of chronic hepatitis B. Proc Natl Acad Sci U S A 2012;109:E2165–2172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Iannacone M, Sitia G, Isogawa M, Marchese P, Castro MG, Lowenstein PR, Chisari FV, et al. Platelets mediate cytotoxic T lymphocyte-induced liver damage. Nat Med 2005;11:1167–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Deng Y, Du Y, Zhang Q, Han X, Cao G. Human cytidine deaminases facilitate hepatitis B virus evolution and link inflammation and hepatocellular carcinoma. Cancer Letters 2014;343:161–171. [DOI] [PubMed] [Google Scholar]
  • 16.Lucifora J, Xia Y, Reisinger F, Zhang K, Stadler D, Cheng X, Sprinzl MF, et al. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science 2014;343:1221–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Alexandrov LB, Kim J, Haradhvala NJ, Huang MN, Tian Ng AW, Wu Y, Boot A, et al. The repertoire of mutational signatures in human cancer. Nature 2020;578:94–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Taub R Liver regeneration: from myth to mechanism. Nat Rev Mol Cell Biol 2004;5:836–847. [DOI] [PubMed] [Google Scholar]
  • 19.Guo JT, Zhou H, Liu C, Aldrich C, Saputelli J, Whitaker T, Barrasa MI, et al. Apoptosis and regeneration of hepatocytes during recovery from transient hepadnavirus infections. J Virol 2000;74:1495–1505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nakamoto Y, Guidotti LG, Kuhlen CV, Fowler P, Chisari FV. Immune Pathogenesis of Hepatocellular Carcinoma. The Journal of Experimental Medicine 1998;188:341–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hagen TM, Huang S, Curnutte J, Fowler P, Martinez V, Wehr CM, Ames BN, et al. Extensive oxidative DNA damage in hepatocytes of transgenic mice with chronic active hepatitis destined to develop hepatocellular carcinoma. Proceedings of the National Academy of Sciences 1994;91:12808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Marrero JA, Kulik LM, Sirlin CB, Zhu AX, Finn RS, Abecassis MM, Roberts LR, et al. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018;68:723–750. [DOI] [PubMed] [Google Scholar]
  • 23.Chemin I, Zoulim F. Hepatitis B virus induced hepatocellular carcinoma. Cancer Lett 2009;286:52–59. [DOI] [PubMed] [Google Scholar]
  • 24.Hsieh Y-H, Su I-J, Wang H-C, Chang W-W, Lei H-Y, Lai M-D, Chang W-T, et al. Pre-S mutant surface antigens in chronic hepatitis B virus infection induce oxidative stress and DNA damage. Carcinogenesis 2004;25:2023–2032. [DOI] [PubMed] [Google Scholar]
  • 25.Li Y, Xia Y, Cheng X, Kleiner ED, Hewitt MS, Sproch J, Li T, et al. Hepatitis B Surface Antigen Activates Unfolded Protein Response in Forming Ground Glass Hepatocytes of Chronic Hepatitis B. Viruses 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Huang SN, Chisari FV. Strong, sustained hepatocellular proliferation precedes hepatocarcinogenesis in hepatitis B surface antigen transgenic mice. Hepatology 1995;21:620–626. [PubMed] [Google Scholar]
  • 27.Chisari FV, Klopchin K, Moriyama T, Pasquinelli C, Dunsford HA, Sell S, Pinkert CA, et al. Molecular pathogenesis of hepatocellular carcinoma in hepatitis B virus transgenic mice. Cell 1989;59:1145–1156. [DOI] [PubMed] [Google Scholar]
  • 28.Chusri P, Kumthip K, Hong J, Zhu C, Duan X, Jilg N, Fusco DN, et al. HCV induces transforming growth factor beta1 through activation of endoplasmic reticulum stress and the unfolded protein response. Sci Rep 2016;6:22487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lin W, Tsai WL, Shao RX, Wu G, Peng LF, Barlow LL, Chung WJ, et al. Hepatitis C virus regulates transforming growth factor beta1 production through the generation of reactive oxygen species in a nuclear factor kappaB-dependent manner. Gastroenterology 2010;138:2509–2518, 2518.e2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol 2017;14:397–411. [DOI] [PubMed] [Google Scholar]
  • 31.Nault JC, Ningarhari M, Rebouissou S, Zucman-Rossi J. The role of telomeres and telomerase in cirrhosis and liver cancer. Nat Rev Gastroenterol Hepatol 2019;16:544–558. [DOI] [PubMed] [Google Scholar]
  • 32.Ding X, He M, Chan AWH, Song QX, Sze SC, Chen H, Man MKH, et al. Genomic and Epigenomic Features of Primary and Recurrent Hepatocellular Carcinomas. Gastroenterology 2019;157:1630–1645 e1636. [DOI] [PubMed] [Google Scholar]
  • 33.Dhanasekaran R, Nault J-C, Roberts LR, Zucman-Rossi J. Genomic Medicine and Implications for Hepatocellular Carcinoma Prevention and Therapy. Gastroenterology 2019;156:492–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wells RG. Location, location, location: Cell-level mechanics in liver fibrosis. Hepatology 2016;64:32–33. [DOI] [PubMed] [Google Scholar]
  • 35.Neuveut C, Wei Y, Buendia MA. Mechanisms of HBV-related hepatocarcinogenesis. Journal of Hepatology 2010;52:594–604. [DOI] [PubMed] [Google Scholar]
  • 36.Yang HI, Lu SN, Liaw YF, You SL, Sun CA, Wang LY, Hsiao CK, et al. Hepatitis B e antigen and the risk of hepatocellular carcinoma. N Engl J Med 2002;347:168–174. [DOI] [PubMed] [Google Scholar]
  • 37.Chen CJ, Yang HI, Su J, Jen CL, You SL, Lu SN, Huang GT, et al. Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. JAMA 2006;295:65–73. [DOI] [PubMed] [Google Scholar]
  • 38.Levrero M, Zucman-Rossi J. Mechanisms of HBV-induced hepatocellular carcinoma. J Hepatol 2016;64:S84–S101. [DOI] [PubMed] [Google Scholar]
  • 39.Zhao LH, Liu X, Yan HX, Li WY, Zeng X, Yang Y, Zhao J, et al. Genomic and oncogenic preference of HBV integration in hepatocellular carcinoma. Nat Commun 2016;7:12992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Sung WK, Zheng H, Li S, Chen R, Liu X, Li Y, Lee NP, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat Genet 2012;44:765–769. [DOI] [PubMed] [Google Scholar]
  • 41.Lau C-C, Sun T, Ching Arthur KK, He M, Li J-W, Wong Alissa M, Co Ngai N, et al. Viral-Human Chimeric Transcript Predisposes Risk to Liver Cancer Development and Progression. Cancer Cell 2014;25:335–349. [DOI] [PubMed] [Google Scholar]
  • 42.Mason WS, Gill US, Litwin S, Zhou Y, Peri S, Pop O, Hong ML, et al. HBV DNA Integration and Clonal Hepatocyte Expansion in Chronic Hepatitis B Patients Considered Immune Tolerant. Gastroenterology 2016;151:986–998 e984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nault J-C, Datta S, Imbeaud S, Franconi A, Mallet M, Couchy G, Letouzé E, et al. Recurrent AAV2-related insertional mutagenesis in human hepatocellular carcinomas. Nature Genetics 2015;47:1187. [DOI] [PubMed] [Google Scholar]
  • 44.Xia Y, Cheng X, Li Y, Valdez K, Chen W, Liang TJ. Hepatitis B Virus Deregulates the Cell Cycle To Promote Viral Replication and a Premalignant Phenotype. J Virol 2018;92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ligat G, Schuster C, Baumert TF. Hepatitis B Virus Core Variants, Liver Fibrosis, and Hepatocellular Carcinoma. Hepatology 2019;69:5–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ma N-F, Lau SH, Hu L, Xie D, Wu J, Yang J, Wang Y, et al. COOH-Terminal Truncated HBV X Protein Plays Key Role in Hepatocarcinogenesis. Clinical Cancer Research 2008;14:5061. [DOI] [PubMed] [Google Scholar]
  • 47.Sze KMF, Chu GKY, Lee JMF, Ng IOL. C-terminal truncated hepatitis B virus x protein is associated with metastasis and enhances invasiveness by c-jun/matrix metalloproteinase protein 10 activation in hepatocellular carcinoma. Hepatology 2013;57:131–139. [DOI] [PubMed] [Google Scholar]
  • 48.Rodrigo C, Leung P, Lloyd AR, Bull RA, Luciani F, Grebely J, Dore GJ, et al. Genomic variability of within-host hepatitis C variants in acute infection. Journal of Viral Hepatitis 2019;26:476–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Microbiome and cancer. New York, NY: Springer Science+Business Media, 2019: pages cm. [Google Scholar]
  • 50.Bandiera S, Pfeffer S, Baumert TF, Zeisel MB. miR-122 – A key factor and therapeutic target in liver disease. Journal of Hepatology 2015;62:448–457. [DOI] [PubMed] [Google Scholar]
  • 51.Tsai W-C, Hsu S-D, Hsu C-S, Lai T-C, Chen S-J, Shen R, Huang Y, et al. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. The Journal of Clinical Investigation 2012;122:2884–2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Moriya K, Fujie H, Shintani Y, Yotsuyanagi H, Tsutsumi T, Ishibashi K, Matsuura Y, et al. The core protein of hepatitis C virus induces hepatocellular carcinoma in transgenic mice. Nat Med 1998;4:1065–1067. [DOI] [PubMed] [Google Scholar]
  • 53.Fujinaga H, Tsutsumi T, Yotsuyanagi H, Moriya K, Koike K. Hepatocarcinogenesis in hepatitis C: HCV shrewdly exacerbates oxidative stress by modulating both production and scavenging of reactive oxygen species. Oncology 2011;81 Suppl 1:11–17. [DOI] [PubMed] [Google Scholar]
  • 54.Trinder D, Ayonrinde OT, Olynyk JK. HCV, iron, and oxidative stress: the new choreography of hepcidin. Gastroenterology 2008;134:348–351. [DOI] [PubMed] [Google Scholar]
  • 55.Nahon P, Sutton A, Rufat P, Ziol M, Thabut G, Schischmanoff PO, Vidaud D, et al. Liver iron, HFE gene mutations, and hepatocellular carcinoma occurrence in patients with cirrhosis. Gastroenterology 2008;134:102–110. [DOI] [PubMed] [Google Scholar]
  • 56.Koh C, Heller T, Glenn JS. Pathogenesis of and New Therapies for Hepatitis D. Gastroenterology 2019;156:461–476 e461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bruno R, Puoti M, Sacchi P, Filice C, Carosi G, Filice G. Management of hepatocellular carcinoma in human immunodeficiency virus-infected patients. J Hepatol 2006;44:S146–150. [DOI] [PubMed] [Google Scholar]
  • 58.Ozturk M. p53 mutation in hepatocellular carcinoma after aflatoxin exposure. Lancet 1991;338:1356–1359. [DOI] [PubMed] [Google Scholar]
  • 59.Sell S, Hunt JM, Dunsford HA, Chisari FV. Synergy between hepatitis B virus expression and chemical hepatocarcinogens in transgenic mice. Cancer Res 1991;51:1278–1285. [PubMed] [Google Scholar]
  • 60.Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, Couchy G, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nature Genetics 2015;47:505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ng AWT, Poon SL, Huang MN, Lim JQ, Boot A, Yu W, Suzuki Y, et al. Aristolochic acids and their derivatives are widely implicated in liver cancers in Taiwan and throughout Asia. Science Translational Medicine 2017;9:eaan6446. [DOI] [PubMed] [Google Scholar]
  • 62.Kuper H, Tzonou A, Kaklamani E, Hsieh CC, Lagiou P, Adami HO, Trichopoulos D, et al. Tobacco smoking, alcohol consumption and their interaction in the causation of hepatocellular carcinoma. Int J Cancer 2000;85:498–502. [PubMed] [Google Scholar]
  • 63.Turati F, Edefonti V, Talamini R, Ferraroni M, Malvezzi M, Bravi F, Franceschi S, et al. Family history of liver cancer and hepatocellular carcinoma. Hepatology 2012;55:1416–1425. [DOI] [PubMed] [Google Scholar]
  • 64.Hassan MM, Spitz MR, Thomas MB, Curley SA, Patt YZ, Vauthey JN, Glover KY, et al. The association of family history of liver cancer with hepatocellular carcinoma: a case-control study in the United States. J Hepatol 2009;50:334–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Loomba R, Liu J, Yang HI, Lee MH, Lu SN, Wang LY, Iloeje UH, et al. Synergistic effects of family history of hepatocellular carcinoma and hepatitis B virus infection on risk for incident hepatocellular carcinoma. Clin Gastroenterol Hepatol 2013;11:1636–1645 e1631–1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Terrault NA, Lok ASF, McMahon BJ, Chang K-M, Hwang JP, Jonas MM, Brown RS Jr, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD 2018 hepatitis B guidance. Hepatology 2018;67:1560–1599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017;169:1327–1341.e1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Huang KL, Mashl RJ, Wu Y, Ritter DI, Wang J, Oh C, Paczkowska M, et al. Pathogenic Germline Variants in 10,389 Adult Cancers. Cell 2018;173:355–370 e314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sawai H, Nishida N, Khor S-S, Honda M, Sugiyama M, Baba N, Yamada K, et al. Genome-wide association study identified new susceptible genetic variants in HLA class I region for hepatitis B virus-related hepatocellular carcinoma. Scientific Reports 2018;8:7958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Jiang DK, Sun J, Cao G, Liu Y, Lin D, Gao YZ, Ren WH, et al. Genetic variants in STAT4 and HLA-DQ genes confer risk of hepatitis B virus-related hepatocellular carcinoma. Nat Genet 2013;45:72–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Li S, Qian J, Yang Y, Zhao W, Dai J, Bei JX, Foo JN, et al. GWAS identifies novel susceptibility loci on 6p21.32 and 21q21.3 for hepatocellular carcinoma in chronic hepatitis B virus carriers. PLoS Genet 2012;8:e1002791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.An P, Xu J, Yu Y, Winkler CA. Host and Viral Genetic Variation in HBV-Related Hepatocellular Carcinoma. Frontiers in Genetics 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Levrero M, Zucman-Rossi J. Mechanisms of HBV-induced hepatocellular carcinoma. J Hepatol 2016;64:S84–101. [DOI] [PubMed] [Google Scholar]
  • 74.Laurent–Puig P, Legoix P, Bluteau O, Belghiti J, Franco D, Binot F, Monges G, et al. Genetic alterations associated with hepatocellular carcinomas define distinct pathways of hepatocarcinogenesis. Gastroenterology 2001;120:1763–1773. [DOI] [PubMed] [Google Scholar]
  • 75.Boyault S, Rickman DS, de Reyniès A, Balabaud C, Rebouissou S, Jeannot E, Hérault A, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology 2007;45:42–52. [DOI] [PubMed] [Google Scholar]
  • 76.Ye C, Tao R, Cao Q, Zhu D, Wang Y, Wang J, Lu J, et al. Whole-genome DNA methylation and hydroxymethylation profiling for HBV-related hepatocellular carcinoma. Int J Oncol 2016;49:589–602. [DOI] [PubMed] [Google Scholar]
  • 77.Park IY, Sohn BH, Yu E, Suh DJ, Chung YH, Lee JH, Surzycki SJ, et al. Aberrant Epigenetic Modifications in Hepatocarcinogenesis Induced by Hepatitis B Virus X Protein. Gastroenterology 2007;132:1476–1494. [DOI] [PubMed] [Google Scholar]
  • 78.Zheng D-L, Zhang L, Cheng N, Xu X, Deng Q, Teng X-M, Wang K-S, et al. Epigenetic modification induced by hepatitis B virus X protein via interaction with de novo DNA methyltransferase DNMT3A. Journal of Hepatology 2009;50:377–387. [DOI] [PubMed] [Google Scholar]
  • 79.Gao Q, Zhu H, Dong L, Shi W, Chen R, Song Z, Huang C, et al. Integrated Proteogenomic Characterization of HBV-Related Hepatocellular Carcinoma. Cell 2019;179:561–577 e522. [DOI] [PubMed] [Google Scholar]
  • 80.Pezzuto F, Izzo F, Buonaguro L, Annunziata C, Tatangelo F, Botti G, Buonaguro FM, et al. Tumor specific mutations in TERT promoter and CTNNB1 gene in hepatitis B and hepatitis C related hepatocellular carcinoma. Oncotarget 2016;7:54253–54262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Duong FH, Christen V, Lin S, Heim MH. Hepatitis C virus-induced up-regulation of protein phosphatase 2A inhibits histone modification and DNA damage repair. Hepatology 2010;51:741–751. [DOI] [PubMed] [Google Scholar]
  • 82.Higgs MR, Lerat H, Pawlotsky J-M. Downregulation of Gadd45β Expression by Hepatitis C Virus Leads to Defective Cell Cycle Arrest. Cancer Research 2010;70:4901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Boni C, Bertoletti A, Penna A, Cavalli A, Pilli M, Urbani S, Scognamiglio P, et al. Lamivudine treatment can restore T cell responsiveness in chronic hepatitis B. J Clin Invest 1998;102:968–975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Serti E, Chepa-Lotrea X, Kim YJ, Keane M, Fryzek N, Liang TJ, Ghany M, et al. Successful Interferon-Free Therapy of Chronic Hepatitis C Virus Infection Normalizes Natural Killer Cell Function. Gastroenterology 2015;149:190–200 e192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Singal AK, Salameh H, Kuo YF, Fontana RJ. Meta-analysis: the impact of oral anti-viral agents on the incidence of hepatocellular carcinoma in chronic hepatitis B. Alimentary Pharmacology & Therapeutics 2013;38:98–106. [DOI] [PubMed] [Google Scholar]
  • 86.Block TM, Alter H, Brown N, Brownstein A, Brosgart C, Chang KM, Chen PJ, et al. Research priorities for the discovery of a cure for chronic hepatitis B: Report of a workshop. Antiviral Res 2018;150:93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lampertico P, Papatheodoridis GV. Is Tenofovir Superior to Entecavir in Reducing the Risk of Hepatocellular Carcinoma in Chronic Hepatitis B? The Controversy Continues. Gastroenterology 2020;158:42–44. [DOI] [PubMed] [Google Scholar]
  • 88.Yip TC, Wong VW, Chan HL, Tse YK, Lui GC, Wong GL. Tenofovir Is Associated With Lower Risk of Hepatocellular Carcinoma Than Entecavir in Patients With Chronic HBV Infection in China. Gastroenterology 2020;158:215–225 e216. [DOI] [PubMed] [Google Scholar]
  • 89.Flemming JA, Terrault NA. Tenofovir vs Entecavir for Hepatocellular Carcinoma Prevention in Patients With Chronic Hepatitis B: One of These Things Is Not Like the Other. JAMA Oncology 2019;5:17–18. [DOI] [PubMed] [Google Scholar]
  • 90.Su F, Ioannou GN. Hepatocellular Carcinoma Risk After Direct-Acting Antiviral Therapy. Clinical Liver Disease 2019;13:6–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ioannou GN, Feld JJ. What Are the Benefits of a Sustained Virologic Response to Direct-Acting Antiviral Therapy for Hepatitis C Virus Infection? Gastroenterology 2019;156:446–460.e442. [DOI] [PubMed] [Google Scholar]
  • 92.Simon TG, Duberg AS, Aleman S, Chung RT, Chan AT, Ludvigsson JF. Association of Aspirin with Hepatocellular Carcinoma and Liver-Related Mortality. N Engl J Med 2020;382:1018–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lee M, Chung GE, Lee JH, Oh S, Nam JY, Chang Y, Cho H, et al. Antiplatelet therapy and the risk of hepatocellular carcinoma in chronic hepatitis B patients on antiviral treatment. Hepatology 2017;66:1556–1569. [DOI] [PubMed] [Google Scholar]
  • 94.Lee TY, Hsu YC, Tseng HC, Yu SH, Lin JT, Wu MS, Wu CY. Association of Daily Aspirin Therapy With Risk of Hepatocellular Carcinoma in Patients With Chronic Hepatitis B. JAMA Intern Med 2019;179:633–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R, Wolchok JD, et al. Safety and Tumor Responses with Lambrolizumab (Anti–PD-1) in Melanoma. N Engl J Med 2013;369:134–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, et al. Safety, Activity, and Immune Correlates of Anti–PD-1 Antibody in Cancer. N Engl J Med 2012;366:2443–2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, Kim TY, 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 2017;389:2492–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Maini MK, Pallett LJ. Defective T-cell immunity in hepatitis B virus infection: why therapeutic vaccination needs a helping hand. Lancet Gastroenterol Hepatol 2018;3:192–202. [DOI] [PubMed] [Google Scholar]
  • 99.Gane E, Verdon DJ, Brooks AE, Gaggar A, Nguyen AH, Subramanian GM, Schwabe C, et al. Anti-PD-1 Blockade with Nivolumab with and without Therapeutic Vaccination for Virally Suppressed Chronic Hepatitis B: A Pilot Study. J Hepatol 2019. [DOI] [PubMed] [Google Scholar]
  • 100.Gardiner D, Lalezari J, Lawitz E, DiMicco M, Ghalib R, Reddy KR, Chang KM, et al. A randomized, double-blind, placebo-controlled assessment of BMS-936558, a fully human monoclonal antibody to programmed death-1 (PD-1), in patients with chronic hepatitis C virus infection. PLoS One;8:e63818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Finn RS, Qin S, Ikeda M, Galle PR, Ducreux M, Kim T-Y, Kudo M, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. New England Journal of Medicine 2020;382:1894–1905. [DOI] [PubMed] [Google Scholar]
  • 102.Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med 2011;3:95ra73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Fisicaro P, Barili V, Montanini B, Acerbi G, Ferracin M, Guerrieri F, Salerno D, et al. Targeting mitochondrial dysfunction can restore antiviral activity of exhausted HBV-specific CD8 T cells in chronic hepatitis B. Nat Med 2017;23:327–336. [DOI] [PubMed] [Google Scholar]
  • 104.Qasim W, Brunetto M, Gehring AJ, Xue S-A, Schurich A, Khakpoor A, Zhan H, et al. Immunotherapy of HCC metastases with autologous T cell receptor redirected T cells, targeting HBsAg in a liver transplant patient. Journal of Hepatology 2015;62:486–491. [DOI] [PubMed] [Google Scholar]
  • 105.Kieckhaefer JE, Maina F, Wells RG, Wangensteen KJ. Liver Cancer Gene Discovery Using Gene Targeting, Sleeping Beauty, and CRISPR/Cas9. Semin Liver Dis 2019;39:261–274. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES