Abstract
Chronic hepatitis B virus (HBV) infection is a major risk factor for hepatocellular carcinoma (HCC). Pre-S2 mutant represents an HBV oncoprotein that is accumulated in the endoplasmic reticulum (ER) and manifests as type II ground glass hepatocytes (GGHs). Pre-S2 mutant can induce ER stress and initiate multiple ER stress-dependent or -independent cellular signal pathways, leading to growth advantage of type II GGH. Importantly, the mammalian target of rapamycin (mTOR) signal pathways are consistently activated throughout the liver tumorigenesis in pre-S2 mutant transgenic mice and in human HCC tissues, leading to hepatocyte proliferation, metabolic disorders, and HCC tumorigenesis. In this review, we summarize the pre-S2 mutant-induced mTOR signal pathways and its implications in HBV-related HCC tumorigenesis. Clinically, the presence of pre-S2 mutant exhibits a high resistance to antiviral treatment and carries a high risk of HCC development in patients with chronic HBV infection. Targeting at pre-S2 mutant-induced mTOR signal pathways may thus provide potential strategies for the prevention or therapy of HBV-associated HCC.
Keywords: Hepatitis B virus (HBV), Hepatocellular carcinoma (HCC), Ground glass hepatocytes (GGHs), Pre-S2 mutant, Mammalian target of rapamycin (mTOR)
Introduction
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related deaths worldwide, and a continued increase in incident rate is predicted1,2. Identification of novel therapeutic targets for HCC is thus urgently needed.
Chronic hepatitis B virus (HBV) infection is a major risk factor for the development of HCC3,4. Several hypo theses are proposed to explain the mechanisms of HBV-related tumorigenesis, such as insertional mutagenesis of HBV genome, inflammation, regeneration, and transactivating functions of HBV gene products, such as X protein (HBx) and truncated middle surface protein5–7.
Studies from our group recognize the pre-S2 mutant, which harbors deletion mutation (nucleotide 4–57 deletion) in the pre-S2 region of HBV large surface antigen (LHBs), as a viral oncoprotein that is accumulated in the endoplasmic reticulum (ER) and manifests as type II ground glass hepatocytes (GGHs)8,9. The retention of pre-S2 mutant protein in ER can induce ER stress and initiate an ER stress-dependent nuclear factor-κB/cyclo-oxygenase-2 signal pathway to protect hepatocytes from apoptosis9,10. Additionally, pre-S2 mutant can induce an ER stress-independent c-Jun activation domain-binding protein (1/p27/retinoblastoma/cyclin A) signal pathway to promote cell cycle progression11,12. The pre-S2 mutant-induced ER stress can also cause DNA damage, centrosome overduplication, and genomic instability13–15. The transforming ability of pre-S2 mutant has been investigated in an immortalized human hepatocyte line HH411. In addition, transgenic mice carrying pre-S2 mutant can develop HCC16. These studies support that combined effects of the pre-S2 mutant-induced signal pathways lead to growth advantage of type II GGHs and eventually HCC development17,18.
Our recent studies further reveal that the mammalian target of rapamycin (mTOR) signal pathway plays a critical role in pre-S2 mutant-driven tumorigenesis. Activation of mTOR signal pathways is consistently observed throughout the liver tumorigenesis in pre-S2 mutant transgenic mice and in human HCC tissues, leading to hepatocyte proliferation, metabolic disorders, and HCC tumorigenesis19–21. This review summarizes our works focused on the pre-S2 mutant-induced mTOR signal pathways and its implications in HBV-related HCC tumorigenesis. We also propose potential therapeutic strategies targeted at pre-S2 mutant-induced mTOR signal pathways for HCC.
Pre-S2 Mutant-Induced VEGF-A/VEGFR-2/AKT/mTOR Signal Pathway Promotes Hepatocyte Proliferation
HBV infection can induce chronic inflammation and contribute to HCC formation through the expression of cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)22,23. HBx has been shown to alter cytokine expression to modulate the immune response and proliferation of hepatocytes24,25. In transgenic mice of large surface antigen, the overproduction of large surface antigen can cause inflammation and regenerative hyperplasia to induce HCC development26,27. Therefore, inflammatory cytokines or growth factors may play a role in the disease progression from a precursor lesion to HCC through activation of growth factor/receptor signaling involving phosphatidylinositol 3-kinase/protein kinase B (Akt)/mTOR or Ras/Raf-1/extracellular signal-regulated kinase (ERK)28,29.
As expected, a human cytokine/growth factor antibody array reveals that the expression of pre-S2 mutant in HuH-7 hepatocytes enhances the secretion of several growth factors, including vascular endothelial growth factor (VEGF)-A and -D, transforming growth factor (TGF)-β1 and -β3, fibroblast growth factor (FGF)-7 and −9, and hepatocyte growth factor19. VEGF-A is selected for detailed study because of its angiogenesis and role in growth in the early stage lesions of human carcinogenesis30,31. The increased level of transcription, protein expression, and secretion of VEGF-A in hepatocytes expressing pre-S2 mutant is confirmed by real-time polymerase chain reaction (RT-PCR), Western blotting, and ELISA19, respectively. The expression of VEGF-A can be reduced by treatment with an ER stress inhibitor vomitoxin in hepatocytes expressing pre-S2 mutant19, indicating that pre-S2 mutant upregulates VEGF-A through ER stress. In addition, the enhanced proliferation of hepatocytes expressing pre-S2 mutant is significantly suppressed by the addition of neutralizing VEGF antibody to the culture supernatant19. The culture supernatant of VEGF-A from hepatocytes expressing pre-S2 mutant can increase human umbilical vein endothelial cell (HUVEC) proliferation19, suggesting that the pre-S2 mutant-upregulated VEGF-A promotes cell growth via an autocrine/paracrine loop.
To further study the mechanism of VEGF-A-induced hepatocyte proliferation, we first assess the expression of VEGF receptor (VEGFR)-1 and -2, which are reported to function in the regulation of VEGF-A signaling. The expression of VEGFR-2 mRNA and protein is upregulated in hepatocytes expressing pre-S2 mutant; however, the expression of VEGFR-1 is not increased19. Subsequently, we demonstrate an enhanced expression of the VEGF-A downstream signal molecules Akt, mTOR, and its phosphorylated (p) form (p-Akt, p-mTOR) in hepatocytes expressing pre-S2 mutant, which can be attenuated by VEGF-A neutralization19. Unexpectedly, another VEGF-A/VEGFR signal pathway, Raf-1 and ERK, shows no enhanced expression or activation. Collectively, these results demonstrate that pre-S2 mutant can activate an ER stress-dependent VEGF-A/VEGFR-2/Akt/mTOR signal pathway to promote hepatocyte proliferation (Fig. 1).
Figure 1.

Schematic overview of pre-S2 mutant-induced mTOR signal pathways. In chronic HBV infection, pre-S2 mutant accumulates in the ER of hepatocytes and induces ER stress. Through the induction of ER stress, the pre-S2 mutant can upregulate VEGF-A. The upregulated VEGF-A then functions in an autocrine (or paracrine) manner mediated by VEGFR-2 to activate Akt and mTOR signals. The activated mTOR signal can promote hepatocyte proliferation directly or indirectly by initiating two metabolic pathways, one involving YY1/MYC/SLC2A1 to stimulate aerobic glycolysis and another involving SREBF1/ACLY/FADS2 to promote lipid biosynthesis. The pre-S2 mutant-induced mTOR signal can additionally feedback suppress HBV large surface gene expression through YY1 recruitment of HDAC1-mediated transcriptional repression. HBV, hepatitis B virus; ER, endoplasmic reticulum; VEGF-A, vascular endothelial growth factor-A; VEGFR-2, VEGF receptor-2; Akt, protein kinase B; mTOR, mammalian target of rapamycin; YY1, Yin Yang 1; SLC2A1, solute carrier family 2 (facilitated glucose transporter) member 1; SREBF1, sterol regulatory element-binding transcription factor 1; ACLY, adenosine triphosphate (ATP) citrate lyase; FADS2, fatty acid desaturase 2; HDAC1, histone deacetylase 1.
In order to evaluate the role of pre-S2 mutant-induced VEGF-A/Akt/mTOR signal pathway in HCC tumorigenesis, we established transgenic mice overexpressing pre-S2 mutant in their livers. The extent of expression of each biomarker is then evaluated by Western blotting and normalized with age-matched nontransgenic liver tissues. Upregulation of VEGF-A, p-Akt, and p-mTOR signals is detected at as early as 1 month of age; the upregulated VEGF-A signal then subsides at 6 months, followed by p-Akt1/2/3 at 12 months32. The p-mTOR signal is consistently upregulated at all time points32, indicating that mTOR is a key regulator of pre-S2 mutant-driven hepatocarcinogenesis. Unlike the sequential activation of the VEGF-A/Akt/mTOR signal cascade, no clear pattern is observed for Raf-1/ERK signals32. To validate the signal activation in transgenic mice, 58 pairs of HBV-related nontumorous and HCC tissues are studied for signal activation. In consonance with the data in the transgenic mouse model, p-mTOR signal is significantly upregulated in tumor tissues32, supporting the important role of mTOR signal in HBV tumorigenesis.
A previous study demonstrated for the first time that type II GGHs exhibit enhanced VEGF-A expression through ER stress induced by the accumulation of pre-S2 mutant in ER19. The enhanced expression and secretion of VEGF-A can activate Akt/mTOR signaling to promote HBV-related hepatocarcinogenesis through VEGFR-2, thereby providing a potential mechanism for the progression from a benign precursor lesion of GGHs to HCC, providing a potential target for chemoprevention in the high-risk group of patients with chronic HBV infection. In addition, HBV is shown to play a crucial role in modulating the accumulation and activation of both cellular components of the tumor microenvironment, such as immune cells and fibroblasts, and noncellular components of the tumor microenvironment, such as cytokines and growth factors, markedly influencing disease progression and prognosis of HCC33. The autocrine/paracrine effects of pre-S2 mutant-upregulated VEGF-A on hepatocyte and/or endothelial cell proliferation may provide a novel insight into the role of pre-S2 mutant in the tumor microenvironment of HCC.
PRE-S2 Mutant-Induced mTOR/YY1/HDAC1 Signal Pathway Feedback Inhibits Large Surface Antigen Synthesis
One intriguing observation in chronic HBV infection is the low detection rate of HBV surface antigen (HBsAg), usually below 20% of cases in human HCC tissues, whereas HBsAg can be detected in almost 100% of cases in paired nontumorous livers34. The same finding is observed in HBsAg-expressing transgenic mice, which are accompanied by a decreased or absent expression of HBsAg in transgenic HCCs35. These observations indicate that the decreased HBsAg expression is a consistent phenomenon during the process of HBV-associated tumorigenesis. Although the levels of HBV DNA and HBsAg usually decline along with the natural course of chronic HBV infection36,37, there exists such a possibility that host cell factors may become activated to inhibit HBsAg expression or HBV replication during HBV-related tumorigenesis.
This speculation gains support from our clinical observation that shows a significantly inverse relationship between decreased LHBs and enhanced p-mTOR expressions in 13 of 20 paired human HBV-related HCC tissues38. A similar phenomenon is observed in HuH-7 hepatocytes expressing pre-S2 mutant, showing that the pre-S2 mutant-induced mTOR activation occurs at 48 h with concurrently decreased LHB RNA expression, followed by the decrease of LHB protein expression at 72 h after transfection38. Blockage of mTOR activation by the mTOR inhibitor rapamycin or mTOR-specific RNA interference can restore both RNA and protein expression levels of LHBs in hepatocytes expressing pre-S2 mutant38. Importantly, secreted LHBs in culture supernatant show the same patterns38, implying that serum HBsAg level may be concurrently decreased when mTOR becomes activated during HBV tumorigenesis. Together, these results demonstrate that the pre-S2 mutant-induced mTOR activation feedback suppresses LHB expression and secretion.
To elucidate the mechanism of LHB suppression by mTOR activation, we first perform a luciferase reporter assay in hepatocytes expressing pre-S2 mutant to detect the luciferase activity driven by the pre-S1 promoter, which controls the transcription of the LHB gene. The pre-S1 promoter-driven luciferase activity is decreased in hepatocytes expressing pre-S2 mutant and can be restored by mTOR inhibitor or RNAi treatment38, suggesting that mTOR activation represses pre-S1 promoter activity. Further assays with various deletion mutants of pre-S1 promoter reporter plasmids show that nucleotides 2,812–2,816 of the pre-S1 promoter is the minimal region for mediating mTOR signal-induced transcriptional repression38. Subsequently, the 2,812–2,816 site of the pre-S1 promoter is confirmed as the binding site of the transcription factor Yin Yang 1 (YY1) by electrophoretic mobility shift assay and DNA affinity precipitation assay38. Pre-S2 mutant-induced mTOR activation can enhance YY1 protein expression and nuclear localization38. Accumulating evidence indicates that YY1 can execute transcriptional repression by complexing with corepressors, among which histone deacetylase 1 and 2 (HDAC1 and HDAC2) are the most relevant39,40. Selective knockdown of HDAC1, but not HDAC2, protects the pre-S1 promoter from repression by pre-S2 mutant-induced mTOR activation in hepatocytes. Moreover, the YY1 antibody can coimmunoprecipitate higher levels of HDAC1 from pre-S2 mutant-expressed cells than control cells in an mTOR-dependent manner38, suggesting that HDAC1 is physically associated with YY1 and contributes to mTOR activation-induced transcriptional repression of LHBs (Fig. 1).
A previous study, for the first time, demonstrated one interesting negative feedback regulation of surface antigen synthesis by the activation of the mTOR signal during the progression of HBV tumorigenesis38. The decreased levels of HBsAg and HBV DNA in serum or hepatocytes, therefore, may not necessarily represent a good sign of disease improvement during the natural course of HCC development, but instead it may indicate a disease progression toward tumorigenesis, especially at the advanced stage of the diseases. This finding, together with the detection of pre-S mutations in serum41–43, should provide an additional hallmark to predict disease progression in the follow-up of patients with chronic HBV infection. In addition, several mTOR inhibitors have been developed at various phases of clinical trials44. According to our findings in a previous study study38, targeting mTOR signaling for HBV-related HCC may potentially lead to HBV reactivation. There are increasing reports on the reactivation of HBV replication and hepatitis flare-up in HBV-related HCC patients receiving anti cancer treatments45,46. Our results provide an explanation for the untoward effect of mTOR inhibitors on HCC patients with chronic HBV infection and emphasize the necessity of combining antiviral agents when mTOR inhibitors are used for HBV-related HCC therapy.
PRE-S2 Mutant-Induced mTOR/YY1/MYC/SLC2A1 Signal Pathway Induces Aerobic Glycolysis in Hepatocytes
Metabolic changes are common features in the development of many types of human cancers47. It has been reported that cancer cells frequently display high rates of aerobic glycolysis in comparison to their nontransformed counterparts, a phenomenon known as the “Warburg effect,” to support the increased demand of macromolecules for cell growth and proliferation48. Recently, several reports have uncovered multiple metabolic changes in HCC, including elevated glycolysis, which is one of the principal changes linked to highly proliferative malignant phenotype49–51. A previous study based on HBV transgenic mice has also consistently revealed a metabolic alteration of hepatocytes from the glycogen storage (glycogenotic) state toward an increase in glycolysis (the glycogen-poor state) during neoplastic transformation52. However, the underlying mechanism of HBV in the regulation of aerobic glycolysis in HCC development remains unclear.
mTOR is a highly conserved serine/threonine kinase that controls cell growth and proliferation53. In addition to its better known functions in promoting protein synthesis, mTOR is now emerging as a key regulator of cellular metabolism and cancer54. Research has documented that mTOR activation is sufficient to stimulate specific metabolic pathways, including aerobic glycolysis55. Our studies have demonstrated that the pre-S2 mutant can activate mTOR through the induction of ER stress-dependent VEGF-A/VEGFR-2/Akt signaling in GGHs to promote tumorigenesis19. The activated mTOR signal can further upregulate YY138, a transcription factor involved in cell proliferation and regulation of oncogenes56. We thus propose that pre-S2 mutant-induced mTOR activation may regulate aerobic glycolysis through YY1 signal cascade in HBV-related tumorigenesis.
In supporting our notion, the pre-S2 mutant transgenic mice exhibit glycogen depletion in HCC tissues as determined by periodic acid–Schiff staining and colorimetric-based assay20. The cDNA microarray data of pre-S2 mutant transgenic livers were adopted to identify the YY1-activated oncogenes. RT-PCR was performed to confirm the selected genes' transcription levels. The transcription factor MYC was identified as the only gene showing significant upregulation (≥1.5-fold) in transgenic HCCs relative to the nontransgenic livers20. By further analysis of the microarray data for potential MYC-activated glycolytic genes, the expression of the solute carrier family 2 (facilitated glucose transporter) member 1 (SLC2A1) was found significantly increased in transgenic HCCs20. These observations led us to test whether the pre-S2 mutant may regulate tumor glycolysis through activation of the mTOR/YY1/MYC/SLC2A1 signal cascade. Indeed, Western blot analysis of pre-S2 mutant transgenic mice livers showed that the expression levels of p-mTOR and YY1 were significantly elevated at as early as 1 month of age and persistently activated throughout the study period, while MYC and SLC2A1 expression was upregulated only upon tumor formation20. Moreover, the mTOR/YY1/MYC/SLC2A1 signal cascade was induced in HuH-7 and HepG2 hepatocytes expressing the pre-S2 mutant; selective knockdown of the upstream activators by RNA interference can sequentially diminish the activation of downstream targets20. The pre-S2 mutant-induced mTOR signal cascade can promote SLC2A1 translocation to the cell surface, where it functions in mediating cellular glucose uptake, resulting in stimulation of aerobic glycolysis and ultimately hepatocyte proliferation20 (Fig. 1).
To ascertain the association of the mTOR/YY1/MYC/SLC2A1 signal pathway with human HBV-related hepatocarcinogenesis, Western blot analysis was performed on 30 pairs of HBV-related HCCs and adjacent nontumorous livers for the expression of each biomarker. The p-mTOR, YY1, MYC, and SLC2A1 signals were consistently and significantly expressed at higher levels in HCCs than in the paired nontumorous livers in 18 of 30 tissue pairs20, supporting the essential role of the mTOR/YY1/MYC/SLC2A1 signal pathway in HBV tumorigenesis. Furthermore, in the model of chemopreventive study in transgenic mice harboring both pre-S2 mutant and HBx, the combined resveratrol and silymarin product can significantly decrease the transgenic tumor size compared with the untreated group57. In a previous study conducted in our laboratory, we observed a lower expression level of the mTOR/YY1/MYC/SLC2A1 signaling pathway in the treated group than in the untreated group20, suggesting that the mTOR/YY1/MYC/SLC2A1 signaling pathway mediates the chemopreventive effect of combined resveratrol and silymarin product on tumor growth.
This study, for the first time, demonstrates the contributing role of pre-S2 mutant in metabolic disturbances of HBV-related HCC development. The pre-S2 mutant can stimulate aerobic glycolysis through the activation of the mTOR/YY1/MYC signaling to upregulate SLC2A1. Upon the activation of SLC2A1 at the advanced stage of tumorigenesis, hepatocytes undergo a metabolic switch from the glycogen-storage state toward increased aerobic glycolysis. The increased SLC2A1 expression in HCC not only indicates an increased utilization of energy but can also directly cause tumorigenesis58. Therefore, our findings suggest that the pre-S2 mutant may promote tumorigenesis by sustaining high activation rates of aerobic glycolysis through the mTOR signal cascade.
PRE-S2 Mutant-Induced mTOR/SREBF1/ACLY/FADS2 Signal Pathway Disturbs Lipid Metabolism in Hepatocytes
Growing evidence indicates that cancer cells show specific alterations in lipid metabolisms that are important for cell growth and proliferation59,60. HCC has also been linked to nonalcoholic fatty liver, obesity, and related metabolic disorders61. Numerous reports uncover aberrant lipidomic profiles in human HCCs and mouse HCC models62,63. Furthermore, aberrations of lipid metabolism often are seen in chronic HBV infection64. However, the contributing role of disturbed lipid biosynthesis in HBV tumorigenesis remains to be clarified.
To elucidate the role of lipid metabolism in HBV tumorigenesis, we investigated the dynamic pattern of lipid metabolism in pre-S2 mutant-induced tumorigenesis. We first analyzed lipid profiles in transgenic mice harboring pre-S2 mutant. By Oil red O staining, mild and diffused fatty change in hepatocytes was observed in 1-month-old transgenic mice compared to the age-matched non-transgenic mice21. The staining intensity weakens as the disease progresses and eventually disappears in the later stages (6 and 12 months)21. Remarkably, large amounts of enlarged lipid droplets were found accumulated in transgenic tumors rather than the surrounding non-tumors21. Furthermore, the levels of triglycerides and cholesterol in transgenic livers were measured by colorimetric-based assays and showed consistency with the accumulation pattern found in the Oil red O staining21. These results indicate that lipid metabolism is disturbed in pre-S2 mutant-induced tumorigenesis.
It is now becoming clear that mTOR promotes de novo lipogenesis by inducing the cleavage of the sterol regulatory element-binding transcription factor 1 (SREBF1), which then translocates to the nucleus and induces the expression of many lipogenesis-related genes65,66. Considering that the pre-S2 mutant can induce reprogramming of glucose metabolism for hepatocyte proliferation through activation of mTOR signal pathway20, it is reasonable to investigate whether the pre-S2 mutant may also promote lipid biosynthesis through mTOR activation during the process of HBV tumorigenesis. The cDNA microarray data of pre-S2 mutant transgenic livers were adopted to identify the candidates of SREBF1 target lipogenic genes. The adenosine triphosphate (ATP) citrate lyase (ACLY) was the only gene that showed significant upregulation (≥1.5-fold) in transgenic HCCs compared with the non-transgenic livers21. This finding leads us to hypothesize that the pre-S2 mutant may regulate lipid metabolism through the activation of the mTOR/SREBF1/ACLY signal cascade. To test this hypothesis, Western blot analysis was performed to examine the expression of the signal molecules in different stages of pre-S2 mutant transgenic livers. As previously observed, the expression of p-mTOR was significantly elevated throughout the study period21. Interestingly, the precursor form of SREBF1 was upregulated in the middle to the late stage, 6 and 12 months, while the cleaved nuclear form of SREBF1 was found increased only upon tumor formation21. Both total and phosphorylated ACLY exhibit biphasic overexpression at the early and tumor stages21. Moreover, we demonstrate that the pre-S2 mutant can activate ACLY through mTOR/SREBF1 signaling to promote de novo lipogenesis and cell proliferation in HuH-7 and HepG2 hepatocytes21 (Fig. 1). The pre-S2 mutant can additionally upregulate the lipogenic enzyme fatty acid desaturase 2 (FADS2) through ACLY-dependent histone acetylation in hepatocytes21. FADS2 promoter-driven luciferase activities are increased in hepatocytes expressing the pre-S2 mutant; this upregulation can be abrogated by RNA interference-mediated ACLY inhibition and restored by treatment with the histone deacetylase inhibitor tricostatin A21. The essential role of the mTOR/SREBF1/ACLY/FADS2 signal pathway in human HBV-related tumorigenesis is further validated in 30 chronic HBV-infected HCC patients, showing a significantly increased expression of the signal pathway in 20 of 30 HCC tissues compared to the paired nontumorous liver tissues21.
This study, together with the role of the pre-S2 mutant in regulation of glucose metabolism, proposes a novel molecular mechanism to explain the pre-S2 mutant-induced metabolic disturbances in HBV-related tumorigenesis21. In chronic HBV infection, the pre-S2 mutant can stimulate aerobic glycolysis through an ER stress-dependent mTOR/YY1/MYC/SLC2A1 signal pathway. The aerobic glycolysis by-product, citrate, can be converted into acetyl-CoA, the raw material of triglycerides and cholesterol, by catalysis of ACLY, which is upregulated by another pre-S2 mutant-induced mTOR/SREBF1 signal pathway. Thus, ACLY plays an important role in linking the glucose metabolism to the endogenous biosynthesis of triglycerides and cholesterol at the advanced stage of tumorigenesis. Our findings suggest that the pre-S2 mutant plays a role in HBV tumorigenesis by disturbing normal metabolism, including elevating glycolysis and promoting lipid biosynthesis through the mTOR signal cascade. Targeting the glycolytic mTOR/YY1/MYC/SLC2A1 and the lipogenic mTOR/SREBF1/ACLY/FADS2 signal pathways may be a promising therapeutic strategy for HCC therapy.
PRE-S2 Mutant-Induced mTOR Signal Pathways Represent Potential Therapeutic Targets for HCC
Oral nucleos(t)ide analogs (NAs), including lamivudine, adefovir, entecavir, telbivudine, and tenofovir, are currently used to treat chronic HBV infection67. Early observations reveal that NA treatment not only significantly reduces the incidence of HCC68 but also lowers HCC recurrence rate69. Recently, a study revealed the resistance of pre-S2 mutant to NAs, providing an explanation for the high proportion of patients who still suffer from HCC recurrence after surgery despite anti-HBV therapy70. This highlights the importance of target therapies against pre-S2 mutant-induced signaling pathways that should be taken into consideration.
Resveratrol and silymarin are naturally occurring compounds that have anticarcinogenic effects on various types of cancers71,72. In the model of a chemopreventive study in transgenic mice harboring both pre-S2 mutant and HBx, the combined resveratrol and silymarin product has been shown to significantly ameliorate fatty liver and inhibit HCC growth57. Western blot analysis performed on the expression profiles of the mTOR signal cascade in the tumor adjacent tissues reveals that the treated group expresses a lower level of mTOR/YY1/MYC/SLC2A1 signaling than the untreated group20. We further examine the in vitro effect of resveratrol and silymarin on mTOR/YY1/MYC/SLC2A1 signaling cascade in HBV tumorigenesis. Consistent with pre-S2 mutant alone, HuH-7 and HepG2 hepatocytes expressing both pre-S2 mutant and HBx show activation of the mTOR signal cascade, which can be inhibited by the combined resveratrol and silymarin treatment20. These results indicate that the combined resveratrol and silymarin product may represent a potential agent for the prevention or therapy of HCC in high-risk chronic HBV carriers.
The bi-aryl urea sorafenib is an oral multikinase inhibitor that inhibits cell surface tyrosine kinase receptors including VEGFR and downstream intracellular serine/threonine kinases such as Raf-173. Sorafenib is the only approved therapy for advanced HCC74. The principal mechanism of sorafenib in action is that it competitively inhibits ATP binding to the catalytic domains of the respective kinases75. Several VEGF/VEGFR-based therapies are developed on the basis of different strategies including small molecular inhibition (e.g., sunitinib, Zactima, vatalanib) and antiligand targeting (e.g., bevacizumab and VEGF trap)29. As the name indicates, the primary target of VEGF/VEGFR-based therapy is focusing on the antiangiogenesis effects. The pre-S2 mutant induced ER stress-dependent VEGF-A/VEGFR-2/Akt/mTOR signal pathway, and the downstream mTOR signaling involved in hepatocarcinogenesis could also be neutralized by severing the source of VEGF-A/VEGFR signal cascade under sorafenib treatment.
Given mTOR involvement in many forms of cancers, it is a favorable target in cancer therapy76,77. Everolimus is a rapamycin analog, an oral mTOR inhibitor that binds to the FK506-binding protein 12 and directly interacts with mTOR complex 1 (mTORC1), inhibiting its downstream signaling78. However, mTOR inhibition induces insulin receptor substrate-1 expression and abrogates feedback inhibition of the pathway, resulting in Akt activation, which weakens its therapeutic effects79. One report suggests that targeting Akt and ERK signaling combined with rapamycin-based therapeutic approaches may be a new strategy for enhancing the treatment efficacy and inhibition of mTORC1 results in activation of Akt and ERK, but inhibiting mTORC2 leads to Akt and ERK suppression80. Moreover, a study shows that inhibition of mTORC2 induces glycogen synthase kinase 3-dependent and F-box and WD repeat domain-containing 7-mediated proteasomal degradation of SREBF1 and suppresses lipogenesis in cancer cells81. These results indicate that an mTOR inhibitor that inhibits both mTORC1 and mTORC2 could be the next generation of mTOR signaling-based target therapy.
The pre-S2 mutant induces mTOR activation and recruits the YY1-HDAC1 complex to feedback suppress transcription from the pre-S1 promoter38. Inhibition of mTOR can result in HBV reactivation, and it is demonstrated in a phase I clinical trial on everolimus that HBV reactivation is observed in four HBsAg-seropositive patients and indicates that prophylactic antiviral therapy should be mandatory for such patients82. One of The European Association for the Study of the Liver (EASL) 2012 guidelines recommend that HBV+ candidates for chemotherapy and immunosuppressive therapy should be tested for HBV DNA levels and should receive preemptive NA administration during therapy (regardless of HBV DNA levels) and for 12 months after cessation of therapy (A1).
In addition, several groups have demonstrated that HBsAg gene-modified dendritic cell-based vaccine can induce a specific immune response and strong antitumor effects in vitro or in mouse model83–85. Considering that hepatocytes harboring pre-S2 mutant exhibit stronger growth advantage than hepatocytes expressing HBsAg and that HBsAg expression frequently decreases in HCCs possibly due to the negative regulation by the pre-S2 mutant, it is reasonable to speculate that the pre-S2 mutant may be a more powerful antigen than HBsAg for developing a dendritic cell-based vaccine for HCC therapy.
It is a critical issue that viral oncoproteins can be continuously produced in the liver for years under NA treatment, presumably using cccDNA or integrated HBV genome as the template86,87. Distinct from oral NA, pegylated interferon (Peg-IFN) has been shown to effectively suppress cccDNA and reduce serum HBsAg level87,88. Moreover, lymphotoxin-β receptor (LT-bR) and downstream APOBEC3 cytidine deaminase pathway has also been proposed to be an alternative mechanism to specifically degrade cccDNA89. Therefore, combining NAs and drugs that degrade cccDNA, such as Peg-IFN and LTbR activator, to accelerate pre-S2 mutant elimination and reduce HCC development or recurrence is worth further investigation. In addition, label-retaining cancer cells are recently described as a novel subpopulation of HCC-derived cancer stem cells and display a high resistance to sorafenib treatment90. This phenomenon also highlights the importance of the development of novel cancer therapeutic drugs and/or combined therapeutic strategies for HCC.
Conclusion
Clinically, the prevalence of pre-S2 mutant is 37% in chronic HBV carriers and as high as 60% in HBV-related HCC patients17. The presence of pre-S2 mutant exhibits a high resistance to antiviral treatment70 and carries a high risk of HCC development41,42 in patients with chronic HBV infection. In this review, we highlight that mTOR dysregulation occurs with liver tumorigenesis in pre-S2 mutant transgenic mice and in human HCC tissues and represents the most important molecular mechanism initiated by pre-S2 mutant. Targeting at pre-S2 mutant-induced mTOR signal pathways may thus provide potential strategies for the prevention or therapy of HBV-associated HCC.
Acknowledgments
This work was supported by grants from the Ministry of Science and Technology. The authors declare no conflicts of interest.
References
- 1.Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet 2012; 379(9822): 1245–55. [DOI] [PubMed] [Google Scholar]
- 2.Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in globocan 2012. Int J Cancer 2015; 136(5): E359–86. [DOI] [PubMed] [Google Scholar]
- 3.Beasley RP, Hwang LY, Lin CC, Chien CS. Hepatocellular carcinoma and hepatitis B virus. a prospective study of 22 707 men in Taiwan. Lancet 1981; 2(8256): 1129–33. [DOI] [PubMed] [Google Scholar]
- 4.Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet 2003; 362(9399): 1907–17. [DOI] [PubMed] [Google Scholar]
- 5.Hildt E, Munz B, Saher G, Reifenberg K, Hofschneider PH. The Pres2 activator MHBs(t) of hepatitis B virus activates c-raf-1/Erk2 signaling in transgenic mice. EMBO J. 2002; 21(4): 525–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Park NH, Song IH, Chung YH. Chronic hepatitis B in hepatocarcinogenesis. Postgrad Med J. 2006; 82(970): 507–15.16891440 [Google Scholar]
- 7.Su IJ, Hsieh WC, Tsai HW, Wu HC. Chemoprevention and novel therapy for hepatocellular carcinoma associated with chronic hepatitis B virus infection. Hepatobiliary Surg Nutr. 2013; 2(1): 37–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fan YF, Lu CC, Chang YC, Chang TT, Lin PW, Lei HY, Su IJ. Identification of a pre-s2 mutant in hepatocytes expressing a novel marginal pattern of surface antigen in advanced diseases of chronic hepatitis B virus infection. J Gastroenterol Hepatol. 2000; 15(5): 519–28. [DOI] [PubMed] [Google Scholar]
- 9.Wang HC, Wu HC, Chen CF, Fausto N, Lei HY, Su IJ. Different types of ground glass hepatocytes in chronic hepatitis B virus infection contain specific pre-S mutants that may induce endoplasmic reticulum stress. Am J Pathol. 2003; 163(6): 2441–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hung JH, Su IJ, Lei HY, Wang HC, Lin WC, Chang WT, Huang W, Chang WC, Chang YS, Chen CC, Lai MD. Endoplasmic reticulum stress stimulates the expression of cyclooxygenase-2 through activation of NF-KAPPAB and PP38 mitogen-activated protein kinase. J Biol Chem. 2004; 279(45): 46384–92. [DOI] [PubMed] [Google Scholar]
- 11.Wang HC, Chang WT, Chang WW, Wu HC, Huang W, Lei HY, Lai MD, Fausto N, Su IJ,. Hepatitis B virus pre-S2 mutant upregulates cyclin a expression and induces nodular proliferation of hepatocytes. Hepatology 2005; 41(4): 761–70. [DOI] [PubMed] [Google Scholar]
- 12.Hsieh YH, Su IJ, Wang HC, Tsai JH, Huang YJ, Chang WW, Lai MD, Lei HY, Huang W. Hepatitis B virus pre-S2 mutant surface antigen induces degradation of cyclin-dependent kinase inhibitor p27Kip1 through c-Jun activation domain-binding protein 1. Mol Cancer Res. 2007; 5(10): 1063–72. [DOI] [PubMed] [Google Scholar]
- 13.Hsieh YH, Su IJ, Wang HC, Chang WW, Lei HY, Lai MD, Chang WT, Huang W. Pre-s mutant surface antigens in chronic hepatitis B virus infection induce oxidative stress and DNA damage. Carcinogenesis 2004; 25(10): 2023–32. [DOI] [PubMed] [Google Scholar]
- 14.Hsieh YH, Hsu JL, Su IJ, Huang W. Genomic instability caused by hepatitis B virus: Into the hepatoma inferno. Front Biosci (Landmark Ed) 2011; 16: 2586–97. [DOI] [PubMed] [Google Scholar]
- 15.Wang LH, Huang W, Lai MD, Su IJ. Aberrant cyclin a expression and centrosome overduplication induced by hepatitis B virus pre-S2 mutants and its implication in hepatocarcinogenesis. Carcinogenesis 2012; 33(2): 466–72. [DOI] [PubMed] [Google Scholar]
- 16.Wang HC, Huang W, Lai MD, Su IJ. Hepatitis B virus pre-S mutants, endoplasmic reticulum stress and hepatocarcinogenesis. Cancer Sci. 2006; 97(8): 683–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Su IJ, Wang HC, Wu HC, Huang WY. Ground glass hepatocytes contain pre-S mutants and represent preneoplastic lesions in chronic hepatitis B virus infection. J Gastroenterol Hepatol. 2008; 23(8 Pt 1): 1169–74. [DOI] [PubMed] [Google Scholar]
- 18.Su IJ, Wang LH, Hsieh WC, Wu HC, Teng CF, Tsai HW, Huang W. The emerging role of hepatitis B virus pre-S2 deletion mutant proteins in HBV tumorigenesis. J Biomed Sci. 2014; 21: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Yang JC, Teng CF, Wu HC, Tsai HW, Chuang HC, Tsai TF, Hsu YH, Huang W, Wu LW, Su IJ. Enhanced expression of vascular endothelial growth factor-A in ground glass hepatocytes and its implication in hepatitis B virus hepatocarcinogenesis. Hepatology 2009; 49(6): 1962–71. [DOI] [PubMed] [Google Scholar]
- 20.Teng CF, Hsieh WC, Wu HC, Lin YJ, Tsai HW, Huang W, Su IJ. Hepatitis B virus pre-S2 mutant induces aerobic glycolysis through mammalian target of rapamycin signal cascade. PLoS One 2015; 10(4): e0122373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Teng CF, Wu HC, Hsieh WC, Tsai HW, Su IJ. Activation of ATP citrate lyase by mTOR signal induces disturbed lipid metabolism in hepatitis B virus pre-S2 mutant tumorigenesis. J Virol. 2015; 89(1): 605–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sheron N, Lau J, Daniels H, Goka J, Eddleston A, Alexander GJ, Williams R. Increased production of tumour necrosis factor ALPHA in chronic hepatitis B virus infection. J Hepatol. 1991; 12(2): 241–45. [DOI] [PubMed] [Google Scholar]
- 23.Song le H, Binh VQ, Duy DN, Kun JF, Bock TC, Kremsner PG, Luty AJ. Serum cytokine profiles associated with clinical presentation in Vietnamese infected with hepatitis B virus. J Clin Virol. 2003; 28(1): 93–103. [DOI] [PubMed] [Google Scholar]
- 24.Yoo YD, Ueda H, Park K, Flanders KC, Lee YI, Jay G, Kim SJ. Regulation of transforming growth factor-BETA 1 expression by the hepatitis B virus (HBV) X transactivator. Role in HBV pathogenesis. J Clin Invest. 1996; 97(2): 388–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lee Y, Park US, Choi I, Yoon SK, Park YM, Lee YI. Human interleukin 6 gene is activated by hepatitis B virus-X protein in human hepatoma cells. Clin Cancer Res. 1998; 4(7): 1711–17. [PubMed] [Google Scholar]
- 26.Chisari FV, Klopchin K, Moriyama T, Pasquinelli C, Dunsford HA, Sell S, Pinkert CA, Brinster RL, Palmiter RD. Molecular pathogenesis of hepatocellular carcinoma in hepatitis B virus transgenic mice. Cell 1989; 59(6): 1145–56. [DOI] [PubMed] [Google Scholar]
- 27.Chisari FV, Filippi P, Buras J, McLachlan A, Popper H, Pinkert CA, Palmiter RD, Brinster RL. Structural and pathological effects of synthesis of hepatitis B virus large envelope polypeptide in transgenic mice. Proc Natl Acad Sci USA 1987; 84(19): 6909–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Avila MA, Berasain C, Sangro B, Prieto J. New therapies for hepatocellular carcinoma. Oncogene 2006; 25(27): 3866–84. [DOI] [PubMed] [Google Scholar]
- 29.Hopfner M, Schuppan D, Scherubl H. Growth factor receptors and related signalling pathways as targets for novel treat ment strategies of hepatocellular cancer. World J Gastroenterol. 2008; 14(1): 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996; 86(3): 353–64. [DOI] [PubMed] [Google Scholar]
- 31.Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT, De Bruijn EA. Vascular endothelial growth factor and angiogenesis. Pharmacol Rev. 2004; 56(4): 549–80. [DOI] [PubMed] [Google Scholar]
- 32.Wu HC, Tsai HW, Teng CF, Hsieh WC, Lin YJ, Wang LH, Yuan Q, Su IJ. Ground-glass hepatocytes co-expressing hepatitis B virus X protein and surface antigens exhibit enhanced oncogenic effects and tumorigenesis. Hum Pathol. 2014; 45(6): 1294–1301. [DOI] [PubMed] [Google Scholar]
- 33.Yang P, Markowitz GJ, Wang XF. The hepatitis B virus-associated tumor microenvironment in hepatocellular carcinoma. Natl Sci Rev. 2014; 1(3): 396–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang WL, London WT, Feitelson MA. Hepatitis B X antigen in hepatitis B virus carrier patients with liver cancer. Cancer Res. 1991; 51(18): 4971–77. [PubMed] [Google Scholar]
- 35.Farza H, Dragani TA, Metzler T, Manenti G, Tiollais P, Della Porta G, Pourcel C,. Inhibition of hepatitis B virus surface antigen gene expression in carcinogen-induced liver tumors from transgenic mice. Mol Carcinog. 1994; 9(4): 185–92. [DOI] [PubMed] [Google Scholar]
- 36.Martinot-Peignoux M, Boyer N, Colombat M, Akremi R, Pham BN, Ollivier S, Castelnau C, Valla D, Degott C, Marcellin P. Serum hepatitis B virus DNA levels and liver histology in inactive HBsAg carriers. J Hepatol. 2002; 36(4): 543–6. [DOI] [PubMed] [Google Scholar]
- 37.Su TH, Hsu CS, Chen CL, Liu CH, Huang YW, Tseng TC, Liu CJ, Chen PJ, Lai MY, Chen DS, Kao JH. Serum hepatitis B surface antigen concentration correlates with HBV DNA level in patients with chronic hepatitis B. Antivir Ther. 2010; 15(8): 1133–9. [DOI] [PubMed] [Google Scholar]
- 38.Teng CF, Wu HC, Tsai HW, Shiah HS, Huang W, Su IJ. Novel feedback inhibition of surface antigen synthesis by mammalian target of rapamycin (MTOR) signal and its implication for hepatitis B virus tumorigenesis and therapy. Hepatology 2011; 54(4): 1199–1207. [DOI] [PubMed] [Google Scholar]
- 39.Coull JJ, Romerio F, Sun JM, Volker JL, Galvin KM, Davie JR, Shi Y, Hansen U, Margolis DM. The human factors YY1 and LSF repress the human immunodeficiency virus type 1 long terminal repeat via recruitment of histone deacetylase 1. J Virol. 2000; 74(15): 6790–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Luke MP, Sui G, Liu H, Shi Y. Yin yang 1 physically interacts with HOXA11 and represses HOXA11-dependent transcription. J Biol Chem. 2006; 281(44): 33226–32. [DOI] [PubMed] [Google Scholar]
- 41.Chen CH, Hung CH, Lee CM, Hu TH, Wang JH, Wang JC, Lu SN, Changchien CS. Pre-S deletion and complex mutations of hepatitis B virus related to advanced liver disease in HBeAg-negative patients. Gastroenterology 2007; 133(5): 1466–74. [DOI] [PubMed] [Google Scholar]
- 42.Sinn DH, Choi MS, Gwak GY, Paik YH, Lee JH, Koh KC, Paik SW, Yoo BC. Pre-S mutation is a significant risk factor for hepatocellular carcinoma development: A long-term retrospective cohort study. Dig Dis Sci. 2013; 58(3): 751–8. [DOI] [PubMed] [Google Scholar]
- 43.Tsai HW, Lin YJ, Lin PW, Wu HC, Hsu KH, Yen CJ, Chan SH, Huang W, Su IJ. A clustered ground-glass hepatocyte pattern represents a new prognostic marker for the recurrence of hepatocellular carcinoma after surgery. Cancer 2011; 117(13): 2951–60. [DOI] [PubMed] [Google Scholar]
- 44.Treiber G. Mtor inhibitors for hepatocellular cancer: A forward-moving target. Expert Rev Anticancer Ther. 2009; 9(2): 247–61. [DOI] [PubMed] [Google Scholar]
- 45.Lok AS, Liang RH, Chiu EK, Wong KL, Chan TK, Todd D. Reactivation of hepatitis B virus replication in patients receiving cytotoxic therapy. Report of a prospective study. Gastroenterology 1991; 100(1): 182–8. [DOI] [PubMed] [Google Scholar]
- 46.Yeo W, Chan PK, Zhong S, Ho WM, Steinberg JL, Tam JS, Hui P, Leung NW, Zee B, Johnson PJ. Frequency of hepatitis B virus reactivation in cancer patients undergoing cytotoxic chemotherapy: A prospective study of 626 patients with identification of risk factors. J Med Virol. 2000; 62(3): 299–307. [DOI] [PubMed] [Google Scholar]
- 47.Cantor JR, Sabatini DM. Cancer cell metabolism: One hallmark, many faces. Cancer Discov. 2012; 2(10): 881–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hsu PP, Sabatini DM. Cancer cell metabolism: Warburg and beyond. Cell 2008; 134(5): 703–7. [DOI] [PubMed] [Google Scholar]
- 49.Bannasch P. Glycogenotic hepatocellular carcinoma with glycogen-ground-glass hepatocytes: A heuristically highly relevant phenotype. World J Gastroenterol. 2012; 18(46): 6701–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Beyoglu D, Imbeaud S, Maurhofer O, Bioulac-Sage P, Zucman-Rossi J, Dufour JF, Idle JR,. Tissue metabolomics of hepatocellular carcinoma: Tumor energy metabolism and the role of transcriptomic classification. Hepatology 2013; 58(1): 229–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Huang Q, Tan Y, Yin P, Ye G, Gao P, Lu X, Wang H, Xu G. Metabolic characterization of hepatocellular carcinoma using nontargeted tissue metabolomics. Cancer Res. 2013; 73(16): 4992–5002. [DOI] [PubMed] [Google Scholar]
- 52.Toshkov I, Chisari FV, Bannasch P. Hepatic preneoplasia in hepatitis B virus transgenic mice. Hepatology 1994; 20(5): 1162–72. [PubMed] [Google Scholar]
- 53.Laplante M, Sabatini DM. MTOR signaling at a glance. J Cell Sci. 2009; 122(Pt 20): 3589–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Cornu M, Albert V, Hall MN. mTOR in aging, metabolism, and cancer. Curr Opin Genet Dev. 2013; 23(1): 53–62. [DOI] [PubMed] [Google Scholar]
- 55.Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD,. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell. 2010; 39(2): 171–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhang Q, Stovall DB, Inoue K, Sui G. The oncogenic role of Yin Yang 1. Crit Rev Oncog. 2011; 16(3–4): 163–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Hsieh WC, Yang CW, Huang YS, Chao TW, Tsai TF, Su IJ. Chemoprevention of HBV-related hepatocellular carcinoma by the combined product of resveratrol and silymarin in transgenic mice. Functional Foods in Health and Disease 2013; 3: 341–52. [Google Scholar]
- 58.Amann T, Maegdefrau U, Hartmann A, Agaimy A, Marienhagen J, Weiss TS, Stoeltzing O, Warnecke C, Scholmerich J, Oefner PJ, Kreutz M, Bosserhoff AK, Hellerbrand C. Glut1 expression is increased in hepatocellular carcinoma and promotes tumorigenesis. Am J Pathol. 2009; 174(4): 1544–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 2007; 7(10): 763–77. [DOI] [PubMed] [Google Scholar]
- 60.Santos CR, Schulze A. Lipid metabolism in cancer. FEBS J. 2012; 279(15): 2610–23. [DOI] [PubMed] [Google Scholar]
- 61.Siegel AB, Zhu AX. Metabolic syndrome and hepatocellular carcinoma: Two growing epidemics with a potential link. Cancer 2009; 115(24): 5651–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Patterson AD, Maurhofer O, Beyoglu D, Lanz C, Krausz KW, Pabst T, Gonzalez FJ, Dufour JF, Idle JR. Aberrant lipid metabolism in hepatocellular carcinoma revealed by plasma metabolomics and lipid profiling. Cancer Res. 2011; 71(21): 6590–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Muir K, Hazim A, He Y, Peyressatre M, Kim DY, Song X, Beretta L. Proteomic and lipidomic signatures of lipid metabolism in NASH-associated hepatocellular carcinoma. Cancer Res. 2013; 73(15): 4722–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Su TC, Lee YT, Cheng TJ, Chien HP, Wang JD. Chronic hepatitis B virus infection and dyslipidemia. J Formos Med Assoc. 2004; 103(4): 286–91. [PubMed] [Google Scholar]
- 65.Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F. SREBP transcription factors: Master regulators of lipid homeostasis. Biochimie 2004; 86(11): 839–48. [DOI] [PubMed] [Google Scholar]
- 66.Laplante M, Sabatini DM. An emerging role of mTOR in lipid biosynthesis. Curr Biol. 2009; 19(22): R1046–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Singal AK, Fontana RJ. Meta-analysis: Oral anti-viral agents in adults with decompensated hepatitis B virus cirrhosis. Aliment Pharmacol Ther. 2012; 35(6): 674–89. [DOI] [PubMed] [Google Scholar]
- 68.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. Aliment Pharmacol Ther. 2013; 38(2): 98–106. [DOI] [PubMed] [Google Scholar]
- 69.Wu CY, Chen YJ, Ho HJ, Hsu YC, Kuo KN, Wu MS, Lin JT. Association between nucleoside analogues and risk of hepatitis B virus-related hepatocellular carcinoma recurrence following liver resection. JAMA 2012; 308(18): 1906–14. [DOI] [PubMed] [Google Scholar]
- 70.Tsai HW, Lin YJ, Wu HC, Chang TT, Wu IC, Cheng PN, Yen CJ, Chan SH, Huang W, Su IJ. Resistance of ground glass hepatocytes to oral antivirals in chronic hepatitis B patients and implication for the development of hepatocellular carcinoma. Oncotarget 2016; 7(19): 27724–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Joe AK, Liu H, Suzui M, Vural ME, Xiao D, Weinstein IB. Resveratrol induces growth inhibition, S-phase arrest, apoptosis, and changes in biomarker expression in several human cancer cell lines. Clin Cancer Res. 2002; 8(3): 893–903. [PubMed] [Google Scholar]
- 72.Agarwal R, Agarwal C, Ichikawa H, Singh RP, Aggarwal BB. Anticancer potential of silymarin: From bench to bed side. Anticancer Res. 2006; 26(6B): 4457–98. [PubMed] [Google Scholar]
- 73.Keating GM, Santoro A. Sorafenib: A review of its use in advanced hepatocellular carcinoma. Drugs 2009; 69(2): 223–40. [DOI] [PubMed] [Google Scholar]
- 74.Raza A, Sood GK. Hepatocellular carcinoma review: Current treatment, and evidence-based medicine. World J Gastroenterol. 2014; 20(15): 4115–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Wilhelm S, Chien DS. Bay 43-9006: Preclinical data. Curr Pharm Des. 2002; 8(25): 2255–7. [DOI] [PubMed] [Google Scholar]
- 76.Ashworth RE, Wu J. Mammalian target of rapamycin inhibition in hepatocellular carcinoma. World J Hepatol. 2014; 6(11): 776–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Huang Z, Wu Y, Zhou X, Qian J, Zhu W, Shu Y, Liu P. Clinical efficacy of mTOR inhibitors in solid tumors: A systematic review. Future Oncol. 2015; 11(11): 1687–99. [DOI] [PubMed] [Google Scholar]
- 78.Houghton PJ. Everolimus. Clin Cancer Res. 2010; 16(5): 1368–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, Lane H, Hofmann F, Hicklin DJ, Ludwig DL, Baselga J, Rosen N. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates akt. Cancer Res. 2006; 66(3): 1500–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Chen XG, Liu F, Song XF, Wang ZH, Dong ZQ, Hu ZQ, Lan RZ, Guan W, Zhou TG, Xu XM, Lei H, Ye ZQ, Peng EJ, Du LH, Zhuang QY. Rapamycin regulates AKT and ERK phosphorylation through mTORC1 and mTORC2 signaling pathways. Mol Carcinog. 2010; 49(6): 603–10. [DOI] [PubMed] [Google Scholar]
- 81.Li S, Oh YT, Yue P, Khuri FR, Sun SY. Inhibition of mTOR complex 2 induces GSK3/FBXW7-dependent degradation of sterol regulatory element-binding protein 1 (SREBP1) and suppresses lipogenesis in cancer cells. Oncogene 2016; 35(5): 642–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Shiah HS, Chen CY, Dai CY, Hsiao CF, Lin YJ, Su WC, Chang JY, Whang-Peng J, Lin PW, Huang JD, Chen LT,. Randomised clinical trial: Comparison of two everolimus dosing schedules in patients with advanced hepatocellular carcinoma. Aliment Pharmacol Ther. 2013; 37(1): 62–73. [DOI] [PubMed] [Google Scholar]
- 83.Qiu SJ, Lu L, Qiao C, Wang L, Wang Z, Xiao X, Qian S, Fung JJ, Ye SL, Bonham CA. Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transduced by adenoviral vectors encoding HBsAg: Comparison to protein immunization. J Cancer Res Clin Oncol. 2005; 131(7): 429–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Shi M, Qian S, Chen WW, Zhang H, Zhang B, Tang ZR, Zhang Z, Wang FS. Hepatitis B virus (HBV) antigen-pulsed monocyte-derived dendritic cells from HBV-associated hepatocellular carcinoma patients significantly enhance specific T cell responses in vitro. Clin Exp Immunol. 2007; 147(2): 277–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Yang JY, Cao DY, Xue Y, Yu ZC, Liu WC. Improvement of dendritic-based vaccine efficacy against hepatitis B virus-related hepatocellular carcinoma by two tumor-associated antigen gene-infected dendritic cells. Hum Immunol. 2010; 71(3): 255–62. [DOI] [PubMed] [Google Scholar]
- 86.Babinet C, Farza H, Morello D, Hadchouel M, Pourcel C. Specific expression of hepatitis B surface antigen (HBsAg) in transgenic mice. Science 1985; 230(4730): 1160–3. [DOI] [PubMed] [Google Scholar]
- 87.Wursthorn K, Lutgehetmann M, Dandri M, Volz T, Buggisch P, Zollner B, Longerich T, Schirmacher P, Metzler F, Zankel M, Fischer C, Currie G, Brosgart C, Petersen J. Peginterferon ALPHA-2B plus adefovir induce strong cccdna decline and HBsAg reduction in patients with chronic hepatitis B. Hepatology 2006; 44(3): 675–84. [DOI] [PubMed] [Google Scholar]
- 88.Moucari R, Mackiewicz V, Lada O, Ripault MP, Castelnau C, Martinot-Peignoux M, Dauvergne A, Asselah T, Boyer N, Bedossa P, Valla D, Vidaud M, Nicolas-Chanoine MH, Marcellin P,. Early serum HBsAg drop: A strong predictor of sustained virological response to pegylated interferon ALPHA-2A in HBeAg-negative patients. Hepatology 2009; 49(4): 1151–7. [DOI] [PubMed] [Google Scholar]
- 89.Lucifora J, Xia Y, Reisinger F, Zhang K, Stadler D, Cheng X, Sprinzl MF, Koppensteiner H, Makowska Z, Volz T, Remouchamps C, Chou WM, Thasler WE, Huser N, Durantel D, Liang TJ, Munk C, Heim MH, Browning JL, Dejardin E, Dandri M, Schindler M, Heikenwalder M, Protzer U. Specific and nonhepatotoxic degradation of nuclear hepatitis B virus cccDNA. Science 2014; 343(6176): 1221–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Xin HW, Ambe CM, Hari DM, Wiegand GW, Miller TC, Chen JQ, Anderson AJ, Ray S, Mullinax JE, Koizumi T, Langan RC, Burka D, Herrmann MA, Goldsmith PK, Stojadinovic A, Rudloff U, Thorgeirsson SS, Avital I. Label-retaining liver cancer cells are relatively resistant to sorafenib. Gut 2013; 62(12): 1777–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
