109
Views
3
CrossRef citations to date
0
Altmetric
Review

Translational Regulation in Hepatocellular Carcinogenesis

, &
Pages 4359-4369 | Published online: 14 Oct 2021

Abstract

The mortality of hepatocellular carcinoma (HCC) is distributed unevenly worldwide. One of the major causes is hepatitis B or hepatitis C virus infection and the development and progression of liver cirrhosis. The carcinogenesis of HCC is among others regulated via the mTOR (mechanistic target of rapamycin) signaling pathway and represents a possible method of targeted treatment. The aim of our article was to address the most recent clinical advances and findings of basic studies on the mTOR signaling pathway and the involved factors. Risk factors play a key role in dysregulation of the signaling pathway, where both mTORCs are upregulated and protein synthesis is altered. eIFs and, to a lesser extent, eEFs play an essential role in this process. Whether the factor will be upregulated or downregulated, among others, depends on hepatitis B/C virus infection. The amount of a particular factor in a patient sample lets us know whether HCC recurrence will occur, what is the likelihood of chemoresistance, and what outcome is predicted for patients with an increased value. Our analysis shows that in addition to mTOR, eIF3, eIF4, and eIF5 play an important role, as they can serve as biomarkers for non- and virus-related HCC.

Introduction

Hepatocellular carcinoma (HCC) is a major health problem, as it was the third most common cause of mortality worldwide in 2020.Citation1 Between 1990 and 2016 it ranked as the fifth most common cause of cancer in men and the seventh in women.Citation2

Major risk factors include chronic infection with hepatitis B (HBV) or hepatitis C virus (HCV), exposure to aflatoxin B1, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcohol-related liver disease like alcoholic steatohepatitis (ASH), diabetes and obesity.Citation1,Citation3,Citation4

Development of HCC begins with exposure to one or more risk factors, which trigger inflammation of liver parenchyma with fibrotic deposition. When cirrhosis and decreased activity occur, the likelihood of developing HCC increases. Changes in the genome, as well as epigenetic modifications, play an important role in further development. Although several authors have already dealt with the molecular profile of HCC and the associated progression or recurrence of the disease,Citation5,Citation6 two main molecular groups were identified: proliferative and non-proliferative HCC.Citation7 A classification based on metabolism was presented.Citation8

HCC classification, treatment, and survival follow the Barcelona Clinic Liver Cancer staging and treatment strategy (BCLC). BCLC presents five prognostic subclasses and specific treatments for each: surgical resection, liver transplantation, ablation, chemoembolization, and systemic therapy.Citation9 Despite the known risk factors and various options for monitoring and diagnosis, 40% of patients with an early stage (stage 0 and A) are suitable for treatment with resection, transplantation or local ablation. Stage B patients profit from chemoembolization and patients with advanced cancer (stage C) receive systemic therapy using sorafenib.Citation10,Citation11 BCLC staging could be upgraded with additional molecular-based information as it is associated with patient recurrence and outcome. Dysregulation of the signaling pathways participating in HCC development could represent additional information for therapy decision making.Citation12

Dysregulation in multiple signaling pathways such as phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR), Ras mitogen-activated protein kinase (Ras/Raf/MAPK), Hedgehog (HH), Wnt/β-catenin, Janus kinase-signal transducer activator of transcription factor (JAK/STAT) signaling pathway was found in HCC carcinogenesis.Citation13,Citation14

PI3K/AKT/mTOR signaling pathway has been extensively studied in the development of HCC with and without viral background. Dysregulation of this pathway is key in hepatocarcinogenesis.Citation15

The present review article addresses advances in the mTOR signaling pathway and its factors, allowing us to better understand the treatment possibilities of patients with HCC.

PI3K/AKT/mTOR Pathway

The PI3K/AKT/mTOR pathway is an intracellular signaling pathway that is activated by insulin and various growth factors (). PI3K activation phosphorylates and activates AKT, which, among others, activates mTOR, affecting the transcription of p70 or 4EBP1. mTOR has two complexes, mTORC1 and mTORC2. Activation of the signal pathway results in fatty acid synthesis, glycogen synthesis, cell survival, proliferation, autophagy, glycolysis and protein synthesis.Citation16

Figure 1 An overview of PI3K/AKT/mTOR signaling pathway shows activation of PI3K through receptor tyrosine kinases (RTK) and G-protein coupled receptors (GPCR). It acts directly on AKT or indirectly via mTORC2. Activation of inhibition of adenosine triphosphate (ATP)-citrate lyase, glycogen synthase kinase 3 (GSK3), forkhead box protein O (FOXO), Rab GTPase-activating protein (AS160) and tuberous sclerosis 1/2. As a result fatty acid synthesis, glycogen synthesis, cell survival, proliferation, autophagy, glycolysis and protein synthesis occur.

Abbreviations: PIP3, phosphatidylinositol (3,4,5)-trisphosphate; GS, glycogen synthase; GLUT4, glucose transporter type 4; Rheb, Ras homolog enriched in brain; p70S6K, ribosomal protein S6 kinase beta-1; S6, ribosomal protein S6.
Figure 1 An overview of PI3K/AKT/mTOR signaling pathway shows activation of PI3K through receptor tyrosine kinases (RTK) and G-protein coupled receptors (GPCR). It acts directly on AKT or indirectly via mTORC2. Activation of inhibition of adenosine triphosphate (ATP)-citrate lyase, glycogen synthase kinase 3 (GSK3), forkhead box protein O (FOXO), Rab GTPase-activating protein (AS160) and tuberous sclerosis 1/2. As a result fatty acid synthesis, glycogen synthesis, cell survival, proliferation, autophagy, glycolysis and protein synthesis occur.

The PI3K/AKT/mTOR signaling pathway is essential for protein synthesis, which is divided into four steps: initiation, elongation, termination, and ribosome recycling. Eukaryotic translation initiation factors (eIFs) thus enter the initiation phase followed by elongation, which is accompanied by eukaryotic translation elongation factors (eEFs). mTOR phosphorylation leads via additional phosphorylation steps to the phosphorylation of 4E-BP-1. 4E-BP1 dissociates and allows the eIF4F complex formation. This results in a cap-dependent activation of translation initiation. The formation of a 43S pre-initiation complex consisting of a 40S small ribosomal unit, eIF3, eIF1, eIF1A, eIF2 and eIF5 starts. Later, the eIF4F complex recognizes the 5’(prime) end of mRNA and binds to the 43S pre-initiation complex. The scanning of the start codon begins. After recognition of the start codon, the recruitment of the 60S ribosome and finally the formation of the 80S ribosome follows. Subsequently, all eIFs are released.Citation17,Citation18

mRNA chain then enters the elongation phase, where amino acids are added into a growing polypeptide chain. There are binding sites for tRNA on the ribosome, namely P (peptidyl) and A (aminoacyl). Upon completion of initiation, Met-tRNAiMet is bound to the P site of the ribosome. The Met-tRNAiMet anticodon is paired with the initial messenger RNA (mRNA) codon, and the second open reading frame (ORF) codon is in place. Elongation begins with aminoacyl-tRNA (RNA transfer) to the A site of the ribosome. Following guanosine triphosphate (GTP) binding, the eukaryotic eEf1A translation elongation factor is activated, and a ternary complex is formed. The eEF1A/GTP/aminoacyl-tRNA complex binds to site A. A peptide bond is formed between amino acids during peptidyl transfer with the amino acid cleaving from the tRNA located at the site P. The mRNA shifts from the 5’ to 3’ end. tRNA that was previously at site A is moved to site P; from site P to exit point (site E), where it leaves the ribosome. This task is performed with an elongation factor 2, which hydrolyzes the GTP.Citation19Citation21

PI3K/AKT/mTOR in HCC

HCC PI3K/AKT/mTOR signaling () is dysregulated, and both mTOR complexes are involved: mTORC1 regulates protein synthesis, autophagy, glucose and lipid metabolism, and mTORC2 stimulates liver tumorigenesis. Both mTOR complexes were reported to be upregulated in 40–50% of HCC patients.Citation22

Figure 2 Non-virus associated regulation of mTOR is regulated by PI3K-AKT, which regulates FOXO (1 and 3a) as also protein translation by S6 and eIF4E. Liver kinase B1 (LKB1) - adenosine monophosphate-activated protein kinase (AMPK) pathway can activate TSC1-TSC2 complex, which regulates Ras (small GTPase) homolog enriched in brain (Rheb) and activates mTORC1. Activation of mTORC1 can be regulated by mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) pathway.

Figure 2 Non-virus associated regulation of mTOR is regulated by PI3K-AKT, which regulates FOXO (1 and 3a) as also protein translation by S6 and eIF4E. Liver kinase B1 (LKB1) - adenosine monophosphate-activated protein kinase (AMPK) pathway can activate TSC1-TSC2 complex, which regulates Ras (small GTPase) homolog enriched in brain (Rheb) and activates mTORC1. Activation of mTORC1 can be regulated by mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) pathway.

Under physiological conditions, the liver is an organ highly involved in lipid metabolism. The liver transforms glucose into fatty acids, which is regulated by the PI3K/AKT/mTOR pathway with insulin and enzymes. It was shown that upregulation of lipid synthesis causes hepatosteatosis and consequently NASH and HCC.Citation23 mTORC2 stimulates the uptake of fatty acids directly from the blood or is involved in de novo synthesis of fatty acids.Citation24

NAFLD is caused by free fatty acids and their accumulation in hepatocytes. 30–40% of patients who later develop NASHCitation25 Center for pathogenesis of NASH are free fatty acids produced by lipolysis in adipose tissue. In the case of over elevated levels of free fatty acids, lipotoxic elements can be formed, which leads to endoplasmic reticulum stress, oxidant stress and activation of inflammation cells.Citation26 Elevated levels of fatty acids can induce NAFLD, which might progress to NASH and lead to HCC development.Citation25,Citation27 It was also shown that increased resistance to insulin was connected to decreased mTORC2-activity.Citation25

Chronic alcohol consumption can lead to liver cirrhosis due to oxidative stress and the formation of steatosis by promoting hepatic lipogenesis. Increased phosphorylation of mTOR, activation of mTORC1 and downstream kinase S6K1 in hepatocytes has to take place.Citation28 Occasional alcohol consumption promotes autophagy as a protective mechanism against hepatocyte damage from alcohol.Citation28,Citation29 Chronic alcohol consumption is frequently associated with oxidative stress, inhibition of hepatocellular autophagy and apoptosis through inhibition of AMPK and activation of mTOR.Citation30

Dysregulated bile acids are associated with liver diseases as increased cell proliferation and autophagy are mediated by AMPK/mTOR pathway. Due to a protective mechanism enhanced cancer cell viability, and consequently, metastases could be formed.Citation31

One of the risk factors for HCC is a chronic infection with HBV or HCV. It is typical for viruses that require their own translation of the host’s proteins, for which they use several different mechanisms, PI3K/AKT/mTOR, for example. As mentioned, this signal pathway pertains to the cap-dependent translation, where 4E-BP is needed for the regulation of the creation of the eIF4F complex. The interaction between eIF4E and 4E-BP is regulated, on the one hand, by phosphorylation and, on the other hand, by viruses, both in negative and positive ways.Citation32 The activation of this pathway, caused by viruses, inhibits apoptosis and promotes the survival of infected cells for further viral replication.Citation33

HCV uses the PI3K/AKT signal pathway to enter a cell. It has been evidenced that the HCV NS5A protein binds itself to PI3K and activates the PI3K/AKT signaling pathway ().Citation34,Citation35

Figure 3 Overview of the regulation of mTOR related to HBV and HCV infection. HCV infection activates NS5A (nonstructural protein 5A). Its P85 form modulates with endoplasmic reticulum membrane or dysregulates association of FKBP5 (FK506 binding protein 5) with mTOR and PTEN is downregulated. HBV upregulates VEGFR-2 (vascular endothelial growth factor) by pre-S1 promoter and the AKT/mTOR pathway is activated. As a negative feedback, YY1-HDAC1 (mammalian zinc-finger transcription factor - Histone deacetylase 1) can inhibit pre-S1 transcription. HBx can activate PI3K and downstream pathway.

Figure 3 Overview of the regulation of mTOR related to HBV and HCV infection. HCV infection activates NS5A (nonstructural protein 5A). Its P85 form modulates with endoplasmic reticulum membrane or dysregulates association of FKBP5 (FK506 binding protein 5) with mTOR and PTEN is downregulated. HBV upregulates VEGFR-2 (vascular endothelial growth factor) by pre-S1 promoter and the AKT/mTOR pathway is activated. As a negative feedback, YY1-HDAC1 (mammalian zinc-finger transcription factor - Histone deacetylase 1) can inhibit pre-S1 transcription. HBx can activate PI3K and downstream pathway.

Even though the use of the PI3K/AKT/mTOR is useful to the viruses, it at the same time limits the procreation of HBV,Citation36 because the lessened RNA-transcription and lessened the HBV X (HBx) protein plays an essential role in pathogenesis since it encourages cell cycle progression, deactivates negative growth factors and has an effect on the tumor-suppressing genes.Citation37 Upon HBx-transfection, autophagy is heightened by activating the PI3K/AKT/mTOR signal pathway.Citation38 In cases of HCV-coinfection, the HBV-replication is suppressed.Citation39

Translation Control Upon Viral Infection and as Response to Non-Virally Triggered Stress

eIFs

eIFs are part of the translation process and together with other PI3K/AKT/mTOR pathway members are dysregulated in malignancies,Citation40 but the studies involving eIFs and eEFs in HCC are scarce.

As already published by our group, eIF4E and eIF3I are involved in non-virus related HCC formation. Additionally, eIF4E and eIF3I were downregulated for HBV- and HCV-related HCC, as well as for chronic hepatitis B and C. eIF5 was downregulated in non-virus related HCC, HCV-related HCC and chronic hepatitis C, but was overexpressed in HBV-related HCC. The eIF subunits p-2a, 2a, 3B, 3C, 3D, 3J, p-4B, 4G and 6 were upregulated in HCV-associated HCC. With the help of eIF3 subunits B, C, I, eIF4E, eIF4G and eIF5, a reduction in protein expression in HCV was observed. In HBV-associated HCC and chronic hepatitis B, overexpression of p-eIF2a, p-eIF4B and eIF4G was seen. Downregulation of eIF2a, eIF3D, eIF3H, eIF3I, eIF3J, eIF4E and eIF6 in HBV-associated HCC and chronic hepatitis B was observed.Citation41

Another component of the translation, eIF3, is a protein complex that can act as a tumor suppressor or tumor promoting factor.Citation42 In HCC, overexpression of eIF3h and eIF3i was observed. Interaction of overexpressed eIF3i with AKT1 prevents PP2A-mediated dephosphorylation and consequently activation of AKT1 signaling. If eIF3i specific inhibitors are applied, tumorigenesis is suppressed.Citation43

Overexpression of eIF4E is associated with a higher risk of HCC recurrence, but the factor is overexpressed in other liver cancers as well.Citation40 Another factor, namely eIF4AIII, was found to be overexpressed in HCC and promoted tumor cell proliferation and migration.Citation44 eIF4AII was identified as a host factor supporting HCV replication in different replication systems, in contrast to eIF4AI where an opposite function was observed.Citation45 A study on HepG2 cells showed that eIF4E phosphorylation with Mnk inhibitor blockage could be a new therapeutic option against chemoresistant HCC.Citation46 It has also been suggested that patients with HCC who have elevated eIF4E factor in tumor samples have an increased likelihood of tumor recurrence.Citation40 In a case where sorafenib, a kinase inhibitor, was used as a line of therapy, drug resistance can occur. It was shown that the eIF4E-eIF4G complex reduces the effectiveness of therapy.Citation47

The eIF5 factor was shown to be increased in HCC.Citation48,Citation49 In patients with elevated expression level of eIF5A1, multiple tumor nodules were associated, and in eIF5A2, increased venous infiltration was associated. Inhibition of both isoforms was manifested by reduced HCC proliferation and invasiveness.Citation48 In patients receiving chemotherapy, HCC cells go through epithelial–mesenchymal transition (EMT) and, as a result, chemotherapy is less effective, but a link to eIF5A2 has been found in chemoresistant samples. If eIF5A2 is inhibited, EMT occurs to a lesser extent, and chemotherapy is more effective.Citation50 As suggested by Tang et al, eIF5A2 in the case of chemoresistance suppresses HCC autophagic cell death.Citation51 It was also shown that eIF5A2 could promote cell proliferation and trigger metabolic pathways for glucose metabolism and fatty acid biosynthesis in HCC. The factor could thus not only be a new target for therapy but could also serve as a new prognostic and predictive marker.Citation52 eIF5A2 is considered to be a factor that promotes metastasis. Inhibition of the eIF5A2 results in a minor invasion and metastasis, for which the reactive oxygen species pathway is thought to be responsible.Citation53,Citation54 For another factor from eIF5 family, namely eIF5B, it was shown that its expression was increased in HCC tissue samples. The factor is associated with a poorer outcome. The proliferation and invasion of cancer cells is accelerated by ASAP1 expression.Citation55 In general, eIF5 is believed to serve as a biomarker for non-virus-related and virus-related HCC.Citation41

To our knowledge, no study can be found to directly indicate the expression profile of eIF6 in HCC. However, it was shown that through competitively binding with miR-588 eIF6 expression is promoted and PI3K/AKT/mTOR signaling pathway activated by p38-inhibited cutaneous squamous cell carcinoma-associated lincRNA (PICSAR) in HCC.Citation56

eEFs

Recently, more attention has been paid to eEF1 and eEF2 and their subunits and complexes, as they are involved in the elongation step.

eEF1A1 regulates cell cycle and promotes malignant cells proliferation. In the case of HCC, downregulation of eEF1A1 inhibited tumor cell proliferation.Citation57 Based on mRNA study, it was shown that eEF1A was increased in HCC compared to non-tumor liver. In cell lines, eEF1A1 and eEF1A2 are correlated with cell growth and differentiation stage.Citation58

It was shown that eEF2 promotes angiogenesis in tumor progression of HCCCitation59 and is a potential drug target.Citation60 Additionally, it might serve as a prognostic marker.Citation60

Ongoing Clinical Trials

Regarding the extent and nature of HCC atezolizumab, bevacizumab (first line) or sorafenib, lenvatinib (first line), regorafenib (second line), pembrolizumab or nivolumab, hormonal compounds (tamoxifen, antiandrogens, and seocalcitiol) and systemic chemotherapy are standard therapy to treat HCCCitation61 ().

Figure 4 Shows mTOR and PI3K inhibitors targeting the mTOR pathway. BEZ235 and XL765 both target mTOR and PI3K and PI-103 targets PI3K. Rapamycin, temsirolimus, everolimus, ridaforolimus, PP242, AZD8055, OSI-027, INK128 and UDCA inhibit mTOR.

Figure 4 Shows mTOR and PI3K inhibitors targeting the mTOR pathway. BEZ235 and XL765 both target mTOR and PI3K and PI-103 targets PI3K. Rapamycin, temsirolimus, everolimus, ridaforolimus, PP242, AZD8055, OSI-027, INK128 and UDCA inhibit mTOR.

Current ongoing clinical trials (*clinicaltrials.gov) investigating the role of different mTOR inhibitors are summarized in and .

Table 1 Drugs and Its Targets

Table 2 Clinical Trials for HCC and mTOR

In a prospective randomized trial, the effect of sirolimus (rapamycin, an mTOR inhibitor) on the HCC recurrence after liver transplantation was studied (Clinicaltrials.gov: NCT00355862). Results showed that treatment with mTOR-inhibitor for ≥3 months improved the outcome. A subgroup of patients with elevated alpha-fetoprotein where better outcome was observed, indicating the usefulness of mTOR-inhibitors in this group of patients.Citation62

Another prospective-randomized open-label international trial (ClinicalTrials.gov NCT00355862) investigated the effect of sirolimus in liver transplanted patients. It was shown that patients could benefit from sirolimus as recurrence-free survival and overall survival were better in the first 3 to 5 years.Citation63

An open-label, parallel-group, noncomparative, randomised study investigated the maximum tolerated dose of the mTOR-inhibitor everolimus for patients with advanced HCC. It was recommended that 7.5 mg of everolimus should be taken daily with acceptable tolerability and evidence of efficiency (ClinicalTrials.gov NCT00390195).Citation64

Future Consideration

Although the risk factors for hepatocellular carcinogenesis are known, treatment is successful to a low extent as HCC-based mortality is still high. As described, different signaling pathways play an important role in HCC development, also the PI3K/AKT/mTOR pathway with and without viral interplay.

Recently, new techniques have opened up additional possibilities for studying the carcinogenesis of HCC. One of them is liquid biopsy, which is one of the least studied in HCC.Citation68,Citation69 Another promising area is molecular profiling of the tumor, which would allow us an individual approach to treatment. However, there is a problem with inter- and intraheterogeneity of the tumor, as well as the heterogeneity of the tumor in its development. Studies of molecular mechanisms as well as clinical studies suggest that much remains unclear in the field of hepatocellular carcinogenesis as well as targeted therapy and offers us opportunities to research and develop new target drugs in the future.Citation70,Citation71

Disclosure

The authors declare no conflicts of interest.

References

  • SungH, FerlayJ, SiegelRL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;1–41. doi:10.3322/caac.21660.
  • FitzmauriceC, AkinyemijuTF, Al LamiFH, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016: a systematic analysis for the global burden of disease study global burden of disease study. JAMA Oncol. 2018;4(11):1553–1568. doi:10.1001/jamaoncol.2018.270629860482
  • LiberalR, GrantCR. Cirrhosis and autoimmune liver disease: current understanding. World J Hepatol. 2016;8(28):1157–1168. doi:10.4254/wjh.v8.i28.115727729952
  • SchlesingerS, AleksandrovaK, PischonT, et al. Diabetes mellitus, insulin treatment, diabetes duration, and risk of biliary tract cancer and hepatocellular carcinoma in a European Cohort. Ann Oncol. 2013;24(9):2449–2455. doi:10.1093/annonc/mdt20423720454
  • HuangY-K, QiuF. Current perspectives of recurrence and progression in hepatocellular carcinoma. J Xiangya Med. 2017;2(9):68. doi:10.21037/jxym.2017.09.01
  • HoyosS, EscobarJ, CardonaD, et al. Factors associated with recurrence and survival in liver transplant patients with HCC - A single center retrospective study. Ann Hepatol. 2015;14(1):58–63. doi:10.1016/s1665-2681(19)30801-425536642
  • ChiangDY, VillanuevaA, HoshidaY, et al. Focal gains of vascular endothelial growth factor A and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68(16):6779–6788. doi:10.1158/0008-5472.CAN-08-0742.Focal18701503
  • YangC, HuangX, LiuZ, QinW, WangC. Metabolism-associated molecular classification of hepatocellular carcinoma. Mol Oncol. 2020;14(4):896–913. doi:10.1002/1878-0261.1263931955511
  • FornerA, ReigM, BruixJ. Hepatocellular carcinoma. Lancet. 2018;391(10127):1301–1314. doi:10.1016/S0140-6736(18)30010-229307467
  • LlovetJM, RealMI, MontañaX, et al. Arterial embolisation or chemoembolisation versus symptomatic treatment in patients with unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet. 2002;359(9319):1734–1739. doi:10.1016/S0140-6736(02)08649-X12049862
  • BruixJ, ShermanM. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–1022. doi:10.1002/hep.2419921374666
  • QuetglasIM, MoeiniA, PinyolR, LlovetJM. Integration of genomic information in the clinical management of HCC. Best Pract Res Clin Gastroenterol. 2014;28(5):831–842. doi:10.1016/j.bpg.2014.08.00425260311
  • PsyrriA, ArkadopoulosN, VassilakopoulouM, SmyrniotisV, DimitriadisG. Pathways and targets in hepatocellular carcinoma. Expert Rev Anticancer Ther. 2012;12(10):1347–1357. doi:10.1586/era.12.11323176622
  • DimriM, SatyanarayanaA. Molecular signaling pathways and therapeutic targets in hepatocellular carcinoma. Cancers (Basel). 2020;12(2):491. doi:10.3390/cancers12020491
  • MatterMS, DecaensT, AndersenJB, ThorgeirssonSS. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J Hepatol. 2014;60(4):855–865. doi:10.1016/j.jhep.2013.11.031.Targeting24308993
  • XieJ, WangX, ProudCG. mTOR inhibitors in cancer therapy. F1000Research. 2016;5:2078. doi:10.12688/f1000research.9207.1
  • AliMU, Ur RahmanMS, JiaZ, JiangC. Eukaryotic translation initiation factors and cancer. Tumor Biol. 2017;39(6):101042831770980. doi:10.1177/1010428317709805
  • SpilkaR, ErnstC, MehtaAK, HaybaeckJ. Eukaryotic translation initiation factors in cancer development and progression. Cancer Lett. 2013;340(1):9–21. doi:10.1016/j.canlet.2013.06.01923830805
  • VoorheesRM, RamakrishnanV. Structural basis of the translational elongation cycle. Annu Rev Biochem. 2013;82:203–236. doi:10.1146/annurev-biochem-113009-09231323746255
  • KappLD, LorschJR. The molecular mechanics of eukaryotic translation. Ann Rev Biochem. 2004;73:657–704. doi:10.1146/annurev.biochem.73.030403.08041915189156
  • BhatM, RobichaudN, HuleaL, SonenbergN, PelletierJ, TopisirovicI. Targeting the translation machinery in cancer. Nat Rev Drug Discov. 2015;14(4):261–278. doi:10.1038/nrd450525743081
  • SahinF, KannangaiR, AdegbolaO, WangJ, SuG, TorbensonM. mTOR and P70 S6 kinase expression in primary liver neoplasms. Clin Cancer Res. 2004;10(24):8421–8425. doi:10.1158/1078-0432.CCR-04-094115623621
  • LuX, PaliogiannisP, CalvisiDF, ChenX. Role of the mammalian target of rapamycin pathway in liver cancer: from molecular genetics to targeted therapies. Hepatology. 2021;73(S1):49–61. doi:10.1002/hep.3131032394479
  • GuriY, ColombiM, DazertE, et al. mTORC2 promotes tumorigenesis via lipid synthesis. Cancer Cell. 2017;32(6):807–823.e12. doi:10.1016/j.ccell.2017.11.01129232555
  • ByrneCD, TargherG. NAFLD: a multisystem disease. J Hepatol. 2015;62(1):S47–S64. doi:10.1016/j.jhep.2014.12.01225920090
  • FriedmanSL, Neuschwander-TetriBA, RinellaM, SanyalAJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24. doi:10.1038/s41591-018-0104-9
  • LallyJSV, GhoshalS, DeperaltaDK, et al. Inhibition of acetyl-CoA carboxylase (ACC) by phosphorylation or by the liver-specific inhibitor, ND-654, suppresses lipogenesis and hepatocellular carcinoma. Cell Metabol. 2019;29(1):174–182. DOI:10.1016/j.cmet.2018.08.020.Inhibition
  • ChenH, ShenF, SherbanA, et al. DEPTOR suppresses lipogenesis and ameliorates hepatic steatosis and acute-on-chronic liver injury in alcoholic liver disease. Hepatology. 2018;68(2):496–514. doi:10.1002/hep.2984929457836
  • CeniE, MelloT, GalliA. Pathogenesis of alcoholic liver disease: role of oxidative metabolism. World J Gastroenterol. 2014;20(47):17756–17772. doi:10.3748/wjg.v20.i47.1775625548474
  • SidB, VerraxJ, CalderonPB. Role of AMPK activation in oxidative cell damage: implications for alcohol-induced liver disease. Biochem Pharmacol. 2013;86(2):200–209. doi:10.1016/j.bcp.2013.05.00723688501
  • XieG, WangX, HuangF, et al. Dysregulated hepatic bile acids collaboratively promote liver carcinogenesis. Int J Cancer. 2016;139(8):1764–1775. doi:10.1002/ijc.3021927273788
  • RobertsLO, JoplingCL, JacksonRJ, WillisAE. Viral strategies to subvert the mammalian translation machinery. Prog Mol Biol Transl Sci. 2009;90(C):313–367. doi:10.1016/S1877-1173(09)90009-620374746
  • CoorayS. The pivotal role of phosphatidylinositol 3-kinase-Akt signal transduction in virus survival. J Gen Virol. 2004;85(5):1065–1076. doi:10.1099/vir.0.19771-015105524
  • StreetA, MacdonaldA, McCormickC, HarrisM. Hepatitis C virus NS5A-mediated activation of phosphoinositide 3-kinase results in stabilization of cellular β-catenin and stimulation of β-catenin-responsive transcription. J Virol. 2005;79(8):5006–5016. doi:10.1128/jvi.79.8.5006-5016.200515795286
  • StreetA, MacdonaldA, CrowderK, HarrisM. The hepatitis C virus NS5A protein activates a phosphoinositide 3-kinase-dependent survival signaling cascade. J Biol Chem. 2004;279(13):12232–12241. doi:10.1074/jbc.M31224520014709551
  • XiangK, WangB. Role of the PI3K-AKT-mTOR pathway in hepatitis B virus infection and replication. Mol Med Rep. 2018;17(3):4713–4719. doi:10.3892/mmr.2018.839529328380
  • NeuveutC, WeiY, BuendiaMA. Mechanisms of HBV-related hepatocarcinogenesis. J Hepatol. 2010;52(4):594–604. doi:10.1016/j.jhep.2009.10.03320185200
  • WangP, GuoQS, WangZW, QianHX. HBx induces HepG-2 cells autophagy through PI3K/Akt-mTOR pathway. Mol Cell Biochem. 2013;372(1–2):161–168. doi:10.1007/s11010-012-1457-x23001846
  • GuoH, ZhouT, JiangD, et al. Regulation of hepatitis B virus replication by the phosphatidylinositol 3-kinase-Akt signal transduction pathway. J Virol. 2007;81(18):10072–10080. doi:10.1128/jvi.00541-0717609269
  • WangXL, CaiHP, GeJH, SuXF. Detection of eukaryotic translation initiation factor 4E and its clinical significance in hepatocellular carcinoma. World J Gastroenterol. 2012;18(20):2540–2544. doi:10.3748/wjg.v18.i20.254022654452
  • Golob-SchwarzlN, KrassnigS, ToeglhoferAM, et al. New liver cancer biomarkers: PI3K/AKT/mTOR pathway members and eukaryotic translation initiation factors. Eur J Cancer. 2017;83:56–70. doi:10.1016/j.ejca.2017.06.00328715695
  • SilveraD, FormentiSC, SchneiderRJ. Translational control in cancer. Nat Rev Cancer. 2010;10(4):254–266. doi:10.1038/nrc282420332778
  • WangYW, LinKT, ChenSC, et al. Overexpressed-eIF3I interacted and activated oncogenic Akt1 is a theranostic target in human hepatocellular carcinoma. Hepatology. 2013;58(1):239–250. doi:10.1002/hep.2635223460382
  • ZhangL, ChenY, BaoC, ZhangX, LiH. Eukaryotic initiation Factor 4AIII facilitates hepatocellular carcinoma cell proliferation, migration, and epithelial-mesenchymal transition process via antagonistically binding to WD repeat domain 66 with miRNA-2113. J Cell Physiol. 2020;235(11):8199–8209. doi:10.1002/jcp.2947531975383
  • AhmedCS, WinlowPL, ParsonsAL, JoplingCL. Eukaryotic translation initiation factor 4AII contributes to microRNA-122 regulation of hepatitis C virus replication. Nucleic Acids Res. 2018;46(12):6330–6343. doi:10.1093/nar/gky26229669014
  • LiuY, SunL, SuX, GuoS. Inhibition of eukaryotic initiation factor 4E phosphorylation by cercosporamide selectively suppresses angiogenesis, growth and survival of human hepatocellular carcinoma. Biomed Pharmacother. 2016;84:237–243. doi:10.1016/j.biopha.2016.09.03827662474
  • FangC, XieH, ZhaoJ, et al. eIF4E-eIF4G complex inhibition synergistically enhances the effect of sorafenib in hepatocellular carcinoma. Anti Cancer Drugs. 2021;1–7. doi:10.1097/CAD.000000000000107432932275
  • LeeNP, TsangFH, ShekFH, et al. Prognostic significance and therapeutic potential of eukaryotic translation initiation factor 5A (eIF5A) in hepatocellular carcinoma. Int J Cancer. 2010;127(4):968–976. doi:10.1002/ijc.2510019998337
  • ShekFH, FatimaS, LeeNP. Implications of the use of eukaryotic translation initiation factor 5A (eIF5A) for prognosis and treatment of hepatocellular carcinoma. Int J Hepatol. 2012;2012:1–6. doi:10.1155/2012/760928
  • LouB, FanJ, WangK, et al. N1-guanyl-1,7-diaminoheptane (GC7) enhances the therapeutic efficacy of doxorubicin by inhibiting activation of eukaryotic translation initiation factor 5A2 (eIF5A2) and preventing the epithelial-mesenchymal transition in hepatocellular carcinoma cells. Exp Cell Res. 2013;319(17):2708–2717. doi:10.1016/j.yexcr.2013.08.01023958463
  • TangY, ChenK, LuanX, et al. Knockdown of eukaryotic translation initiation factor 5A2 enhances the therapeutic efficiency of doxorubicin in hepatocellular carcinoma cells by triggering lethal autophagy. Int J Oncol. 2020;57(6):1368–1380. doi:10.3892/ijo.2020.514333174013
  • CaoTT, LinSH, FuL, et al. Eukaryotic translation initiation factor 5A2 promotes metabolic reprogramming in hepatocellular carcinoma cells. Carcinogenesis. 2017;38(1):94–104. doi:10.1093/carcin/bgw11927879277
  • LiuRR, LvYS, TangYX, et al. Eukaryotic translation initiation factor 5A2 regulates the migration and invasion of hepatocellular carcinoma cells via pathways involving reactive oxygen species. Oncotarget. 2016;7(17):24348–24360. doi:10.18632/oncotarget.832427028999
  • TangDJ, DongSS, MaNF, et al. Overexpression of eukaryotic initiation factor 5A2 enhances cell motility and promotes tumor metastasis in hepatocellular carcinoma. Hepatology. 2010;51(4):1255–1263. doi:10.1002/hep.2345120112425
  • WangZG, ZhengH, GaoW, et al. eIF5B increases ASAP1 expression to promote HCC proliferation and invasion. Oncotarget. 2016;7(38):62327–62339. doi:10.18632/oncotarget.1146927694689
  • LiuZ, MoH, SunL, et al. Long noncoding RNA PICSAR/miR-588/EIF6 axis regulates tumorigenesis of hepatocellular carcinoma by activating PI3K/AKT/mTOR signaling pathway. Cancer Sci. 2020;111(11):4118–4128. doi:10.1111/cas.1463132860321
  • HuangJ, ZhengC, ShaoJ, ChenL, LiuX, ShaoJ. Overexpression of eEF1A1 regulates G1-phase progression to promote HCC proliferation through the STAT1-cyclin D1 pathway. Biochem Biophys Res Commun. 2017;494(3–4):542–549. doi:10.1016/j.bbrc.2017.10.11629079187
  • GrassiG, ScaggianteB, FarraR, et al. The expression levels of the translational factors eEF1A 1/2 correlate with cell growth but not apoptosis in hepatocellular carcinoma cell lines with different differentiation grade. Biochimie. 2007;89(12):1544–1552. doi:10.1016/j.biochi.2007.07.00717825975
  • ZhouY, LiY, XuS, et al. Eukaryotic elongation factor 2 kinase promotes angiogenesis in hepatocellular carcinoma via PI3K/Akt and STAT3. Int J Cancer. 2020;146(5):1383–1395. doi:10.1002/ijc.3256031286509
  • PottLL, HagemannS, ReisH, et al. Eukaryotic elongation factor 2 is a prognostic marker and its kinase a potential therapeutic target in HCC. Oncotarget. 2017;8(7):11950–11962. doi:10.18632/oncotarget.1444728060762
  • GordanJD, KennedyEB, Abou-AlfaGK, et al. Systemic therapy for advanced hepatocellular carcinoma: ASCO guideline. J Clin Oncol. 2020;38(36):4317–4345. doi:10.1200/JCO.20.0267233197225
  • SchnitzbauerAA, FilmannN, AdamR, et al. mTOR inhibition is most beneficial after liver transplantation for hepatocellular carcinoma in patients with active tumors. Ann Surg. 2020;272(5):855–862. doi:10.1097/SLA.000000000000428032889867
  • GeisslerEK, SchnitzbauerAA, ZölkeC, et al. Sirolimus use in liver transplant recipients with hepatocellular carcinoma: a randomized, multicenter, open-label phase 3 trial. Transplantation. 2016;100(1):116–125. doi:10.1097/TP.000000000000096526555945
  • ShiahHS, ChenCY, DaiCY, et al. Randomised clinical trial: comparison of two everolimus dosing schedules in patients with advanced hepatocellular carcinoma. Aliment Pharmacol Ther. 2013;37(1):62–73. doi:10.1111/apt.1213223134470
  • LlovetJM, RicciS, MazzaferroV, et al. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359(4):378–390. doi:10.1056/nejmoa070885718650514
  • KoeberleD, DufourJF, DemeterG, et al. Sorafenib with or without everolimus in patients with advanced hepatocellular carcinoma (HCC): a randomized multicenter, multinational Phase II trial (SAKK 77/08 and SASL 29). Ann Oncol. 2016;27(5):856–861. doi:10.1093/annonc/mdw05426884590
  • HuangTE, DengYN, HsuJL, et al. Evaluation of the anticancer activity of a bile acid-dihydroartemisinin hybrid ursodeoxycholic-dihydroartemisinin in hepatocellular carcinoma cells. Front Pharmacol. 2020;11:1–14. doi:10.3389/fphar.2020.59906732116689
  • PezzutoF, BuonaguroL, BuonaguroFM, TorneselloML. The role of circulating free DNA and microRNA in non-invasive diagnosis of HBV- and HCV-related hepatocellular carcinoma. Int J Mol Sci. 2018;19(4):1007. doi:10.3390/ijms19041007
  • MaraveliaP, SilvaDN, RovestiG, et al. Liquid biopsy in hepatocellular carcinoma: opportunities and challenges for immunotherapy. Cancers. 2021;1–19.
  • KasebAO, SanchezNS, SenS, et al. Molecular profiling of hepatocellular carcinoma using circulating cell-free DNA. Clin Cancer Res. 2019;25(20):6107–6118. doi:10.1158/1078-0432.CCR-18-334131363003
  • DemoryA, NaultJC. Molecular perspectives for the treatment of hepatocellular carcinoma. Acta Gastroenterol Belg. 2020;83(2):309–312.32603051