Abstract
Chronic fibrotic liver disease caused by viral or metabolic etiologies is a high-risk condition for developing hepatocellular carcinoma (HCC). Even after curative treatment of early-stage HCC tumor, the carcinogenic microenvironment persists in the remnant diseased liver and supports development of de novo HCC tumors (de novo HCC recurrence). Therefore, prevention of HCC development in patients at risk of not only first primary but also second primary HCC tumors is theoretically the most impactful strategy to improve patient prognosis. However, no such therapy has been established to date. One major challenge is identification of clinically relevant targets, which can be achieved by utilizing the reverse-engineering strategy of chemoprevention discovery, integrating omics information from clinical cohorts with completed follow-up for cancer development. Clinical and experimental studies have suggested etiology-specific and generic candidate HCC chemoprevention strategies, including statins, antidiabetic drugs, selective molecular targeted agents, and dietary and nutritional substances. Clinical testing of the candidate compounds can be cost-effectively performed by combining it with HCC risk biomarker evaluation to specify the target patient population most likely benefit from the therapy. Non-toxic, generic agents will have broad clinical applicability across the diverse HCC etiologies and clinical contexts, and are expected to substantially improve the still dismal prognosis of HCC.
Keywords: hepatocellular carcinoma, chemoprevention, cirrhosis, precision medicine, recurrence
Introduction
Hepatocellular carcinoma (HCC) is the major histological type of primary liver cancer, and the second leading cause of cancer mortality globally.1–4 In the United States, HCC mortality has been rising over the past 30 years across virtually all regions of the nation, especially along the Texas-Mexico border.5 HCC typically develops in chronically diseased livers with progressive fibrosis caused by viral and metabolic etiologies.6 Rapidly emerging predisposing conditions include non-alcoholic fatty liver disease (NAFLD), estimated to affect one-third to one-fourth of global population due to the obesity epidemic and metabolic disorders such as diabetes.7 Chronic infection by hepatitis C virus (HCV) has been a major HCC etiology, especially in developed countries.6 The recent development and widespread use of direct-acting antivirals (DAAs) have enabled a revolutionary improvement in viral clearance rate. Despite earlier concern about the higher incidence of HCC development and recurrence after DAA-based treatment compared to interferon-based regimens, more recent meta-analyses reported no obvious difference between the two strategies.8,9 However, the risk of developing HCC persists even after a decade of viral cure.8 It is predicted that HCV-related HCC incidence will keep increasing in the coming decades unless HCV diagnosis and treatment uptake rates are substantially improved.6,8
It is important to note that the magnitude of HCC risk needs to be determined according to each etiology and specific clinical scenario.10 It is clinically well known that male patients are more susceptible to HCC development in all major etiologies,11,12 including NAFLD. Studies have suggested the involvement of several molecular pathways in the sex disparity, including estrogen receptor signaling, the interleukin (IL)-6 pathway, and nuclear receptor signaling, such as through the peroxisome proliferator-activated receptor (PPAR) γ, liver × receptors, and farnesoid × receptor. These pathways may need to be taken into account when developing HCC chemopreventive strategies according to sex.13–21 Besides the major viral and metabolic etiologies of HCC, several predisposing factors have been noted. Family history has been implicated, with higher HCC risk in hepatitis B virus (HBV)/HCV–infected population.22 Type 2 diabetes and obesity have been shown to independently increase the risk of developing HCC.12,23,24 A study suggested that thalassemia with iron overload accompanied by chronic infection with HBV or HCV could contribute to hepatic injury, which could ultimately lead to HCC development.25 Thus, prognostication with regard to HCC risk under specific clinical scenarios is critical to identify the patients who would most benefit from the preventive interventions studied in recent publications.26–28
In clinical practice guidelines, regular biannual HCC screening is recommended as a measure for early tumor detection in patients with predisposing conditions, although it is still unclear how to screen the emerging at-risk population having relatively low annual incidence rates.6,29,30 More advanced liver fibrosis and portal hypertension have been implicated in increased risk of HCC, whereas HCC can also develop in non-cirrhotic livers in some etiologies, that is, HBV infection and NAFLD.31–34 Localized early-stage HCC tumors mainly diagnosed by contrast-enhanced CT or MRI can be curatively treated by surgical resection, thermal ablation, or liver transplantation. However, even after application of these potentially curative therapies, approximately 70% of the cases suffer tumor recurrence in the remnant diseased liver within 5 years after treatment. Once the tumors have progressed to an advanced stage, the survival benefit of currently available medical therapies is limited to several months and no curative treatment option is available.6,35–42 Thus, chemoprevention of HCC development in patients at risk is theoretically the most impactful strategy, although no such therapy has been established to date.
Clinically, HCC recurrence after curative treatment can be classified into two groups, namely growth of disseminated cancer cells from the treated primary tumor (disseminative recurrence), and development of second primary cancer clonally unrelated to the treated first primary tumor (de novo recurrence).6 Early tumor detection by the regular screening can control the former, but not the latter, which needs chemopreventive intervention. In this article, we provide an overview of generic HCC chemoprevention strategies that are potentially applicable across diverse HCC etiologies and clinical scenarios (Table 1).
Table 1.
Category of agent | Agent | Type of prevention | Study type(s) | Observed effect size |
---|---|---|---|---|
Anti-inflammatory agent | Aspirin | Secondary | Cohort, meta-analysis | Pooled HR = 0.68 |
Metabolic disorder correction | Metformin | Secondary | Case-control, meta-analysis | Pooled OR = 0.52 |
Statins | Secondary | Cohort | HR = 0.29–0.66 | |
Dietary/nutritional agent | Coffee/caffeine | Secondary | Case-control, cohort, meta-analysis | Pooled OR = 0.53–0.71, HR = 0.41 |
Vitamin D/25(OH)D | Secondary | Case-control, cohort | OR = 0.51, HR = 0.53 | |
Targeted agent | mTOR inhibitors | Tertiary | Clinical trial | HR = 0.84 |
Peretinoin | Secondary | Clinical trial | HR = 0.73 (entire study period), HR = 0.27 (> 2 years) |
HCC, hepatocellular carcinoma; HR, hazard ratio; OR, odds ratio; BCAA, branched-chain amino acids. See Ref. 6 for definition of type of prevention.
Anti-inflammatory and anti-fibrotic therapies
Progressive liver fibrosis and subsequent carcinogenesis are typically accompanied by chronic inflammation in the liver that causes hepatocyte injury and regeneration. Therefore, anti-inflammatory therapies have been sought as potential measures for HCC chemoprevention. In HCV-infected patients, low-dose maintenance interferon therapy has been extensively tested for its capability to reduce hepatic inflammation and HCC incidence.6,43–45 Although HCC reduction was observed in patient with more advanced fibrosis and who could complete the planned regimens, the high dropout rate due to toxicity precludes its clinical adaptation. There is conflicting epidemiological evidence about HCC-preventive effect of non-steroidal anti-inflammatory drugs (NSAIDs), including asprin.46 In a meta-analysis of ten U.S. patient cohorts involving 679 HCC cases, aspirin shows a protective effect against HCC development at a hazard ratio of 0.68.47 In mice, the gut microbial environment can induce innate immune signaling through TLR4 and support transformation of neoplastic cells in the liver; these effects are inhibited by gut sterilization.48
Experimental studies have suggested that antifibrotic therapies may serve as HCC chemoprevention by resolving the fibrosis that provides a tissue microenvironment supporting carcinogenesis.49 Several recent clinical trials of antifibrotic agents have shown promising results. Selonsertib, a small molecule inhibitor of apoptosis signal-regulating kinase 1 (ASK1), reduced fibrosis in patients with non-alcoholic steatohepatitis (NASH).50 Cenicriviroc, a dual inhibitor of C-C motif chemokine receptor 2/5 (CCR2/CCR5), reduced liver fibrosis in NASH patients,51 and is now being followed up in a Phase III trial (NCT03028740).
PPARs are nuclear receptor members that transcriptionally regulate several essential processes, including fatty acid metabolism, to maintain energy equilibrium.49 Elafibranor, an agonist of PPARα/δ, reduced steatohepatitis without worsening fibrosis in a phase II trial in NASH patients,52 which led to a phase III trial (NCT02704403). Extended clinical observation in these trials may provide insights about antifibrotic therapy as an option for HCC chemoprevention.
Treatment of metabolic disorders
Metformin, a biguanide derivative, is one of the first-line therapies for type 2 diabetes; it reduces hepatic gluconeogenesis via activation of adenosine monophosphate-activated protein kinase (AMPK) and inhibition of the mitochondrial respiratory chain, cyclic adenosine monophosphate (cAMP), and protein kinase A (PKA) activity, among other mechanisms. Experimental studies have also reported anti-cancer activities for metformin, including blockade of the mammalian target of rapamycin (mTOR) pathway by AMPK activation; antiangiogenesis by suppressing vascular endothelial growth factor (VEGF); induction of cell cycle arrest by decreasing cyclin D1 expression; and induction of cell death by suppressing nuclear factor-κB (NF-κB) pathway or inducing apoptosis.53–57 Multiple epidemiological studies have suggested that metformin use is associated with reduced HCC incidence in patients with chronic liver diseases. A meta-analysis of 19 studies, involving > 500,000 diabetic patients, reported reduced HCC incidence in metformin users compared to placebo or non-users.58 Subgroup analysis showed consistent association with lower HCC incidence in patients with hepatitis virus infection, obesity, or cirrhosis. However, prospective trials have failed to validate these findings,6 and it still needs to be confirmed in future trials. In a rat model of cirrhosis-driven hepatocarcinogenesis, metformin lowers HCC burden by suppressing activation of progenitor cells when the treatment is started earlier.59
Statins are 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors that are widely used for lowering circulating LDL cholesterol levels60,61. Besides this effect, anti-cancer properties for statins have also been noted. In experimental studies, statins inhibit oncogenesis drivers such as Myc, Akt, integrins, Rho-dependent kinase, NF-κB, tumor necrosis factor (TNF)–mediated IL-6 production, the Hippo pathway, and extracellular signal–regulated kinase 1/2 (ERK1/2), whereas AMPK and p38/mitogen-activated protein kinase (MAPK) pathways are activated and p53-dependent apoptosis is induced.62–70 Statins also limit liver fibrogenesis in diet/chemical-induced rodent models by inhibiting hepatic stellate cell activation via nitric oxide synthase, paracrine signals from hepatocytes and endothelial cells, induction of sterol regulatory element-binding protein 1 (SREBP-1) and PPARs.71–74 Statins may reduce cirrhotic portal hypertension via non-canonical Hedgehog signaling.75 A case-control study of type 2 diabetes that included 229 HCC patients reported a reduced risk of HCC development in association with statin use in individuals with liver disease, and this reduction increased with higher statin doses.76,77 A similar association was observed in HBV- or HCV-infected patients.78,79 Analysis of > 7000 HCV-infected individuals in the U.S. Electronically Retrieved Cohort of HCV Infected Veterans (ERCHIVES) database showed that statin use was associated with approximately half the risk of HCC.80 Fibrosis progression was reduced by statins in the HALT-C cohort.81 In cirrhotic patients with HBV, HCV, alcoholism, as well as non-cirrhotic NAFLD, statin use reduced decompensation, death, and HCC.82,83 Meta-analyses of these retrospective observational studies reproduced the trend of reduced HCC incidence despite significant heterogeneity in the study population and confounding factors.17,84 However, the protective effect was not confirmed in meta-analyses of 27 prospective studies involving 175,000 individuals for various cancers, including HCC.84,85 In a systematic comparison based on meta-analysis of five observational studies, fluvastatin showed the highest association with reduced HCC risk compared to other statins.86 Atorvastatin and fluvastatin showed higher association with antifibrotic effects compared to lovastatin, pravastatin, rosuvastatin, and simvastatin.80 Prospective randomized clinical trials are currently underway to determine the role of statins in HCC chemoprevention (Table 2). A preventive effect of simvastatin in patients with cirrhosis is being tested in a phase II trial, using change in serum AFP-L3% as the primary endpoint (NCT02968810). Atorvastatin is being evaluated in a phase IV trial for prevention of de novo recurrence after HCC resection or ablation (NCT03024684).
Table 2.
Trial number | Agent | Type of agent | Phase | Target population | Primary endpoint | Estimated completion data |
---|---|---|---|---|---|---|
NCT02224456 | Tenofovir disoproxil fumarate | Anti-viral | IV | CHB with advanced fibrosis | HCC incidence and progression | 1/2021 |
NCT02968810 | Simvastatin | Statin | II | Cirrhosis | Change in AFP-L3 | 1/2020 |
NCT02779465 | Vitamin D3 | Dietary/nutritional supplement | IV | CHB on anti-viral | Change in serum levels of 25(OH)D | 12/2026 |
NCT02098785 | Caffeine citrate | Dietary/nutritional supplement | I | Healthy | VAP-1 serum levels | 9/2018 |
NCT02273362 | Erlotinib | Kinase inhibitor | I | Cirrhosis | Safety, pEGFR, 186-gene signature | 1/2019 |
NCT03024684 | Atorvastatin | Statin | IV | Early-stage HCC after curative ablation or hepatectomy | HCC recurrence | 1/2022 |
NCT03184493 | Metformin, celecoxib | Anti-inflammation, Anti-diabetic | III | HCC after hepatectomy | HCC recurrence | 6/2021 |
NCT02281266 | Thymalfasin | Immune modulator | IV | HBV-HCC after hepatectomy | Recurrence-free survival | 10/2018 |
NCT02686372 | HBV antigen specific TCR redirected T cell | Immune modulator | I | CHB after transplantation | Safety | 11/2020 |
NCT01924624 | Thalidomide | Immune modulator, anti-angiogenesis | IV | HCC after hepatectomy | Disease-free survival | 12/2019 |
NCT02447679 | Thalidomide, tegafur-uracil | Immune modulator, anti-angiogenesis, cytotoxic agent | II | HCC after hepatectomy | HCC recurrence | Completed, not reported |
NCT03178929 | SAM | Dietary/nutritional supplement | n.a. | Early-stage HCC after radical treatment | HCC recurrence | 8/2020 |
NCT01770431 | Huaier granule | Traditional herbal medicine | IV | HCC after hepatectomy | HCC recurrence, metastasis | Completed, not reported |
NCT02399033 | Xihuang capsules | Traditional herbal medicine | IV | HCC after hepatectomy | HCC recurrence | 12/2019 |
NCT01717066 | Ginsenoside Rg3 | Chemo-sensitizer, anti-angiogenesis | III | HCC after hepatectomy | HCC recurrence | Completed, not reported |
NCT00116454 | 131I-lipiodol | Cytotoxic agent | III | Viral/alcohol-HCC after hepatectomy or ablation (≤ 2 tumors) | HCC recurrence | Completed, not reported |
NCT02767375 | Hepatic arterial infusion chemotherapy | Cytotoxic agent | II-III | HCC after hepatectomy | Disease-free survival | 12/2018 |
HCC, hepatocellular carcinoma; CHB, chronic hepatitis B; AFP-L3, lens culinaris agglutinin-reactive fraction of α-fetoprotein; LSM, liver stiffness measurement; 25(OH)D, 25-hydroxy vitamin D; VAP-1, vascular adhesion protein 1; HBV, hepatitis B virus; TCR, T cell receptor; SAM, S-adenosylmethionine. Information from ClinicalTrials.gov, accessed in March 2018.
Life style change and dietary/nutritional agents
Coffee use has been associated with reduced HCC risk in a series of systematic reviews of multiple liver disease patient cohorts, irrespective of liver disease severity, alcohol usage, body mass index, diabetes, smoking, or hepatitis infection.87 Caffeinated coffee was associated with higher HCC reduction compared to decaffeinated coffee in some cohort studies. The reduced HCC risk with coffee use was associated with reduced inflammation and hepatic injury, based on biomarkers such as IL-6, hepatic transaminases, and γ-glutamyltransferase.88,89 In a multi-ethnic cohort of > 215,000 individuals, a dose-dependent inverse association was observed between coffee use and the progression of chronic liver disease caused by HCV, alcohol, and NAFLD.90 In a prospective cohort of > 480,000 participants that included 201 HCC cases, tea intake was associated with reduction of HCC risk to a lesser extent than coffee.91 Coffee consumption of more than 3 cups per day was associated with improved sensitivity to anti-HCV therapy.92 A phase I trial of caffeine citrate is planned to assess its effect in NASH on serum vascular adhesion protein 1 (VAP-1) level, hepatic inflammation, and fibrosis (NCT02098785).49 Experimentally, the caffeine analog CGS 15943 inhibits HCC cell growth by targeting the PI3K/AKT pathway.93
Dietary phytochemicals, including curcumin (from turmeric extract), resveratrol (a polyphenol in grapes, red wine, and berries), silymarin (a herbal flavonoid), and carotenoids, have been assessed as potential HCC chemoprevention agents, although clinical proof is still lacking.94 Shared features of this class of agents include activation of cytoprotective mechanisms such as the Keap1/Nrf2 pathway, as shown in experimental models.94 Markers of DNA damage such as 8-hydroxydeoxyguanosine (8-OHdG) in urine was reduced in humans by several agents, such as epigallocatechin gallate (EGCG, a green tea polyphenol) and broccoli sprouts, although their HCC-preventive effect has not yet been determined.94 Normalization of the levels of hepatic transaminases by glycyrrhizin (from licorice root extract) has been associated with reduced HCC incidence.95 S-adenosylmethionine (SAM), a ubiquitous, major methyl donor, suppresses HCC development in an experimental model.94 In a phase II trial of SAM in HCV cirrhosis, the levels of the biomarkers for liver injury and oxidative stress as well as of serum α-fetoprotein did not change in the subjects.96
Dietary intake of unsaturated fat was inversely associated with HCC incidence in a European cohort.97 In a population-based cohort of > 90,000 Japanese subjects, consumption of n-3 polyunsaturated fatty acid (PUFA)-rich fish and of individual n-3 PUFAs such as eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and docosahexaenoic acid (DHA) was associated with lower HCC incidence.98 Omega-3 PUFAs, DHA, and EPA inhibit cyclooxygenase 2 (COX2) and suppress HCC growth.99 Fat-1 transgenic mice, which endogenously form omega-3 PUFAs are protected from chemically-induced hepatocarcinogenesis and have reduced hepatic COX2 expression.100 White meat has been associated with a lower risk of chronic liver disease and HCC compared to red meat in a large-scale U.S. cohort.101
Low serum levels of vitamin D (25(OH)D3) have been associated with the risk of HCC, variceal bleeding, and ascites in patients with chronic hepatitis B, and with more frequent death in patients with HCC.102,103 Higher 25(OH)D levels have been associated with lower HCC risk.104 In alcoholic liver disease patients, low 25(OH)D levels have been associated with higher liver damage and mortality.105 Vitamin D3 up-regulated protein 1 (VDUP1) suppresses TNF and NF-κB signaling, and reduces chemical carcinogenesis in rat liver.106 Artificial sunlight therapy increases serum levels of 25(OH)D3 and reduces liver inflammation and fibrosis in NASH rats fed a choline-deficient, L-amino acid-defined (CDAA) diet.107 1,25(OH)2D3 inhibits thioacetamide (TAA)-induced fibrosis.108 SQSTM1 (p62) promotes heterodimerization of the vitamin D receptor (VDR) with retinoid × receptor (RXR) and inhibits hepatic stellate cell activation, liver fibrosis, and HCC progression.109 A phase IV trial of vitamin D3 is planned for the prevention of HBV-related HCC (NCT02779465).
Hereditary hemochromatosis is caused by HFE gene mutations (C282Y and H63D) and associated with increased iron content in the liver and elevated risk of HCC110–112. Iron depletion by chelate therapy and phlebotomy have been explored as measures for HCC chemoprevention not only in hemochromatosis patients but also in viral hepatitis patients, although none of these have been adopted in clinical care.94,113 Phlebotomy reduces hepatic iron content and inflammation and HCC in HCV-infected patients.114 Liver-specific β-catenin knockout increases susceptibility to excess dietary iron, which leads to the development of steatohepatitis, fibrosis, and HCC by activating the AKT, ERK, and NF-κB pathways in mice.115 Deferasirox, an iron chelator, upregulates hepcidin, transferrin receptor 1, and hypoxia inducible factor-1α, and suppresses chemically-induced HCC in mice; however, its toxicity limits its clinical use.116
Dietary supplementation with branched-chain amino acids (BCAA) is used for treatment of hepatic encephalopathy in cirrhotics.117 BCAA induces mTOR-mediated cellular senescence and suppresses liver fibrosis and HCC in rats.118,119 In HCV-transgenic mice, BCAA reduces hepatic iron accumulation by inducing hepcidin-25, which reduces reactive oxygen species.120 In a NASH mouse model, BCAA represses profibrogenic activation of hepatic stellate cells via TGF-β1 and reduces cellular transformation in an mTOR-dependent manner.121 In C57BL/KsJ-db/db obese mice, BCAA reduces inflammation in the liver and white adipose tissues, and inhibits spontaneous hepatic carcinogenesis by inducing PPARγ, p21CIP1, and p27KIP1, and suppressing IL-6, IL-1β, IL-18, and TNF.122 BCAA increased serum hepcidin-25 levels in HCV-infected patients with advanced liver fibrosis. In a prospective observational study, BCAA supplementation was associated with reduced incidence of HCC and death.123 N-acetyl-l-(+)-cysteine (NAC) protects the animals from hepatic injury and fibrogenesis.
Molecular targeted therapies
The phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway is involved in cell survival, making it a candidate HCC chemoprevention target.6 AKT was identified as a key HCC risk driver in a human hepatic transcriptome meta-analysis.124 Clinical studies have suggested that immunosuppression by mTOR pathway inhibition reduces post-transplantation HCC recurrence,125 but adverse effects such as arterial thrombosis and poorer patient and graft survival have been noted.126 In animal models of chemical/obesity-driven HCC, sirolimus (rapamycin) activates IL-6/signal transducer and activator of transcription 3 (STAT3) and enhances HCC development.127 Sirolimus and everolimus have been tested in prospective trials for prevention of post-transplantation HCC recurrence.128 A phase III trial of sirolimus enrolling 525 patients did not improve recurrence-free survival beyond 5 years after transplantation, although subgroup analyses suggested that less advanced tumors or younger age may predict a better outcome.129
Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are bioactive lipids that can regulate cell survival, differentiation, proliferation, and migration.130 In liver, hepatocyte-derived autotaxin (ATX), which converts lysophosphatidylcholine into LPA, is elevated in the serum of HCV-infected and NAFLD patients with liver fibrosis and HCC.131,132 In a transcriptome meta-analysis of human fibrotic livers, LPA receptor 1 (LPAR1) was identified as a pan-etiology HCC risk driver.124 Genetic knockout of ATX as well as pharmacological inhibition of ATX or LPAR1 attenuates progression of liver fibrosis and HCC development in rodent models.124,133
Activation of receptor tyrosine kinase signaling, such as through the epidermal growth factor receptor (EGFR) pathway, in hepatic stellate cells and macrophages has been implicated in hepatocarcinogenesis in rodent models.134,135 A small molecule EGFR inhibitor such as erlotinib reversed a gene signature pattern associated with a high risk of HCC and suppressed HCC development in rodent models.136 Based on these findings, a phase I trial of erlotinib has been started (NCT02273362). A multi-kinase inhibitor, sorafenib, did not affect recurrence-free survival after curative treatment of primary HCC tumors in a phase III trial.137
The renin-angiotensin system has been implicated in pathogenesis of liver fibrosis and HCC risk.94 NF-κB activation mediated by angiotensin II promotes fibrogenesis, which can be inhibited by captopril, an angiotensin-converting enzyme (ACE) inhibitor.138 Angiotensin II type 1 receptor blockers (ARBs) such as losartan and telmisartan can suppress hepatic stellate cell activation, reduce liver fibrosis, and prevent HCC in rodents.139,140 In a retrospective single-center study, ARB use was associated with delayed HCC recurrence and extended patient survival after HCC tumor ablation.141 An ACE inhibitor, perindopril, in combination with vitamin K2 or branched-chain amino acids, reduced HCC recurrence after curative treatment.142,143
Peretinoin (acyclic retinoid) is a synthetic vitamin A analog that inhibits the Wnt and platelet-derived growth factor (PDGF) pathways, induces cellular differentiation and apoptosis of hepatic stem cells, and apparently depletes neoplastic clones.144–146 Peretinoin also suppresses HCV replication and the release of infectious virus in vitro.147 In mice fed a high-fat diet, peretinoin induced autophagy and suppressed HCC development.148 In a phase III trial of peretinoin in patients with curatively treated HCV-related HCC, reduced HCC recurrence was observed after 2 years.149 A follow-up study reported extended overall survival in patients treated with the higher dose.150 Prospective trials in HBV-related HCC patients are underway.144
It has been speculated that the estrogen pathway underlies the sex disparity in HCC risk.151 Genetic variations in estrogen receptor 1 (ESR1) gene were associated with increased HCC risk in a meta-analysis of 87 studies.152 Estrogen replacement as post-menopausal hormone therapy was associated with a reduced risk of HCC and prolonged patient survival.153 The microRNA miR-101 is down-regulated in HCC tissue, and its administration inhibits known HCC chemoprevention targets such as COX2 and Rho-GTPase, and reduces HCC in mice.154
Conclusions
Given the diversity of HCC etiology and clinical conditions, generically applicable chemoprevention therapies will have broader clinical applicability compared to etiology-specific interventions such as anti-viral therapies. Moreover, such generic chemoprevention could be used in combination with etiology-specific therapies for potentially synergistic effects. Identification of clinically relevant HCC chemoprevention targets has been challenging due to the logistical difficulty in validating experimentally-derived hypothesis in humans, which typically requires long observation periods in large patient cohorts. Integrative omics analysis of clinical specimens with already completed clinical follow-up, so-called “reverse-engineering” chemoprevention discovery, may overcome this challenge.124 The prediction of drug toxicity is another critical factor because cirrhotic patients are the major target of chemopreventive therapy.155 Safe generic compounds in combination with HCC risk biomarkers will enable flexible and cost-effective HCC chemoprevention and contribute to substantial improvement of the dismal prognosis of HCC.
Acknowledgements
This work is supported by U.S. NIH/NIDDK R01 Grant DK099558, European Union Grant ERC-2014-AdG-671231 HEPCIR, the Irma T. Hirschl Trust, the U.S. Department of Defense W81XWH-16-1-0363, and the Cancer Prevention and Research Institute of Texas RR180016 (to Y.H.).
Footnotes
Competing interests
The authors declare no competing interests.
References
- 1.Singal AG & El-Serag HB Hepatocellular Carcinoma From Epidemiology to Prevention: Translating Knowledge into Practice. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 13, 2140–2151 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mittal S & El-Serag HB Epidemiology of hepatocellular carcinoma: consider the population. Journal of clinical gastroenterology 47 Suppl, S2–6 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.White DL, Kanwal F & El-Serag HB Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 10, 1342–1359.e1342 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.El-Serag HB Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 142, 1264–1273.e1261 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mokdad AH, et al. Trends and Patterns of Disparities in Cancer Mortality Among US Counties, 1980–2014. JAMA 317, 388–406 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fujiwara N, Friedman SL, Goossens N & Hoshida Y Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J Hepatol 68, 526–549 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Younossi Z, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat Rev Gastroenterol Hepatol 15, 11–20 (2018). [DOI] [PubMed] [Google Scholar]
- 8.Baumert TF, Juhling F, Ono A & Hoshida Y Hepatitis C-related hepatocellular carcinoma in the era of new generation antivirals. BMC medicine 15, 52 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Waziry R, et al. Hepatocellular carcinoma risk following direct-acting antiviral HCV therapy: A systematic review, meta-analyses, and meta-regression. Journal of hepatology 67, 1204–1212 (2017). [DOI] [PubMed] [Google Scholar]
- 10.Fattovich G, Stroffolini T, Zagni I & Donato F Hepatocellular carcinoma in cirrhosis: incidence and risk factors. Gastroenterology 127, S35–50 (2004). [DOI] [PubMed] [Google Scholar]
- 11.Ballestri S, et al. NAFLD as a Sexual Dimorphic Disease: Role of Gender and Reproductive Status in the Development and Progression of Nonalcoholic Fatty Liver Disease and Inherent Cardiovascular Risk. Adv Ther 34, 1291–1326 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ballestri S, Nascimbeni F, Romagnoli D, Baldelli E & Lonardo A The Role of Nuclear Receptors in the Pathophysiology, Natural Course, and Drug Treatment of NAFLD in Humans. Adv Ther 33, 291–319 (2016). [DOI] [PubMed] [Google Scholar]
- 13.Falleti E, et al. Interleukin-6 polymorphisms and gender: relationship with the occurrence of hepatocellular carcinoma in patients with end-stage liver disease. Oncology 77, 304–313 (2009). [DOI] [PubMed] [Google Scholar]
- 14.Nakagawa H, et al. Serum IL-6 levels and the risk for hepatocarcinogenesis in chronic hepatitis C patients: an analysis based on gender differences. Int J Cancer 125, 2264–2269 (2009). [DOI] [PubMed] [Google Scholar]
- 15.Aleksandrova K, et al. Inflammatory and metabolic biomarkers and risk of liver and biliary tract cancer. Hepatology (Baltimore, Md.) 60, 858–871 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ohishi W, et al. Serum interleukin-6 associated with hepatocellular carcinoma risk: a nested case-control study. International journal of cancer 134, 154–163 (2014). [DOI] [PubMed] [Google Scholar]
- 17.Shi M, Zheng H, Nie B, Gong W & Cui X Statin use and risk of liver cancer: an update meta-analysis. BMJ Open 4, e005399 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tang S, et al. Genetic polymorphism of interleukin-6 influences susceptibility to HBV-related hepatocellular carcinoma in a male Chinese Han population. Human immunology 75, 297–301 (2014). [DOI] [PubMed] [Google Scholar]
- 19.Naugler WE, et al. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science (New York, N.Y.) 317, 121–124 (2007). [DOI] [PubMed] [Google Scholar]
- 20.Sander LE, Trautwein C & Liedtke C Is interleukin-6 a gender-specific risk factor for liver cancer? Hepatology (Baltimore, Md.) 46, 1304–1305 (2007). [DOI] [PubMed] [Google Scholar]
- 21.Prieto J Inflammation, HCC and sex: IL-6 in the centre of the triangle. Journal of hepatology 48, 380–381 (2008). [DOI] [PubMed] [Google Scholar]
- 22.Hassan MM, et al. The association of family history of liver cancer with hepatocellular carcinoma: a case-control study in the United States. Journal of hepatology 50, 334–341 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Reeves HL, Zaki MY & Day CP Hepatocellular Carcinoma in Obesity, Type 2 Diabetes, and NAFLD. Digestive diseases and sciences 61, 1234–1245 (2016). [DOI] [PubMed] [Google Scholar]
- 24.Loria P, Lonardo A & Anania F Liver and diabetes. A vicious circle. Hepatology research : the official journal of the Japan Society of Hepatology 43, 51–64 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Moukhadder HM, Halawi R, Cappellini MD & Taher AT Hepatocellular carcinoma as an emerging morbidity in the thalassemia syndromes: A comprehensive review. Cancer 123, 751–758 (2017). [DOI] [PubMed] [Google Scholar]
- 26.Faillaci F, et al. Liver Angiopoietin-2 is a key predictor of de novo or recurrent hepatocellular cancer after HCV direct-acting antivirals. Hepatology (Baltimore, Md.) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Villa E, et al. Neoangiogenesis-related genes are hallmarks of fast-growing hepatocellular carcinomas and worst survival. Results from a prospective study. Gut 65, 861–869 (2016). [DOI] [PubMed] [Google Scholar]
- 28.Coulon S, et al. Angiogenesis in chronic liver disease and its complications. Liver international : official journal of the International Association for the Study of the Liver 31, 146–162 (2011). [DOI] [PubMed] [Google Scholar]
- 29.Heimbach JK, et al. AASLD guidelines for the treatment of hepatocellular carcinoma. Hepatology 67, 358–380 (2018). [DOI] [PubMed] [Google Scholar]
- 30.Tzartzeva K, et al. Surveillance Imaging and Alpha Fetoprotein for Early Detection of Hepatocellular Carcinoma in Patients With Cirrhosis: A Meta-analysis. Gastroenterology (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Baffy G Origins of Portal Hypertension in Nonalcoholic Fatty Liver Disease. Digestive diseases and sciences 63, 563–576 (2018). [DOI] [PubMed] [Google Scholar]
- 32.AISF position paper on nonalcoholic fatty liver disease (NAFLD): Updates and future directions. Digestive and liver disease : official journal of the Italian Society of Gastroenterology and the Italian Association for the Study of the Liver 49, 471–483 (2017). [DOI] [PubMed] [Google Scholar]
- 33.Piscaglia F, et al. Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: A multicenter prospective study. Hepatology 63, 827–838 (2016). [DOI] [PubMed] [Google Scholar]
- 34.Singh S, Muir AJ, Dieterich DT & Falck-Ytter YT American Gastroenterological Association Institute Technical Review on the Role of Elastography in Chronic Liver Diseases. Gastroenterology 152, 1544–1577 (2017). [DOI] [PubMed] [Google Scholar]
- 35.Bruix J, Reig M & Sherman M Evidence-Based Diagnosis, Staging, and Treatment of Patients With Hepatocellular Carcinoma. Gastroenterology 150, 835–853 (2016). [DOI] [PubMed] [Google Scholar]
- 36.Dhir M, et al. A Review and Update of Treatment Options and Controversies in the Management of Hepatocellular Carcinoma. Annals of surgery 263, 1112–1125 (2016). [DOI] [PubMed] [Google Scholar]
- 37.Monga SP beta-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology 148, 1294–1310 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bruix J, Gores GJ & Mazzaferro V Hepatocellular carcinoma: clinical frontiers and perspectives. Gut 63, 844–855 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fong ZV & Tanabe KK The clinical management of hepatocellular carcinoma in the United States, Europe, and Asia: a comprehensive and evidence-based comparison and review. Cancer 120, 2824–2838 (2014). [DOI] [PubMed] [Google Scholar]
- 40.Gaddikeri S, et al. Hepatocellular carcinoma in the noncirrhotic liver. AJR. American journal of roentgenology 203, W34–47 (2014). [DOI] [PubMed] [Google Scholar]
- 41.Kansagara D, et al. Screening for hepatocellular carcinoma in chronic liver disease: a systematic review. Annals of internal medicine 161, 261–269 (2014). [DOI] [PubMed] [Google Scholar]
- 42.Knudsen ES, Gopal P & Singal AG The changing landscape of hepatocellular carcinoma: etiology, genetics, and therapy. The American journal of pathology 184, 574–583 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bruix J, et al. Maintenance therapy with peginterferon Alfa-2b does not prevent hepatocellular carcinoma in cirrhotic patients with chronic hepatitis C. Gastroenterology 140, 1990–1999 (2011). [DOI] [PubMed] [Google Scholar]
- 44.Lok AS, et al. Maintenance peginterferon therapy and other factors associated with hepatocellular carcinoma in patients with advanced hepatitis C. Gastroenterology 140, 840–849 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hoshida Y, Fuchs BC, Bardeesy N, Baumert TF & Chung RT Pathogenesis and prevention of hepatitis C virus-induced hepatocellular carcinoma. Journal of Hepatology 61, S79–S90 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Singh S, Singh PP, Roberts LR & Sanchez W Chemopreventive strategies in hepatocellular carcinoma. Nature Reviews Gastroenterology and Hepatology 11, 45–54 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Petrick JL, et al. Nsaid use and risk of hepatocellular carcinoma and intrahepatic cholangiocarcinoma: The liver cancer pooling project. Cancer Prevention Research 8, 1156–1162 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dapito D, et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 21, 504–516 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Higashi T, Friedman SL & Hoshida Y Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev 121, 27–42 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Loomba R, et al. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: A randomized, phase 2 trial. Hepatology (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Friedman SL, et al. A Randomized, Placebo-Controlled Trial of Cenicriviroc for Treatment of Nonalcoholic Steatohepatitis with Fibrosis. Hepatology (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ratziu V, et al. Elafibranor, an Agonist of the Peroxisome Proliferator-Activated Receptor-α and -δ, Induces Resolution of Nonalcoholic Steatohepatitis Without Fibrosis Worsening. Gastroenterology 150, 1147–1159e1145 (2016). [DOI] [PubMed] [Google Scholar]
- 53.Zheng L, et al. Prognostic significance of AMPK activation and therapeutic effects of metformin in hepatocellular carcinoma. Clinical Cancer Research 19, 5372–5380 (2013). [DOI] [PubMed] [Google Scholar]
- 54.Zhou X, et al. Metformin suppresses hypoxia-induced stabilization of HIF-1α through reprogramming of oxygen metabolism in hepatocellular carcinoma. Oncotarget 7, 873–884 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chen HP, et al. Metformin decreases hepatocellular carcinoma risk in a dose-dependent manner: population-based and in vitro studies. Gut 62, 606–615 (2013). [DOI] [PubMed] [Google Scholar]
- 56.Buzzai M, et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Research 67, 6745–6752 (2007). [DOI] [PubMed] [Google Scholar]
- 57.Tsai HH, et al. Metformin promotes apoptosis in hepatocellular carcinoma through the CEBPD-induced autophagy pathway. Oncotarget 8, 13832–13845 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Cali G, et al. Natural history of Cesarean scar pregnancy on prenatal ultrasound: the crossover sign. Ultrasound Obstet Gynecol 50, 100–104 (2017). [DOI] [PubMed] [Google Scholar]
- 59.DePeralta DK, et al. Metformin prevents hepatocellular carcinoma development by suppressing hepatic progenitor cell activation in a rat model of cirrhosis. Cancer 122, 1216–1227 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lonardo A & Loria P Potential for statins in the chemoprevention and management of hepatocellular carcinoma. Journal of gastroenterology and hepatology 27, 1654–1664 (2012). [DOI] [PubMed] [Google Scholar]
- 61.Argo CK, Loria P, Caldwell SH & Lonardo A Statins in liver disease: a molehill, an iceberg, or neither? Hepatology (Baltimore, Md.) 48, 662–669 (2008). [DOI] [PubMed] [Google Scholar]
- 62.Higashi T, et al. Statin attenuates cell proliferative ability via TAZ (WWTR1) in hepatocellular carcinoma. Med Oncol 33, 123 (2016). [DOI] [PubMed] [Google Scholar]
- 63.Wang J, Tokoro T, Higa S & Kitajima I Anti-inflammatory effect of pitavastatin on NF-kappaB activated by TNF-alpha in hepatocellular carcinoma cells. Biological & pharmaceutical bulletin 29, 634–639 (2006). [DOI] [PubMed] [Google Scholar]
- 64.Relja B, et al. Simvastatin modulates the adhesion and growth of hepatocellular carcinoma cells via decrease of integrin expression and ROCK. Int J Oncol 38, 879–885 (2011). [DOI] [PubMed] [Google Scholar]
- 65.Roudier E, Mistafa O & Stenius U Statins induce mammalian target of rapamycin (mTOR)-mediated inhibition of Akt signaling and sensitize p53-deficient cells to cytostatic drugs. Molecular cancer therapeutics 5, 2706–2715 (2006). [DOI] [PubMed] [Google Scholar]
- 66.Ghalali A, Martin-Renedo J, Hogberg J & Stenius U Atorvastatin Decreases HBx-Induced Phospho-Akt in Hepatocytes via P2X Receptors. Mol Cancer Res 15, 714–722 (2017). [DOI] [PubMed] [Google Scholar]
- 67.Cao Z, et al. MYC phosphorylation, activation, and tumorigenic potential in hepatocellular carcinoma are regulated by HMG-CoA reductase. Cancer Res 71, 2286–2297 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yang PM, et al. Inhibition of autophagy enhances anticancer effects of atorvastatin in digestive malignancies. Cancer Res 70, 7699–7709 (2010). [DOI] [PubMed] [Google Scholar]
- 69.Sutter AP, et al. Cell cycle arrest and apoptosis induction in hepatocellular carcinoma cells by HMG-CoA reductase inhibitors. Synergistic antiproliferative action with ligands of the peripheral benzodiazepine receptor. J Hepatol 43, 808–816 (2005). [DOI] [PubMed] [Google Scholar]
- 70.Kah J, et al. Selective induction of apoptosis by HMG-CoA reductase inhibitors in hepatoma cells and dependence on p53 expression. Oncol Rep 28, 1077–1083 (2012). [DOI] [PubMed] [Google Scholar]
- 71.Wang W, et al. Simvastatin ameliorates liver fibrosis via mediating nitric oxide synthase in rats with non-alcoholic steatohepatitis-related liver fibrosis. PLoS One 8, e76538 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chong LW, et al. Fluvastatin attenuates hepatic steatosis-induced fibrogenesis in rats through inhibiting paracrine effect of hepatocyte on hepatic stellate cells. BMC Gastroenterol 15, 22 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Marrone G, et al. The transcription factor KLF2 mediates hepatic endothelial protection and paracrine endothelial-stellate cell deactivation induced by statins. J Hepatol 58, 98–103 (2013). [DOI] [PubMed] [Google Scholar]
- 74.Marinho TS, et al. Rosuvastatin limits the activation of hepatic stellate cells in diet-induced obese mice. Hepatol Res 47, 928–940 (2017). [DOI] [PubMed] [Google Scholar]
- 75.Uschner FE, et al. Statins activate the canonical hedgehog-signaling and aggravate non-cirrhotic portal hypertension, but inhibit the non-canonical hedgehog signaling and cirrhotic portal hypertension. Sci Rep 5, 14573 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kim G, et al. Effect of statin on hepatocellular carcinoma in patients with type 2 diabetes: A nationwide nested case-control study. Int J Cancer 140, 798–806 (2017). [DOI] [PubMed] [Google Scholar]
- 77.Chen HH, et al. Combination Therapy of Metformin and Statin May Decrease Hepatocellular Carcinoma Among Diabetic Patients in Asia. Medicine 94, e1013 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tsan YT, Lee CH, Wang JD & Chen PC Statins and the risk of hepatocellular carcinoma in patients with hepatitis B virus infection. J Clin Oncol 30, 623–630 (2012). [DOI] [PubMed] [Google Scholar]
- 79.Tsan YT, et al. Statins and the risk of hepatocellular carcinoma in patients with hepatitis C virus infection. J Clin Oncol 31, 1514–1521 (2013). [DOI] [PubMed] [Google Scholar]
- 80.Simon TG, Bonilla H, Yan P, Chung RT & Butt AA Atorvastatin and fluvastatin are associated with dose-dependent reductions in cirrhosis and hepatocellular carcinoma, among patients with hepatitis C virus: Results from ERCHIVES. Hepatology 64, 47–57 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Simon TG, King LY, Zheng H & Chung RT Statin use is associated with a reduced risk of fibrosis progression in chronic hepatitis C. J Hepatol 62, 18–23 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chang FM, et al. Statins decrease the risk of decompensation in hepatitis B virus- and hepatitis C virus-related cirrhosis: A population-based study. Hepatology 66, 896–907 (2017). [DOI] [PubMed] [Google Scholar]
- 83.Lee TY, et al. The occurrence of hepatocellular carcinoma in different risk stratifications of clinically noncirrhotic nonalcoholic fatty liver disease. Int J Cancer 141, 1307–1314 (2017). [DOI] [PubMed] [Google Scholar]
- 84.Singh S, Singh PP, Singh AG, Murad MH & Sanchez W Statins are associated with a reduced risk of hepatocellular cancer: a systematic review and meta-analysis. Gastroenterology 144, 323–332 (2013). [DOI] [PubMed] [Google Scholar]
- 85.Cholesterol Treatment Trialists, C., et al. Lack of effect of lowering LDL cholesterol on cancer: meta-analysis of individual data from 175,000 people in 27 randomised trials of statin therapy. PLoS One 7, e29849 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhou YY, et al. Systematic review with network meta-analysis: statins and risk of hepatocellular carcinoma. Oncotarget 7, 21753–21762 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Kennedy OJ, et al. Coffee, including caffeinated and decaffeinated coffee, and the risk of hepatocellular carcinoma: a systematic review and dose-response meta-analysis. BMJ Open 7, e013739 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Aleksandrova K, et al. The association of coffee intake with liver cancer risk is mediated by biomarkers of inflammation and hepatocellular injury: data from the European Prospective Investigation into Cancer and Nutrition. Am J Clin Nutr 102, 1498–1508 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xiao Q, Sinha R, Graubard BI & Freedman ND Inverse associations of total and decaffeinated coffee with liver enzyme levels in National Health and Nutrition Examination Survey 1999–2010. Hepatology 60, 2091–2098 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Setiawan VW, et al. Coffee Drinking and Alcoholic and Nonalcoholic Fatty Liver Diseases and Viral Hepatitis in the Multiethnic Cohort. Clin Gastroenterol Hepatol 15, 1305–1307 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Bamia C, et al. Coffee, tea and decaffeinated coffee in relation to hepatocellular carcinoma in a European population: multicentre, prospective cohort study. Int J Cancer 136, 1899–1908 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Freedman ND, et al. Coffee consumption is associated with response to peginterferon and ribavirin therapy in patients with chronic hepatitis C. Gastroenterology 140, 1961–1969 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Edling CE, Selvaggi F, Ghonaim R, Maffucci T & Falasca M Caffeine and the analog CGS 15943 inhibit cancer cell growth by targeting the phosphoinositide 3-kinase/Akt pathway. Cancer Biol Ther 15, 524–532 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hoshida Y, Fuchs BC & Tanabe KK Prevention of hepatocellular carcinoma: potential targets, experimental models, and clinical challenges. Curr Cancer Drug Targets 12, 1129–1159 (2012). [PMC free article] [PubMed] [Google Scholar]
- 95.Veldt BJ, et al. Long-term clinical outcome and effect of glycyrrhizin in 1093 chronic hepatitis C patients with non-response or relapse to interferon. Scand J Gastroenterol 41, 1087–1094 (2006). [DOI] [PubMed] [Google Scholar]
- 96.Morgan TR, et al. A Phase II Randomized, Controlled Trial of S-Adenosylmethionine in Reducing Serum alpha-Fetoprotein in Patients with Hepatitis C Cirrhosis and Elevated AFP. Cancer Prev Res (Phila) 8, 864–872 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Duarte-Salles T, et al. Dietary fat, fat subtypes and hepatocellular carcinoma in a large European cohort. Int J Cancer 137, 2715–2728 (2015). [DOI] [PubMed] [Google Scholar]
- 98.Sawada N, et al. Consumption of n-3 fatty acids and fish reduces risk of hepatocellular carcinoma. Gastroenterology 142, 1468–1475 (2012). [DOI] [PubMed] [Google Scholar]
- 99.Lim K, Han C, Dai Y, Shen M & Wu T Omega-3 polyunsaturated fatty acids inhibit hepatocellular carcinoma cell growth through blocking beta-catenin and cyclooxygenase-2. Molecular cancer therapeutics 8, 3046–3055 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Weylandt KH, et al. Suppressed liver tumorigenesis in fat-1 mice with elevated omega-3 fatty acids is associated with increased omega-3 derived lipid mediators and reduced TNF-alpha. Carcinogenesis 32, 897–903 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Freedman ND, et al. Association of meat and fat intake with liver disease and hepatocellular carcinoma in the NIH-AARP cohort. J Natl Cancer Inst 102, 1354–1365 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wong GL, et al. Adverse effects of vitamin D deficiency on outcomes of patients with chronic hepatitis B. Clin Gastroenterol Hepatol 13, 783–790e781 (2015). [DOI] [PubMed] [Google Scholar]
- 103.Finkelmeier F, et al. Severe 25-hydroxyvitamin D deficiency identifies a poor prognosis in patients with hepatocellular carcinoma - a prospective cohort study. Aliment Pharmacol Ther 39, 1204–1212 (2014). [DOI] [PubMed] [Google Scholar]
- 104.Fedirko V, et al. Prediagnostic circulating vitamin D levels and risk of hepatocellular carcinoma in European populations: a nested case-control study. Hepatology 60, 1222–1230 (2014). [DOI] [PubMed] [Google Scholar]
- 105.Trepo E, et al. Marked 25-hydroxyvitamin D deficiency is associated with poor prognosis in patients with alcoholic liver disease. J Hepatol 59, 344–350 (2013). [DOI] [PubMed] [Google Scholar]
- 106.Kwon HJ, et al. Vitamin D3 upregulated protein 1 suppresses TNF-alpha-induced NF-kappaB activation in hepatocarcinogenesis. J Immunol 185, 3980–3989 (2010). [DOI] [PubMed] [Google Scholar]
- 107.Nakano T, et al. Impact of artificial sunlight therapy on the progress of non-alcoholic fatty liver disease in rats. J Hepatol 55, 415–425 (2011). [DOI] [PubMed] [Google Scholar]
- 108.Abramovitch S, et al. Vitamin D inhibits development of liver fibrosis in an animal model but cannot ameliorate established cirrhosis. Am J Physiol Gastrointest Liver Physiol 308, G112–120 (2015). [DOI] [PubMed] [Google Scholar]
- 109.Duran A, et al. p62/SQSTM1 by Binding to Vitamin D Receptor Inhibits Hepatic Stellate Cell Activity, Fibrosis, and Liver Cancer. Cancer Cell 30, 595–609 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kowdley KV Iron, hemochromatosis, and hepatocellular carcinoma. Gastroenterology 127, S79–86 (2004). [DOI] [PubMed] [Google Scholar]
- 111.Harrison SA & Bacon BR Relation of hemochromatosis with hepatocellular carcinoma: epidemiology, natural history, pathophysiology, screening, treatment, and prevention. The Medical clinics of North America 89, 391–409 (2005). [DOI] [PubMed] [Google Scholar]
- 112.Nahon P, et al. Liver iron, HFE gene mutations, and hepatocellular carcinoma occurrence in patients with cirrhosis. Gastroenterology 134, 102–110 (2008). [DOI] [PubMed] [Google Scholar]
- 113.Fan JG, Farrell GC & Asia-Pacific Working Party for Prevention of Hepatocellular, C. Prevention of hepatocellular carcinoma in nonviral-related liver diseases. J Gastroenterol Hepatol 24, 712–719 (2009). [DOI] [PubMed] [Google Scholar]
- 114.Kato J, et al. Long-term phlebotomy with low-iron diet therapy lowers risk of development of hepatocellular carcinoma from chronic hepatitis C. J Gastroenterol 42, 830–836 (2007). [DOI] [PubMed] [Google Scholar]
- 115.Preziosi ME, et al. Mice lacking liver-specific beta-catenin develop steatohepatitis and fibrosis after iron overload. J Hepatol 67, 360–369 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Saeki I, et al. Effects of an oral iron chelator, deferasirox, on advanced hepatocellular carcinoma. World J Gastroenterol 22, 8967–8977 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Gluud LL, et al. Branched-chain amino acids for people with hepatic encephalopathy. Cochrane Database Syst Rev 5, CD001939 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Cha JH, et al. Branched-chain amino acids ameliorate fibrosis and suppress tumor growth in a rat model of hepatocellular carcinoma with liver cirrhosis. PLoS One 8, e77899 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Nakano M, et al. Branched-chain amino acids enhance premature senescence through mammalian target of rapamycin complex I-mediated upregulation of p21 protein. PLoS One 8, e80411 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Korenaga M, et al. Branched-chain amino acids reduce hepatic iron accumulation and oxidative stress in hepatitis C virus polyprotein-expressing mice. Liver Int 35, 1303–1314 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Takegoshi K, et al. Branched-chain amino acids prevent hepatic fibrosis and development of hepatocellular carcinoma in a non-alcoholic steatohepatitis mouse model. Oncotarget 8, 18191–18205 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Terakura D, et al. Preventive effects of branched-chain amino acid supplementation on the spontaneous development of hepatic preneoplastic lesions in C57BL/KsJ-db/db obese mice. Carcinogenesis 33, 2499–2506 (2012). [DOI] [PubMed] [Google Scholar]
- 123.Kawaguchi T, et al. Branched-chain amino acids prevent hepatocarcinogenesis and prolong survival of patients with cirrhosis. Clin Gastroenterol Hepatol 12, 1012–1018e1011 (2014). [DOI] [PubMed] [Google Scholar]
- 124.Nakagawa S, et al. Molecular Liver Cancer Prevention in Cirrhosis by Organ Transcriptome Analysis and Lysophosphatidic Acid Pathway Inhibition. Cancer Cell 30, 879–890 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Duvoux C & Toso C mTOR inhibitor therapy: Does it prevent HCC recurrence after liver transplantation? Transplant Rev (Orlando) 29, 168–174 (2015). [DOI] [PubMed] [Google Scholar]
- 126.Massoud O & Wiesner RH The use of sirolimus should be restricted in liver transplantation. J Hepatol 56, 288–290 (2012). [DOI] [PubMed] [Google Scholar]
- 127.Umemura A, et al. Liver damage, inflammation, and enhanced tumorigenesis after persistent mTORC1 inhibition. Cell Metab 20, 133–144 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Burra P & Rodriguez-Castro KI Neoplastic disease after liver transplantation: Focus on de novo neoplasms. World J Gastroenterol 21, 8753–8768 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Geissler EK, et al. Sirolimus Use in Liver Transplant Recipients With Hepatocellular Carcinoma: A Randomized, Multicenter, Open-Label Phase 3 Trial. Transplantation 100, 116–125 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Binder BY, Williams PA, Silva EA & Leach JK Lysophosphatidic Acid and Sphingosine-1-Phosphate: A Concise Review of Biological Function and Applications for Tissue Engineering. Tissue Eng Part B Rev 21, 531–542 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Erstad DJ, Tager AM, Hoshida Y & Fuchs BC The autotaxin-lysophosphatidic acid pathway emerges as a therapeutic target to prevent liver cancer. Mol Cell Oncol 4, e1311827 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Neuschwander-Tetri BA Non-alcoholic fatty liver disease. BMC Med 15, 45 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Kaffe E, et al. Hepatocyte autotaxin expression promotes liver fibrosis and cancer. Hepatology 65, 1369–1383 (2017). [DOI] [PubMed] [Google Scholar]
- 134.Lanaya H, et al. EGFR has a tumour-promoting role in liver macrophages during hepatocellular carcinoma formation. Nature cell biology 16, 972–977 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Komposch K & Sibilia M EGFR Signaling in Liver Diseases. Int J Mol Sci 17(2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Fuchs BC, et al. Epidermal growth factor receptor inhibition attenuates liver fibrosis and development of hepatocellular carcinoma. Hepatology 59, 1577–1590 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Bruix J, et al. Adjuvant sorafenib for hepatocellular carcinoma after resection or ablation (STORM): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Oncol 16, 1344–1354 (2015). [DOI] [PubMed] [Google Scholar]
- 138.Oakley F, et al. Angiotensin II activates I kappaB kinase phosphorylation of RelA at Ser 536 to promote myofibroblast survival and liver fibrosis. Gastroenterology 136, 2334–2344e2331 (2009). [DOI] [PubMed] [Google Scholar]
- 139.Moreno M, et al. Reduction of advanced liver fibrosis by short-term targeted delivery of an angiotensin receptor blocker to hepatic stellate cells in rats. Hepatology 51, 942–952 (2010). [DOI] [PubMed] [Google Scholar]
- 140.Tamaki Y, et al. Angiotensin II type 1 receptor antagonist prevents hepatic carcinoma in rats with nonalcoholic steatohepatitis. J Gastroenterol 48, 491–503 (2013). [DOI] [PubMed] [Google Scholar]
- 141.Facciorusso A, et al. Angiotensin receptor blockers improve survival outcomes after radiofrequency ablation in hepatocarcinoma patients. J Gastroenterol Hepatol 30, 1643–1650 (2015). [DOI] [PubMed] [Google Scholar]
- 142.Yoshiji H, et al. Combination of vitamin K2 and angiotensin-converting enzyme inhibitor ameliorates cumulative recurrence of hepatocellular carcinoma. J Hepatol 51, 315–321 (2009). [DOI] [PubMed] [Google Scholar]
- 143.Yoshiji H, et al. Combination of branched-chain amino acids and angiotensin-converting enzyme inhibitor suppresses the cumulative recurrence of hepatocellular carcinoma: a randomized control trial. Oncol Rep 26, 1547–1553 (2011). [DOI] [PubMed] [Google Scholar]
- 144.Tan CK Peretinoin as an adjuvant therapy for hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 10, 1201–1210 (2016). [DOI] [PubMed] [Google Scholar]
- 145.Okada H, et al. Acyclic retinoid targets platelet-derived growth factor signaling in the prevention of hepatic fibrosis and hepatocellular carcinoma development. Cancer Res 72, 4459–4471 (2012). [DOI] [PubMed] [Google Scholar]
- 146.Guan HB, et al. Acyclic retinoid induces differentiation and apoptosis of murine hepatic stem cells. Stem Cell Res Ther 6, 51 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Shimakami T, et al. The acyclic retinoid Peretinoin inhibits hepatitis C virus replication and infectious virus release in vitro. Sci Rep 4, 4688 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Okada H, et al. Peretinoin, an acyclic retinoid, suppresses steatohepatitis and tumorigenesis by activating autophagy in mice fed an atherogenic high-fat diet. Oncotarget 8, 39978–39993 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Okita K, et al. Peretinoin after curative therapy of hepatitis C-related hepatocellular carcinoma: a randomized double-blind placebo-controlled study. J Gastroenterol 50, 191–202 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Okita K, et al. Survey of survival among patients with hepatitis C virus-related hepatocellular carcinoma treated with peretinoin, an acyclic retinoid, after the completion of a randomized, placebo-controlled trial. J Gastroenterol 50, 667–674 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Zheng B, Zhu YJ, Wang HY & Chen L Gender disparity in hepatocellular carcinoma (HCC): multiple underlying mechanisms. Sci China Life Sci 60, 575–584 (2017). [DOI] [PubMed] [Google Scholar]
- 152.Sun H, et al. Association between estrogen receptor 1 (ESR1) genetic variations and cancer risk: a meta-analysis. J BUON 20, 296–308 (2015). [PubMed] [Google Scholar]
- 153.Hassan MM, et al. Estrogen Replacement Reduces Risk and Increases Survival Times of Women With Hepatocellular Carcinoma. Clin Gastroenterol Hepatol 15, 1791–1799 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Zheng F, et al. Systemic delivery of microRNA-101 potently inhibits hepatocellular carcinoma in vivo by repressing multiple targets. PLoS genetics 11, e1004873 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kim RS, Goossens N & Hoshida Y Use of big data in drug development for precision medicine. Expert Rev Precis Med Drug Dev 1, 245–253 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]