Review Open Access
Copyright ©The Author(s) 2018. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Oct 7, 2018; 24(37): 4230-4242
Published online Oct 7, 2018. doi: 10.3748/wjg.v24.i37.4230
New prognostic biomarkers of mortality in patients undergoing liver transplantation for hepatocellular carcinoma
Leonardo Lorente, Intensive Care Unit, Hospital Universitario de Canarias, Santa Cruz de Tenerife 38320, Spain
ORCID number: Leonardo Lorente (0000-0003-4902-4065).
Author contributions: Lorente L drafted the manuscript.
Conflict-of-interest statement: The author has no financial or other conflicts of interest to declare related to the submitted manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Leonardo Lorente, MD, PhD, Attending Doctor, Medical Assistant, Intensive Care Unit, Hospital Universitario de Canarias, Ofra, s/n, Santa Cruz de Tenerife 38320, Spain. lorentemartin@msn.com
Telephone: +34-922-678000 Fax: +34-922-647112
Received: May 25, 2018
Peer-review started: May 25, 2018
First decision: July 4, 2018
Revised: August 18, 2018
Accepted: August 24, 2018
Article in press: August 24, 2018
Published online: October 7, 2018

Abstract

The outcome prediction of hepatocellular carcinoma (HCC) patients undergoing liver transplantation (LT) was classically established using various macromorphological factors and serum alpha-fetoprotein levels prior to LT. However, other biomarkers have recently been reported to be associated with the prognosis of HCC patients undergoing to LT. This review summarizes clinical data on these new biomarkers. High blood levels of malondialdehyde, total antioxidant capacity, caspase-cleaved cytokeratin-18, soluble CD40 ligand, substance P, C-reactive protein, and vascular endothelial growth factor, increased neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in blood, high peripheral blood expression of human telomerase reverse transcriptase messenger ribonucleic acid, and high HCC expression of dickkopf-1 have recently been associated with decreased survival rates. In addition, high blood levels of des-gamma-carboxy prothrombin, and high HCC expression of glypican-3, E-cadherin and beta-catenin have been associated with increased HCC recurrence. Additional research is necessary to establish the prognostic role of these biomarkers in HCC prior to LT. Furthermore, some of these biomarkers are also interesting because their potential modulation could help to create new research lines for improving the outcomes of those patients.

Key Words: Liver transplantation, Hepatocellular carcinoma, Biomarkers, Outcome, Survival, Recurrence, Genomic

Core tip: The outcome of liver transplantation (LT) for hepatocellular carcinoma (HCC) patients are generally predicted using various macromorphological factors and serum alpha-fetoprotein levels prior to LT. However, other biomarkers have recently been reported to be associated with the prognosis of HCC patients undergoing LT. Furthermore, some of these biomarkers are also interesting because their potential modulation could help to create new research lines for improving the outcomes of those patients. This review summarizes clinical data on those new biomarkers.



INTRODUCTION

Hepatocellular carcinoma (HCC), the most frequent primary liver malignancy, is one of the most common malignancies and causes of cancer-related deaths[1-3]. In liver transplantation (LT), the primary tumor is removed, and liver failure is treated. Therefore, LT is considered the treatment of choice for some HCC patients[1-11].

Various macromorphological factors assessed prior to LT have been classically used to predict the outcome of HCC patients undergoing LT. These factors include the tumor size, tumor number, degree of differentiation, hepatic microvascular invasion, hepatic macrovascular invasion, being outside the Milan criteria and infiltration[1-11].

However, establishing biomarkers to be assessed prior to LT could strengthen the predictions of prognoses for HCC patients undergoing LT. Currently, the most commonly studied biomarker are the serum alphafetoprotein levels[1-11]. However, other biomarkers have recently been reported to be associated with the prognosis of HCC patients undergoing to LT. This review summarizes clinical data on these new biomarkers.

BIOMARKERS
Malondialdehyde

Oxidative stress can lead to membrane lipid peroxidation, which creates many end products, including malondialdehyde, which is a low molecular weight aldehyde that is produced during the degradation of cellular membrane phospholipids. It is formed when free radicals attack polyunsaturated fatty acids. Malondialdehyde can be released into the extracellular space, and it can ultimately reach the bloodstream. Therefore, it has been used as a circulating biomarker of lipid oxidation[12,13].

Some studies have reported higher levels of serum malondialdehyde in HCC patients[14-16] and patients with chronic liver disease than in healthy controls[17,18]. Additionally, studies have reported that the tumoral tissue of HCC patients has higher malondialdehyde concentrations than non-tumoral tissue[19]. Studies have also found higher free radical intensity in the erythrocytes of HCC patients than in healthy controls[20], higher serum concentrations of reactive oxygen metabolites in HCC patients who exhibit recurrence after curative treatment by radiofrequency ablation or surgical resection[21], and higher circulating lipid peroxide levels prior to LT in patients who do not survive LT than in survivors[22].

A study by our team reported, for the first time, that serum malondialdehyde levels prior to LT were higher in non-surviving patients than in patients who survived for one year after LT. We also found an association between serum malondialdehyde levels in HCC patients prior to LT and their survival at one year after LT[23]. These findings are consistent with those from other studies that have reported an association between circulating malondialdehyde levels and mortality in patients with sepsis[24], traumatic brain injuries[25], brain infarctions[26] and spontaneous intracerebral hemorrhaging[27].

Total antioxidant capacity

The production of reactive oxygen species (ROS) is balanced by the production of antioxidant defenses, and the analysis of total antioxidant capacity (TAC) could provide a global information in respect to the antioxidant status[28].

Some studies have found lower circulating TAC levels in LT patients than in healthy controls[18], and lower circulating TAC levels in HCC patients than in healthy controls[14,15].

A study by our team was the first to find that serum TAC levels prior to LT were lower in non-surviving than in surviving patients during the first one year after LT[29]. Besides, we found that there is an association between low serum TAC levels in HCC patients prior to LT and their survival at one year after LT. In addition, we found a negative association between serum levels of TAC and malondialdehyde; thus, patients with lower serum TAC levels showed higher lipid peroxidation.

I think that those findings could suggest that it is possible that non-survivor LT patients remains during the first one year after LT with low serum TAC levels and high serum malondialdehyde levels (due to a higher lipid peroxidation because the high ROS production is not balanced by an insufficient antioxidant capacity) compared to survivor patients.

Antioxidant agents have been shown to reduce malondialdehyde concentrations in animal models of sepsis[30] and brain trauma[31] as well as in clinical trials involving septic newborns[32], acute ischemic stroke[33] and traumatic brain injuries[34]. Additionally, in a clinical trial of traumatic brain injuries[34], the administration of antioxidant agents reduced mortality rate. Thus, since non-surviving HCC patients showed higher serum malondialdehyde levels prior to LT than surviving patients, it could be interesting to explore the benefit of the administration of antioxidant agents to HCC patients undergoing LT. Antioxidant treatment could potentially improve their prognoses, especially for patients with a higher oxidative state.

Caspase-cleaved cytokeratin-18

Apoptosis, which leads to active and programmed cell elimination, is increased in liver diseases[35-37]. Two main pathways exist (extrinsic and intrinsic) for cell death by apoptosis. The apoptotic extrinsic pathway is initiated when the tumor necrosis factor receptor superfamily (TNFRSF) is activated by its ligand (TNFSF). This leads to the formation of a death signal that activates caspase-8 and ultimately activates caspase-3. The intrinsic apoptotic pathway is activated via oxygen free radicals, interleukin (IL)-1, IL-6 and nitric oxide. These factors release cytochromes from the mitochondria to the cytosol, which activates caspase-3. Thus, both apoptotic pathways ultimately activate caspase 3, which leads to cell death.

Cytokeratin-18 is the main protein found in the intermediate filaments of the liver and is present in most parenchymal and epithelial cells. During hepatocyte apoptosis, cytokeratin-18 is cleaved by caspases and can be released into the bloodstream as caspase-cleaved cytokeratin (CCCK)-18[35-39], which can be detected using M30 monoclonal antibodies[40,41].

Some studies have reported higher circulating CCCK-18 levels in patients with tumoral diseases than in healthy controls[42,43] and in patients with tumoral diseases that had a poor evolution[44-48]. Additionally, HCC patients have higher circulating CCCK-18 levels than healthy controls[49,50] or cirrhotic patients[51,52]. Studies have reported an association between serum CCCK-18 levels and mortality in HCC patients[53].

A study by our team found, for the first time, that serum CCCK-18 levels prior to LT were higher in non-surviving patients than in patients who survived for one year after LT. Additionally, an association was found between serum CCCK-18 levels in HCC patients prior to LT and their survival for one year after LT[54]. These findings are consistent with the results of other studies that have shown that circulating CCCK-18 levels are associated with the prognosis of patients with various tumoral diseases[44-48], HCC[53], sepsis[55], traumatic brain injury[56] and cerebral artery infarction[57]. Additionally, circulating CCCK-18 levels have been associated with metastasis[45], serum AFP levels[46,54] and tumor size[47,48].

Soluble CD40 ligand

Soluble CD40 ligand (sCD40L) is a member in the TNFSF of proteins. It has proinflammatory and prothrombotic effects when bound to its receptor, CD40, which is also a member of the TNFRSF[58-65]. CD40L is mainly found in platelets and activated T-lymphocytes, although it is also present in smooth muscle cells, endothelial cells, microglia, monocytes, and B cells. When CD40L is cleaved, it is released into circulation and is present in its soluble form, sCD40L[58-65].

Some studies have reported higher circulating sCD40L levels in patients with ischemic stroke[66-69], acute coronary syndrome[70,71], and sepsis[72,73] than in healthy subjects. Additionally, high circulating sCD40L levels are associated with a poor prognosis in patients with ischemic stroke[69], acute coronary syndrome[74], sepsis[72,73] and traumatic brain injuries[75]. Patients with chronic hepatitis C virus infection[76], cirrhosis[77], and non-alcoholic fatty liver disease have been shown to exhibit higher circulating sCD40L levels than control subjects[78]. Furthermore, high circulating sCD40L levels are associated with a poor prognosis in HCC patients[79].

A study by our team was the first to report that serum sCD40L levels prior to LT were higher in patients who did not survive for one year after transplantation than in the surviving patients, and an association was also found between serum sCD4L levels in HCC patients prior to LT and survival for one year after LT[80]. These findings are consistent with the results of other studies reporting an association between circulating sCD40L levels and mortality in patients with cerebral infarction[69], acute coronary syndrome[74], sepsis[72,73] and traumatic brain injuries[75].

Circulating sCD40L levels could play a role in patients receiving LT for HCC by their proinflammatory[81,82] and procoagulant[83-88] effects. The proinflammatory effects of sCD40L may be due to an increase in the expression of proinflammatory mediators such as IL-1, IL-6, IL-12, TNF-alpha and interferon-gamma[81,82]. The procoagulant effects of sCD40L may be due to the induction of tissue factor expression[83-86], reduced expression of thrombomodulin[85,86] and its binding to glycoprotein IIb/IIIa platelet receptor[87,88]. These proinflammatory and procoagulant effects could potentially favor the development of vascular thrombosis and organ dysfunction, ultimately resulting in patient death.

The statin administration has been associated with a reduction of circulating sCD40L levels in patients with coronary artery disease[89-91] and an improvement in the prognosis of patients with ischemic stroke[92] and infections[93-96]. Therefore, as non-surviving HCC patients showed higher serum sCD40L levels prior to LT than patients who survived for one year after LT, it could be interesting to explore the benefit of administering sCD40L modulators to HCC patients who are undergoing to LT to improve their prognosis, especially for patients with higher sCD40L levels.

Substance P

Substance P is a member of the tachykinin family, which is distributed by the central and peripheral nervous, respiratory and urinary systems and by the gut. Tachykinins may play a role in nociceptive responses, inflammation, vasodilation and plasma protein extravasation[97-99].

Circulating substance P levels are elevated in patients with liver diseases compared to control subjects[100-106] and in patients with severe liver diseases[104-106].

A study by our team was the first to report that serum levels of substance P prior to LT were higher in patients who did not survive for one year after LT than in surviving patients. The study also found an association between serum levels of substance P in HCC patients prior to LT and mortality within one year after LT[107]. These findings are consistent with the results of other studies that have reported an association between circulating serum P levels and mortality in patients with traumatic brain injuries[108] or ischemic stroke[109].

Substance P plays a role in the inflammatory response by producing inflammatory cytokines such as IL-1, IL-6 and TNF-alpha[110-114]. Various agents that reduce substance P activity have been identified in animal models of ischemic stroke[115-117] and traumatic brain injury[118,119]. These agents have been associated with a reduction in the inflammation process and edema. HCC patients who did not survive for one year after LT showed higher serum substance P levels prior to LT than surviving patients. Therefore, it could be interesting to explore the benefit of administering agents to control substance P activity to HCC patients undergoing LT, especially in patients with high circulating substance P levels.

Neutrophil to lymphocyte ratio and platelet to lymphocyte ratio

The blood neutrophil to lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR) have both been used as biomarkers to evaluate systemic inflammatory responses. A meta-analysis published in 2017 by Zheng et al[120] analyzed the association between the NLR and PLR in the blood of HCC patients prior to receiving different treatments and its overall survival and HCC recurrence. The treatments included curative resection, transarterial chemoembolization (TACE), radiofrequency ablation (RFA), LT and chemotherapy. The authors examined the associations between the NLR and all treatments combined and individually. They found an association between a high NLR and both of the outcomes (poor overall survival and HCC recurrence) for all types of treatment. They also reported an association between a high NLR and survival when specifically analyzing LT. However, no association was found between the NLR and HCC recurrence when specifically analyzing LT. The authors also found an association between a high PLR and poor outcomes for all treatments combined and when analyzing LT specifically.

C-reactive protein

C-reactive protein (CRP) is synthesized in the liver by hepatocytes in response to factors released by macrophages and adipocytes during inflammation and afterwards is released to blood; thus, blood levels of CRP increase in response to inflammation. Elevated blood CRP levels are associated in multivariate analyses with an increase in the risk of HCC recurrence and decreased survival in patients undergoing LT for HCC[121,122], overall in patients with HCC beyond the Milan criteria[121].

Des-gamma-carboxy prothrombin or protein induced by vitamin K absence or antagonist II (PIVKA-II)

Des-gamma-carboxy prothrombin (DCP) is a nonfunctional prothrombin form produced by the liver. The normal liver converts the glutamic acid residues in the N-terminal portion of prothrombin by carboxylation in gamma-carboxyglutamic acid residues before its release into the peripheral blood. In many of HCC cells, the vitamin K dependent carboxylase that produces this carboxylation is absent; thus, an abnormal prothrombin is secreted. Several studies have found in multivariate analyses that high blood DCP levels are associated with a higher risk of HCC recurrence in HCC patients who undergo LT[123-127].

Glypican-3

Glypican (GPC)-3 is a member of the glypican protein family, which plays a role in regulating cell division and growth. One study reported that the protein expression of GPC-3 in HCC tissue samples prior to LT was associated with a higher rate of HCC recurrence after LT[128]; in addition, there was found that GPC-3 was expressed in 68% of HCC tissues, but not in adjacent non-tumoral tissues and in tissues of liver controls. Another study found that serum GPC-3 levels were higher in HCC patients than in cirrhosis patients[129]; however, the study did not examine the prognostic role of serum GPC-3 in HCC patients prior to LT.

Human telomerase reverse transcriptase messenger ribonucleic acid

Human telomerase reverse transcriptase messenger ribonucleic acid (h-TERT mRNA) is a ribonucleoprotein polymerase that maintains telomere ends in chromosomes. High h-TERT mRNA expression in the peripheral blood of HCC patients who undergo LT has been associated with decreased survival and increased HCC recurrence[130,131]; however, in another study, h-TERT mRNA concentrations in the peripheral blood were not associated with HCC recurrence after LT[132]. Therefore, additional research is necessary to determine the prognostic role of h-TERT mRNA expression in the peripheral blood of HCC patients prior to LT.

Matrix metalloproteinase-9

Matrix metalloproteinase (MMP)-9 is a member of the matrix metalloproteinases (MMPs), which are involved in degradation and remodeling of the extracellular matrix. MMPs play a role in physiological functions such as morphogenesis, tissue remodeling and the menstrual cycle. They are also involved in various diseases such as arthritis, tumors, atherosclerosis and sepsis. The activity of MMPs is regulated by several tissue inhibitor of matrix metalloproteinases (TIMPs).

Contradictory results have been found with regard to MMP-9. Patients who undergo LT for HCC and have high MMP-9 expression in the tumor have exhibited an unfavorable rates of overall survival and HCC recurrence[133,134]. Another study in patients undergoing LT due to HCC or for cirrhosis without HCC found that high serum MMP-9 levels at one week after LT were associated with a higher rate of LT rejection[135]. However, one study also reported that high serum MMP-9 levels and low serum TIMP-1 levels in HCC patients receiving different treatments (curative resection, TACE, thermoablation, and LT) were associated with a higher survival rate, although the study did not specifically examine patients receiving LT because the sample size for that group was small[136]. Our group has previously reported a lower survival rate in patients with cerebral artery infarction[137], traumatic brain injury[138] and sepsis[139,140] who have high serum TIMP-1 levels than in patients who have low TIMP-1 levels. Therefore, additional research is necessary to establish the prognostic role of MMP-9 expression in the peripheral blood of HCC patients prior to LT.

E-cadherin

E-cadherin is a member of the cadherin family of proteins, which are cell adhesion molecules that participate in the formation of junctions between cells. One study found that high serum levels of soluble E-cadherin were associated with increased recurrence of HCC after a curative resection[141]. Another study of HCC patients who underwent LT found that E-cadherin expression in the liver was associated with HCC recurrence after LT[142].

Beta-catenin

Beta-catenin is a member of the catenin family of proteins, which also constitute a group of cell adhesion molecules that are involved in the formation of bonds between cells. A study of HCC patients who underwent LT found that beta-catenin expression in the liver was associated with HCC recurrence after LT[142]. However, in other recently published study, no association was found between beta-catenin expression in the liver prior to LT and the survival of HCC patients[143]. Therefore, additional research is necessary to establish the prognostic role of beta-catenin expression in the liver in HCC patients prior to LT.

AFP

AFP is a glycoprotein that is produced by the yolk sac and the fetal liver during fetal development. It is the most abundant plasma protein in the human fetus. Increased values are found in newborns (values gradually decrease to normal over the first year of life), pregnant women (values return to normal after delivery), and patients with various AFP-producing tumors such as HCC and tumors of the ovary and testis.

The blood AFP level is the most extensively studied biomarker in HCC patients undergoing LT. Elevated blood AFP levels are associated with decreased survival[144] as well as an increase in HCC recurrence[145] in patients undergoing LT for HCC.

A review of 13 observational studies published in 2012 involving 12,159 patients who underwent LT for HCC examined the role of pre-LT circulating AFP levels in predicting survival and HCC recurrence[144]. Nine of the 13 studies reported data about pre-LT serum AFP levels and survival. Only four studies reported absolute serum AFP values for all included patients, and the other studies used varying cut-off points for serum AFP levels. This heterogeneity precluded pooling of the data for a valid meta-analysis. The majority of the studies concluded that a high pre-LT serum AFP level was an independent predictor of death following LT for HCC. These studies also suggested that serum AFP levels higher than 1000 ng/mL may predict poorer survival. Ten of the 13 studies reported data on HCC recurrence and pre-LT serum AFP values. All of these studies concluded that high AFP levels were associated with increased HCC recurrence following LT for HCC. The authors were unable to perform a meta-analysis on this research question due to the heterogeneity in the data reported by the studies. Additionally, some of the studies included in the review found that pre-LT serum AFP levels were correlated with vascular invasion and poor differentiation of HCC.

A review and meta-analysis published in 2016 examined the prognostic role of biomarkers in HCC recurrence in patients who underwent LT for HCC[145]. The review included 49 studies with a total of 13693 patients that reported data on pre-LT serum AFP levels and HCC recurrence. However, the studies had 88% heterogeneity due to their use of varying definitions and cut-off values for AFP. Therefore, it was not possible to conduct a valid meta-analysis for this topic. However, a meta-analysis was performed using 17 of the studies with different cut-off values for pre-LT serum AFP levels, but the meta-analysis required a cut-off value higher than 400 ng/mL. In this analysis, an association was found between elevated pre-LT serum AFP levels and the risk of HCC recurrence (HR = 2.69; 95%CI: 2.08-3.47), with a heterogeneity of 46%.

Dickkopf-1

In several studies have been found higher circulating Dickkopf-1 (DKK1) levels in HCC patients than in healthy subjects[146-149] or than in patients with liver cirrosis without HCC[150,151]. In addition, in a meta-analysis published in 2014 including 4 studies[152] and in other recently published study[153] was found that higher DKK1 expression levels in HCC patients were associated with lower survival. Besides, in one study was found that higher DKK1 expression is associated with lower survival and higher recurrence in HCC patients after LT[154].

Vascular endothelial growth factor

In a meta-analysis of 11 studies was found that high serum Vascular endothelial growth factor (VEGF) leves in HCC patients were associated with lower survival[155]. In addition, in one study was found that high plasma VEGF levels in HCC patients previously to LT were associated with HCC recurrence and survival[156].

Caspase-1

Pyroptosis is a form of programmed cell death, which is dependent of caspase-1. In some studies has been found lower caspase-1 expression in HCC tissues[157,158]. In a study was determined caspase-1 expression in HCC patients (from HCC tissues and adjacent normal tissues) and in hepatocyte cell lines[157]. There was found a significant decrease in caspase-1 expression in HCC tissues compared to adjacent normal tissues and hepatocyte cell lines. Besides, the use of berberine increased the expression of caspase-1, decreased cell number, and increased cell swelling in hepatocyte cell lines; and the use of the caspase-1 inhibitor Ac-YVAD-CMK attenuated the effects of berberine.

However, in one study has been found that liver tissue of patients infected with hepatitis C virus (HCV) showed caspase-1-mediated pyroptosis[159]. Besides, in other study of patients with resection of HCC was found lower survival in patients with high of caspase-1 expression in normal tissues[160].

Angiopoietin-2

Angiopoietin-2 is a protein that is involved in angiogenesis and inflammation[161]. In a recently published study of chronic HCV patients treated with direct acting antivirals (DAA) was found that angiopoietin-2 in liver tissue was related with the risk of HCC recurrence or de novo ocurrence.

Another interesting finding of that study was that patients with HCC recurrence or de novo ocurrence had significantly higher portal pressure than patients never developing HCC[162]; and in previous studies was found that portal hypertension was associated with poor prognosis in patients undergoing to LT[163] or with HCC[164,165].

The risk of HCC occurrence or recurrence following DAA remains unclear due to that the results of different studies are contradictories. In a review published in 2017 including 10 studies was found in meta-analyses a higher incidence of HCC occurrence and HCC recurrence with the administration of DAA[166]. However, in meta-regression analyses after adjusting for study follow-up and age, DAA therapy was not associated with higher HCC de novo ocurrence and neither with HCC recurrence. In the study by Faillaci et al[162] was found that the use of DAA was associated with de novo HCC, and that this risk is higher in patients with higher angiopoietin-2 expression.

Genomic

The Cancer Genome Atlas (TCGA) Research Network published in 2017 the genomic characterization of HCC[167]. There were analyzed 363 HCC cases by whole-genome sequencing and DNA copy number, and 196 HCC cases by DNA methylation, mRNA, miRNA, and proteomic expression. In total, 12136 genes had non-silent mutations, and 26 genes were determined to be significantly mutated genes. Of these 26 genes, 18 were reported in at least one previous HCC genome sequencing study and 8 were not previously associated with HCC. Whithin of know mutated genes, the most included TERTpromoter (51%), TP53 (31%), CTNNB1 (27%), ALB (13%), APOB (10%), ARID1A (7%), AXIN1 (8%), ARID2 (5%), BAP1(5%), KEAP1 (5%), RB1 (4%), and NFE2L2 (3%). There were identified 8 novel mutated gene with a low frequence (2%-3% of HCC patients), including LTZR1, EEF1A1, AZIN1, RP1L 1, GPATCH4, CREB3L3, AHCTF1, and HIST1H1C. In addition, other two mutated genes previously associated with other cancer types were associated with HCC in this study, F3B1 and SMARCA4. Besides, integrative clustering of datasets of DNA copy number, DNA methylation, mRNA expression and miRNA expression could define three HCC subtypes (iClust 1 to 3), and iClust1 subtype had a poor prognosis. In addition, the analysis of these mutations and pathways provide potential directions for future potential therapeutic in HCC patients by the use of inhibitors of WNT, MDM4, MET, VEGFA, MCL1, IDH1, TERT. Thus, this genome-wide characterization has been very important in improving our knowledge about mutated genes associated with HCC, prognostic gene signatures and potential treatments[168].

CONCLUSION

Various macromorphological factors measured prior to LT have been classically used to estimate the outcomes of HCC patients undergoing LT. Additionally, the determination of some valid biomarkers prior to LT could help predict the prognoses of HCC patients undergoing LT. The most frequently examined biomarker is the serum AFP level. Recently, an association was reported between decreased survival rates and high blood levels of malondialdehyde, TAC, CCCK-18, sCD40L, substance P, CRP, and VEGF, NLR and PLR in blood, high peripheral blood expression of h-TERT mRNA, and high HCC expression of DKK1. In addition, an association has been found between increased HCC recurrence and high blood levels of Des-gamma-carboxy prothrombin, and high HCC expression of GPC-3, E-cadherin and beta-catenin. Additional research is necessary to establish the prognostic role of these biomarkers for HCC prior to LT. Furthermore, some of these biomarkers are also interesting because their potential modulation could help to create new research lines for improving the outcomes of those patients. Those new biomarkers are summarized on Table 1.

Table 1 New prognostic biomarkers in patients undergoing liver transplantation for hepatocellular carcinoma.
BiomarkerAlterationOutcomeRef.
MalondialdehydeHigh circulating levelsLower survival[]
Total antioxidant capacityHigh circulating levelsLower survival[29]
Caspase-cleaved cytokeratin-18High circulating levelsLower survival[54]
Soluble CD40 ligandHigh circulating levelsLower survival[80]
Substance PHigh circulating levelsLower survival[107]
Neutrophil to lymphocyte ratioHigh circulating ratioLower survival[120]
Platelet to lymphocyte ratioHigh circulating ratioLower survival[120]
C-reactive proteinHigh circulating levelsLower survival[121,122]
Des-gamma-carboxy prothrombinHigh circulating levelsHigher recurrence[123-127]
Glypican-3High HCC expressionHigher recurrence[128]
H-TERT mRNAHigh peripheral blood expressionLower survival[130,131]
E-cadherinHigh HCC expressionHigher recurrence[142]
Beta-cateninHigh HCC expressionHigher recurrence[142]
Dickkopf-1High HCC expressionLower survival[154]
Vascular endothelial growth factorHigh circulating levelsLower survival[156]
Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: Spain

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Gorrell MD, Wang K, Zhu X S- Editor: Wang XJ L- Editor: A E- Editor: Huang Y

References
1.  Bodzin AS, Busuttil RW. Hepatocellular carcinoma: Advances in diagnosis, management, and long term outcome. World J Hepatol. 2015;7:1157-1167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 62]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
2.  Guerrero-Misas M, Rodríguez-Perálvarez M, De la Mata M. Strategies to improve outcome of patients with hepatocellular carcinoma receiving a liver transplantation. World J Hepatol. 2015;7:649-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
3.  European Association For The Study Of The Liver. ; European Organisation For Research And Treatment Of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908-943.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4059]  [Cited by in F6Publishing: 4345]  [Article Influence: 362.1]  [Reference Citation Analysis (2)]
4.  Slotta JE, Kollmar O, Ellenrieder V, Ghadimi BM, Homayounfar K. Hepatocellular carcinoma: Surgeon’s view on latest findings and future perspectives. World J Hepatol. 2015;7:1168-1183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 40]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
5.  Gomaa AI, Waked I. Recent advances in multidisciplinary management of hepatocellular carcinoma. World J Hepatol. 2015;7:673-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 64]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
6.  Clark T, Maximin S, Meier J, Pokharel S, Bhargava P. Hepatocellular Carcinoma: Review of Epidemiology, Screening, Imaging Diagnosis, Response Assessment, and Treatment. Curr Probl Diagn Radiol. 2015;44:479-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 159]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
7.  Toyoda H, Kumada T, Tada T, Sone Y, Kaneoka Y, Maeda A. Tumor Markers for Hepatocellular Carcinoma: Simple and Significant Predictors of Outcome in Patients with HCC. Liver Cancer. 2015;4:126-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 98]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
8.  Cescon M, Bertuzzo VR, Ercolani G, Ravaioli M, Odaldi F, Pinna AD. Liver transplantation for hepatocellular carcinoma: role of inflammatory and immunological state on recurrence and prognosis. World J Gastroenterol. 2013;19:9174-9182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 40]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
9.  Lee HW, Suh KS. Expansion of the criteria for living donor liver transplantation for hepatocellular carcinoma. Curr Opin Organ Transplant. 2016;21:231-237.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
10.  Cillo U, Giuliani T, Polacco M, Herrero Manley LM, Crivellari G, Vitale A. Prediction of hepatocellular carcinoma biological behavior in patient selection for liver transplantation. World J Gastroenterol. 2016;22:232-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
11.  Romano M, De Francesco F, Pirozzi G, Gringeri E, Boetto R, Di Domenico M, Zavan B, Ferraro GA, Cillo U. Expression of cancer stem cell biomarkers as a tool for a correct therapeutic approach to hepatocellular carcinoma. Oncoscience. 2015;2:443-456.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Draper HH, Hadley M. Malondialdehyde determination as index of lipid peroxidation. Methods Enzymol. 1990;186:421-431.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Dalle-Donne I, Rossi R, Colombo R, Giustarini D, Milzani A. Biomarkers of oxidative damage in human disease. Clin Chem. 2006;52:601-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1075]  [Cited by in F6Publishing: 1071]  [Article Influence: 59.5]  [Reference Citation Analysis (0)]
14.  Yahya RS, Ghanem OH, Foyouh AA, Atwa M, Enany SA. Role of interleukin-8 and oxidative stress in patients with hepatocellular carcinoma. Clin Lab. 2013;59:969-976.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Zhao J, Zhao Y, Wang H, Gu X, Ji J, Gao C. Association between metabolic abnormalities and HBV related hepatocelluar carcinoma in Chinese: a cross-sectional study. Nutr J. 2011;10:49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
16.  Tsai SM, Lin SK, Lee KT, Hsiao JK, Huang JC, Wu SH, Ma H, Wu SH, Tsai LY. Evaluation of redox statuses in patients with hepatitis B virus-associated hepatocellular carcinoma. Ann Clin Biochem. 2009;46:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
17.  Trevisani F, Caraceni P, Simoncini M, Micati M, Domenicali M, Dazzani F, Zambruni A, Stefanelli C, Grazi G, Nardo B. Evidence of oxidative imbalance in long-term liver transplant patients. Dig Liver Dis. 2002;34:279-284.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Thorat VN, Suryakar AN, Naik P, Tiwale BM. Total antioxidant capacity and lipid peroxidation in liver transplantation. Indian J Clin Biochem. 2009;24:102-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
19.  Czeczot H, Scibior D, Skrzycki M, Podsiad M. Glutathione and GSH-dependent enzymes in patients with liver cirrhosis and hepatocellular carcinoma. Acta Biochim Pol. 2006;53:237-242.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Váli L, Hahn O, Kupcsulik P, Drahos A, Sárváry E, Szentmihályi K, Pallai Z, Kurucz T, Sípos P, Blázovics A. Oxidative stress with altered element content and decreased ATP level of erythrocytes in hepatocellular carcinoma and colorectal liver metastases. Eur J Gastroenterol Hepatol. 2008;20:393-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
21.  Suzuki Y, Imai K, Takai K, Hanai T, Hayashi H, Naiki T, Nishigaki Y, Tomita E, Shimizu M, Moriwaki H. Hepatocellular carcinoma patients with increased oxidative stress levels are prone to recurrence after curative treatment: a prospective case series study using the d-ROM test. J Cancer Res Clin Oncol. 2013;139:845-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
22.  Liu C, Zhou XS, Geng QM. [Evaluation oxygen free radicals related index before liver transplantation to forejudge prognosis]. Zhongguo Wei Zhong Bing Ji Jiu Yi Xue. 2003;15:560-562.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Lorente L, Rodriguez ST, Sanz P, Abreu-González P, Díaz D, Moreno AM, Borja E, Martín MM, Jiménez A, Barrera MA. Association between Pre-Transplant Serum Malondialdehyde Levels and Survival One Year after Liver Transplantation for Hepatocellular Carcinoma. Int J Mol Sci. 2016;17:500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
24.  Lorente L, Martín MM, Abreu-González P, Domínguez-Rodriguez A, Labarta L, Díaz C, Solé-Violán J, Ferreres J, Cabrera J, Igeño JC. Sustained high serum malondialdehyde levels are associated with severity and mortality in septic patients. Crit Care. 2013;17:R290.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Lorente L, Martín MM, Abreu-González P, Ramos L, Argueso M, Cáceres JJ, Solé-Violán J, Lorenzo JM, Molina I, Jiménez A. Association between serum malondialdehyde levels and mortality in patients with severe brain trauma injury. J Neurotrauma. 2015;32:1-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Lorente L, Martín MM, Abreu-González P, Ramos L, Argueso M, Solé-Violán J, Riaño-Ruiz M, Jiménez A. Serum malondialdehyde levels in patients with malignant middle cerebral artery infarction are associated with mortality. PLoS One. 2015;10:e0125893.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
27.  Lorente L, Martín MM, Abreu-González P, Sabatel R, Ramos L, Argueso M, Solé-Violán J, Riaño-Ruiz M, Jiménez A, García-Marín V. Serum Malondialdehyde Levels and Mortality in Patients with Spontaneous Intracerebral Hemorrhage. World Neurosurg. 2018;113:e542-e547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
28.  Ghiselli A, Serafini M, Natella F, Scaccini C. Total antioxidant capacity as a tool to assess redox status: critical view and experimental data. Free Radic Biol Med. 2000;29:1106-1114.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Lorente L, Rodriguez ST, Sanz P, Pérez-Cejas A, Abreu-González P, Padilla J, Díaz D, González A, Martín MM, Jiménez A. Serum total antioxidant capacity prior to liver transplantation for hepatocellular carcinoma is associated with 1-year liver transplantation survival. J Int Med Res. 2018;46:2641-2649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  Lowes DA, Webster NR, Murphy MP, Galley HF. Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis. Br J Anaesth. 2013;110:472-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 222]  [Article Influence: 20.2]  [Reference Citation Analysis (0)]
31.  Horáková L, Ondrejicková O, Bachratá K, Vajdová M. Preventive effect of several antioxidants after oxidative stress on rat brain homogenates. Gen Physiol Biophys. 2000;19:195-205.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Gitto E, Karbownik M, Reiter RJ, Tan DX, Cuzzocrea S, Chiurazzi P, Cordaro S, Corona G, Trimarchi G, Barberi I. Effects of melatonin treatment in septic newborns. Pediatr Res. 2001;50:756-760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 360]  [Cited by in F6Publishing: 349]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
33.  Ullegaddi R, Powers HJ, Gariballa SE. Antioxidant supplementation enhances antioxidant capacity and mitigates oxidative damage following acute ischaemic stroke. Eur J Clin Nutr. 2005;59:1367-1373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
34.  Saniova B, Drobny M, Lehotsky J, Sulaj M, Schudichova J. Biochemical and clinical improvement of cytotoxic state by amantadine sulphate. Cell Mol Neurobiol. 2006;26:1475-1482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
35.  Luedde T, Kaplowitz N, Schwabe RF. Cell death and cell death responses in liver disease: mechanisms and clinical relevance. Gastroenterology. 2014;147:765-783.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 512]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
36.  Canbay A, Feldstein A, Kronenberger B, Schulze-Osthoff K, Bantel H. [Cytokeratin 18 as marker for non-invasive diagnosis and prognosis of acute and chronic liver diseases]. Z Gastroenterol. 2014;52:290-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
37.  Yilmaz Y. Systematic review: caspase-cleaved fragments of cytokeratin 18 - the promises and challenges of a biomarker for chronic liver disease. Aliment Pharmacol Ther. 2009;30:1103-1109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 68]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
38.  Chu PG, Weiss LM. Keratin expression in human tissues and neoplasms. Histopathology. 2002;40:403-439.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Caulín C, Salvesen GS, Oshima RG. Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis. J Cell Biol. 1997;138:1379-1394.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Leers MP, Kölgen W, Björklund V, Bergman T, Tribbick G, Persson B, Björklund P, Ramaekers FC, Björklund B, Nap M. Immunocytochemical detection and mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis. J Pathol. 1999;187:567-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
41.  Hägg M, Bivén K, Ueno T, Rydlander L, Björklund P, Wiman KG, Shoshan M, Linder S. A novel high-through-put assay for screening of pro-apoptotic drugs. Invest New Drugs. 2002;20:253-259.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Oyama K, Fushida S, Kinoshita J, Okamoto K, Makino I, Nakamura K, Hayashi H, Inokuchi M, Nakagawara H, Tajima H. Serum cytokeratin 18 as a biomarker for gastric cancer. Clin Exp Med. 2013;13:289-295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
43.  Ueno T, Toi M, Bivén K, Bando H, Ogawa T, Linder S. Measurement of an apoptotic product in the sera of breast cancer patients. Eur J Cancer. 2003;39:769-774.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Ulukaya E, Yilmaztepe A, Akgoz S, Linder S, Karadag M. The levels of caspase-cleaved cytokeratin 18 are elevated in serum from patients with lung cancer and helpful to predict the survival. Lung Cancer. 2007;56:399-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 81]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
45.  Yaman E, Coskun U, Sancak B, Buyukberber S, Ozturk B, Benekli M. Serum M30 levels are associated with survival in advanced gastric carcinoma patients. Int Immunopharmacol. 2010;10:719-722.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
46.  de Haas EC, di Pietro A, Simpson KL, Meijer C, Suurmeijer AJ, Lancashire LJ, Cummings J, de Jong S, de Vries EG, Dive C. Clinical evaluation of M30 and M65 ELISA cell death assays as circulating biomarkers in a drug-sensitive tumor, testicular cancer. Neoplasia. 2008;10:1041-1048.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Koelink PJ, Lamers CB, Hommes DW, Verspaget HW. Circulating cell death products predict clinical outcome of colorectal cancer patients. BMC Cancer. 2009;9:88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
48.  Wu YX, Wang JH, Wang H, Yang XY. [Study on expression of Ki-67, early apoptotic protein M30 in endometrial carcinoma and their correlation with prognosis]. Zhonghua Bing Li Xue Za Zhi. 2003;32:314-318.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Gonzalez-Quintela A, Mallo N, Mella C, Campos J, Perez LF, Lopez-Rodriguez R, Tome S, Otero E. Serum levels of cytokeratin-18 (tissue polypeptide-specific antigen) in liver diseases. Liver Int. 2006;26:1217-1224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
50.  Yagmur E, Trautwein C, Leers MP, Gressner AM, Tacke F. Elevated apoptosis-associated cytokeratin 18 fragments (CK18Asp386) in serum of patients with chronic liver diseases indicate hepatic and biliary inflammation. Clin Biochem. 2007;40:651-655.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 45]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
51.  Godin C, Louandre C, Bodeau S, Diouf M, Saidak Z, Conte MA, Chauffert B, Barbare JC, Barget N, Trinchet JC. Biomarkers of apoptosis and necrosis in patients with hepatocellular carcinoma treated with sorafenib. Anticancer Res. 2015;35:1803-1808.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Waidmann O, Köberle V, Bettinger D, Trojan J, Zeuzem S, Schultheiß M, Kronenberger B, Piiper A. Diagnostic and prognostic significance of cell death and macrophage activation markers in patients with hepatocellular carcinoma. J Hepatol. 2013;59:769-779.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
53.  Morris KL, Tugwood JD, Khoja L, Lancashire M, Sloane R, Burt D, Shenjere P, Zhou C, Hodgson C, Ohtomo T. Circulating biomarkers in hepatocellular carcinoma. Cancer Chemother Pharmacol. 2014;74:323-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 43]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
54.  Lorente L, Rodriguez ST, Sanz P, Pérez-Cejas A, Padilla J, Díaz D, González A, Martín MM, Jiménez A, Barrera MA. Prognostic Value of Serum Caspase-Cleaved Cytokeratin-18 Levels before Liver Transplantation for One-Year Survival of Patients with Hepatocellular Carcinoma. Int J Mol Sci. 2016;17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
55.  Lorente L, Martín MM, González-Rivero AF, Ferreres J, Solé-Violán J, Labarta L, Díaz C, Jiménez A, Borreguero-León JM. Serum levels of caspase-cleaved cytokeratin-18 and mortality are associated in severe septic patients: pilot study. PLoS One. 2014;9:e109618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
56.  Lorente L, Martín MM, González-Rivero AF, Argueso M, Ramos L, Solé-Violán J, Cáceres JJ, Jiménez A, Borreguero-León JM. Serum levels of caspase-cleaved cytokeratin-18 in patients with severe traumatic brain injury are associated with mortality: a pilot study. PLoS One. 2015;10:e0121739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
57.  Lorente L, Martín MM, Pérez-Cejas A, Ramos L, Argueso M, Solé-Violán J, Cáceres JJ, Jiménez A, García-Marín V. High serum levels of caspase-cleaved cytokeratin-18 are associated with malignant middle cerebral artery infarction patient mortality. BMC Neurol. 2018;18:32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
58.  Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS, Stefanadis C. The CD40/CD40 ligand system: linking inflammation with atherothrombosis. J Am Coll Cardiol. 2009;54:669-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 243]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
59.  Ferroni P, Santilli F, Guadagni F, Basili S, Davì G. Contribution of platelet-derived CD40 ligand to inflammation, thrombosis and neoangiogenesis. Curr Med Chem. 2007;14:2170-2180.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Aukrust P, Damas JK, Solum NO. Soluble CD40 ligand and platelets: self-perpetuating pathogenic loop in thrombosis and inflammation? J Am Coll Cardiol. 2004;43:2326-2328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 55]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
61.  Anand SX, Viles-Gonzalez JF, Badimon JJ, Cavusoglu E, Marmur JD. Membrane-associated CD40L and sCD40L in atherothrombotic disease. Thromb Haemost. 2003;90:377-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 114]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
62.  Zhang B, Wu T, Chen M, Zhou Y, Yi D, Guo R. The CD40/CD40L system: a new therapeutic target for disease. Immunol Lett. 2013;153:58-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
63.  Zhang T, Pierson RN 3rd, Azimzadeh AM. Update on CD40 and CD154 blockade in transplant models. Immunotherapy. 2015;7:899-911.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (1)]
64.  Pinelli DF, Ford ML. Novel insights into anti-CD40/CD154 immunotherapy in transplant tolerance. Immunotherapy. 2015;7:399-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 46]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
65.  Aloui C, Prigent A, Sut C, Tariket S, Hamzeh-Cognasse H, Pozzetto B, Richard Y, Cognasse F, Laradi S, Garraud O. The signaling role of CD40 ligand in platelet biology and in platelet component transfusion. Int J Mol Sci. 2014;15:22342-22364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 117]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
66.  Mao DJ, Guo RY, Tang YC, Zang YH. [Expression of sCD40L in peripheral blood and NF-κBp65 in PBMC of patients with acute progressive cerebral infarction]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 2011;27:177-179.  [PubMed]  [DOI]  [Cited in This Article: ]
67.  Tuttolomondo A, Di Raimondo D, Di Sciacca R, Casuccio A, Bivona G, Bellia C, Barreca L, Serio A, D’Aguanno G, Ciaccio M. Fetuin-A and CD40 L plasma levels in acute ischemic stroke: differences in relation to TOAST subtype and correlation with clinical and laboratory variables. Atherosclerosis. 2010;208:290-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
68.  Davì G, Tuttolomondo A, Santilli F, Basili S, Ferrante E, Di Raimondo D, Pinto A, Licata G. CD40 ligand and MCP-1 as predictors of cardiovascular events in diabetic patients with stroke. J Atheroscler Thromb. 2009;16:707-713.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Lorente L, Martín MM, González-Rivero AF, Ramos L, Argueso M, Cáceres JJ, Solé-Violán J, Jiménez A, Borreguero-León JM. Association between Serum Soluble CD154 Levels and Mortality in Patients with Malignant Middle Cerebral Artery Infarction. Int J Mol Sci. 2015;16:12147-12158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
70.  Aukrust P, Müller F, Ueland T, Berget T, Aaser E, Brunsvig A, Solum NO, Forfang K, Frøland SS, Gullestad L. Enhanced levels of soluble and membrane-bound CD40 ligand in patients with unstable angina. Possible reflection of T lymphocyte and platelet involvement in the pathogenesis of acute coronary syndromes. Circulation. 1999;100:614-620.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Varo N, de Lemos JA, Libby P, Morrow DA, Murphy SA, Nuzzo R, Gibson CM, Cannon CP, Braunwald E, Schönbeck U. Soluble CD40L: risk prediction after acute coronary syndromes. Circulation. 2003;108:1049-1052.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 276]  [Cited by in F6Publishing: 281]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
72.  Lorente L, Martín MM, Varo N, Borreguero-León JM, Solé-Violán J, Blanquer J, Labarta L, Díaz C, Jiménez A, Pastor E. Association between serum soluble CD40 ligand levels and mortality in patients with severe sepsis. Crit Care. 2011;15:R97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
73.  Martínez de Lizarrondo S, Roncal C, Calvayrac O, Rodríguez C, Varo N, Purroy A, Lorente L, Rodríguez JA, Doeuvre L, Hervás-Stubbs S. Synergistic effect of thrombin and CD40 ligand on endothelial matrix metalloproteinase-10 expression and microparticle generation in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2012;32:1477-1487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
74.  Dominguez-Rodriguez A, Abreu-Gonzalez P, Garcia-Gonzalez MJ, Kaski JC. Soluble CD40 ligand:interleukin-10 ratio predicts in-hospital adverse events in patients with ST-segment elevation myocardial infarction. Thromb Res. 2007;121:293-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
75.  Lorente L, Martín MM, González-Rivero AF, Ramos L, Argueso M, Cáceres JJ, Solé-Violán J, Serrano N, Rodríguez ST, Jiménez A. Serum soluble CD40 Ligand levels are associated with severity and mortality of brain trauma injury patients. Thromb Res. 2014;134:832-836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
76.  Lapiński TW, Pogorzelska J, Grzeszczuk A, Swiderska M, Kowalczuk O, Nikliński J, Flisiak R. The importance of sCD40 and sCD40L concentration in patients with chronic HCV infection and HIV co-infection. Przegl Epidemiol. 2014;68:11-15, 105-108.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Basili S, Raparelli V, Riggio O, Merli M, Carnevale R, Angelico F, Tellan G, Pignatelli P, Violi F; CALC Group. NADPH oxidase-mediated platelet isoprostane over-production in cirrhotic patients: implication for platelet activation. Liver Int. 2011;31:1533-1540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
78.  Sookoian S, Castaño GO, Burgueño AL, Rosselli MS, Gianotti TF, Mallardi P, Martino JS, Pirola CJ. Circulating levels and hepatic expression of molecular mediators of atherosclerosis in nonalcoholic fatty liver disease. Atherosclerosis. 2010;209:585-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
79.  Eltaher SM, El-Gil R, Fouad N, Mitwali R, El-Kholy H. Evaluation of serum levels and significance of soluble CD40 ligand in screening patients with hepatitis C virus-related hepatocellular carcinoma. East Mediterr Health J. 2016;22:603-610.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Lorente L, Rodriguez ST, Sanz P, Pérez-Cejas A, Padilla J, Díaz D, González A, Martín MM, Jiménez A, Barrera MA. High serum soluble CD40L levels previously to liver transplantation in patients with hepatocellular carcinoma are associated with mortality at one year. J Crit Care. 2018;43:316-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
81.  Noelle RJ, Roy M, Shepherd DM, Stamenkovic I, Ledbetter JA, Aruffo A. A 39-kDa protein on activated helper T cells binds CD40 and transduces the signal for cognate activation of B cells. Proc Natl Acad Sci USA. 1992;89:6550-6554.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Mach F, Schönbeck U, Sukhova GK, Bourcier T, Bonnefoy JY, Pober JS, Libby P. Functional CD40 ligand is expressed on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for CD40-CD40 ligand signaling in atherosclerosis. Proc Natl Acad Sci USA. 1997;94:1931-1936.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Zhou L, Stordeur P, de Lavareille A, Thielemans K, Capel P, Goldman M, Pradier O. CD40 engagement on endothelial cells promotes tissue factor-dependent procoagulant activity. Thromb Haemost. 1998;79:1025-1028.  [PubMed]  [DOI]  [Cited in This Article: ]
84.  Hezi-Yamit A, Wong PW, Bien-Ly N, Komuves LG, Prasad KS, Phillips DR, Sinha U. Synergistic induction of tissue factor by coagulation factor Xa and TNF: evidence for involvement of negative regulatory signaling cascades. Proc Natl Acad Sci USA. 2005;102:12077-12082.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
85.  Miller DL, Yaron R, Yellin MJ. CD40L-CD40 interactions regulate endothelial cell surface tissue factor and thrombomodulin expression. J Leukoc Biol. 1998;63:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Slupsky JR, Kalbas M, Willuweit A, Henn V, Kroczek RA, Müller-Berghaus G. Activated platelets induce tissue factor expression on human umbilical vein endothelial cells by ligation of CD40. Thromb Haemost. 1998;80:1008-1014.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Prasad KS, Andre P, He M, Bao M, Manganello J, Phillips DR. Soluble CD40 ligand induces beta3 integrin tyrosine phosphorylation and triggers platelet activation by outside-in signaling. Proc Natl Acad Sci USA. 2003;100:12367-12371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 161]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
88.  André P, Prasad KS, Denis CV, He M, Papalia JM, Hynes RO, Phillips DR, Wagner DD. CD40L stabilizes arterial thrombi by a beta3 integrin--dependent mechanism. Nat Med. 2002;8:247-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 565]  [Cited by in F6Publishing: 550]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
89.  Li J, Zhao SP, Peng DQ, Xu ZM, Zhou HN. Early effect of pravastatin on serum soluble CD40L, matrix metalloproteinase-9, and C-reactive protein in patients with acute myocardial infarction. Clin Chem. 2004;50:1696-1699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
90.  Hamdan R, Hajj F, Kadry Z, Kassab R, Salame E, Aboujaoude S, Azar R, Badaoui G. Benefit and tolerability of the coadministration of ezetimibe and atorvastatin in acute coronary syndrome patients. J Med Liban. 2011;59:65-69.  [PubMed]  [DOI]  [Cited in This Article: ]
91.  Han SH, Koh KK, Quon MJ, Lee Y, Shin EK. The effects of simvastatin, losartan, and combined therapy on soluble CD40 ligand in hypercholesterolemic, hypertensive patients. Atherosclerosis. 2007;190:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
92.  Ní Chróinín D, Asplund K, Åsberg S, Callaly E, Cuadrado-Godia E, Díez-Tejedor E, Di Napoli M, Engelter ST, Furie KL, Giannopoulos S. Statin therapy and outcome after ischemic stroke: systematic review and meta-analysis of observational studies and randomized trials. Stroke. 2013;44:448-456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 168]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
93.  Tleyjeh IM, Kashour T, Hakim FA, Zimmerman VA, Erwin PJ, Sutton AJ, Ibrahim T. Statins for the prevention and treatment of infections: a systematic review and meta-analysis. Arch Intern Med. 2009;169:1658-1667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 190]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
94.  Thomsen RW, Riis A, Kornum JB, Christensen S, Johnsen SP, Sørensen HT. Preadmission use of statins and outcomes after hospitalization with pneumonia: population-based cohort study of 29,900 patients. Arch Intern Med. 2008;168:2081-2087.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 138]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
95.  Hsu J, Andes DR, Knasinski V, Pirsch J, Safdar N. Statins are associated with improved outcomes of bloodstream infection in solid-organ transplant recipients. Eur J Clin Microbiol Infect Dis. 2009;28:1343-1351.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
96.  Dobesh PP, Klepser DG, McGuire TR, Morgan CW, Olsen KM. Reduction in mortality associated with statin therapy in patients with severe sepsis. Pharmacotherapy. 2009;29:621-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
97.  Almeida TA, Rojo J, Nieto PM, Pinto FM, Hernandez M, Martín JD, Candenas ML. Tachykinins and tachykinin receptors: structure and activity relationships. Curr Med Chem. 2004;11:2045-2081.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Pennefather JN, Lecci A, Candenas ML, Patak E, Pinto FM, Maggi CA. Tachykinins and tachykinin receptors: a growing family. Life Sci. 2004;74:1445-1463.  [PubMed]  [DOI]  [Cited in This Article: ]
99.  Satake H, Kawada T. Overview of the primary structure, tissue-distribution, and functions of tachykinins and their receptors. Curr Drug Targets. 2006;7:963-974.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Trivedi M, Bergasa NV. Serum concentrations of substance P in cholestasis. Ann Hepatol. 2010;9:177-180.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  El-Raziky MS, Gohar N, El-Raziky M. Study of substance P, renin and aldosterone in chronic liver disease in Egyptian children. J Trop Pediatr. 2005;51:320-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
102.  Li CP, Lee FY, Hwang SJ, Chang FY, Lin HC, Lu RH, Hou MC, Chu CJ, Chan CC, Luo JC. Role of substance P in the pathogenesis of spider angiomas in patients with nonalcoholic liver cirrhosis. Am J Gastroenterol. 1999;94:502-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
103.  Uemura M, Tsujii T, Kikuchi E, Fukui H, Tsukamoto N, Matsumura M, Fujimoto M, Koizumi M, Takaya A, Kojima H. Increased plasma levels of substance P and disturbed water excretion in patients with liver cirrhosis. Scand J Gastroenterol. 1998;33:860-866.  [PubMed]  [DOI]  [Cited in This Article: ]
104.  Lee FY, Lin HC, Tsai YT, Chang FY, Lu RH, Hou MC, Li CP, Chu CJ, Wang SS, Lee SD. Plasma substance P levels in patients with liver cirrhosis: relationship to systemic and portal hemodynamics. Am J Gastroenterol. 1997;92:2080-2084.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Fernández-Rodriguez CM, Prieto J, Quiroga J, Zozoya JM, Andrade A, Núñez M, Sangro B, Penas J. Plasma levels of substance P in liver cirrhosis: relationship to the activation of vasopressor systems and urinary sodium excretion. Hepatology. 1995;21:35-40.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Hörtnagl H, Singer EA, Lenz K, Kleinberger G, Lochs H. Substance P is markedly increased in plasma of patients with hepatic coma. Lancet. 1984;1:480-483.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Lorente L, Rodriguez ST, Sanz P, Pérez-Cejas A, Padilla J, Díaz D, González A, Martín MM, Jiménez A, Cerro P. Patients with high serum substance P levels previously to liver transplantation for hepatocellular carcinoma have higher risk of one-year liver transplantation mortality. Oncotarget. 2018;9:21552-21559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
108.  Lorente L, Martín MM, Almeida T, Hernández M, Ramos L, Argueso M, Cáceres JJ, Solé-Violán J, Jiménez A. Serum substance P levels are associated with severity and mortality in patients with severe traumatic brain injury. Crit Care. 2015;19:192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
109.  Lorente L, Martín MM, Almeida T, Pérez-Cejas A, Ramos L, Argueso M, Riaño-Ruiz M, Solé-Violán J, Hernández M. Serum Levels of Substance P and Mortality in Patients with a Severe Acute Ischemic Stroke. Int J Mol Sci. 2016;17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
110.  Lotz M, Vaughan JH, Carson DA. Effect of neuropeptides on production of inflammatory cytokines by human monocytes. Science. 1988;241:1218-1221.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  Laurenzi MA, Persson MA, Dalsgaard CJ, Haegerstrand A. The neuropeptide substance P stimulates production of interleukin 1 in human blood monocytes: activated cells are preferentially influenced by the neuropeptide. Scand J Immunol. 1990;31:529-533.  [PubMed]  [DOI]  [Cited in This Article: ]
112.  Ansel JC, Brown JR, Payan DG, Brown MA. Substance P selectively activates TNF-alpha gene expression in murine mast cells. J Immunol. 1993;150:4478-4485.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Rameshwar P, Gascón P. Substance P (SP) mediates production of stem cell factor and interleukin-1 in bone marrow stroma: potential autoregulatory role for these cytokines in SP receptor expression and induction. Blood. 1995;86:482-490.  [PubMed]  [DOI]  [Cited in This Article: ]
114.  Yamaguchi M, Kojima T, Kanekawa M, Aihara N, Nogimura A, Kasai K. Neuropeptides stimulate production of interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha in human dental pulp cells. Inflamm Res. 2004;53:199-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 69]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
115.  Turner RJ, Helps SC, Thornton E, Vink R. A substance P antagonist improves outcome when administered 4 h after onset of ischaemic stroke. Brain Res. 2011;1393:84-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
116.  Turner RJ, Vink R. NK1 tachykinin receptor treatment is superior to capsaicin pre-treatment in improving functional outcome following acute ischemic stroke. Neuropeptides. 2014;48:267-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
117.  Yu Z, Cheng G, Huang X, Li K, Cao X. Neurokinin-1 receptor antagonist SR140333: A novel type of drug to treat cerebral ischemia. Neuroreport. 1997;8:2117-2119.  [PubMed]  [DOI]  [Cited in This Article: ]
118.  Gabrielian L, Helps SC, Thornton E, Turner RJ, Leonard AV, Vink R. Substance P antagonists as a novel intervention for brain edema and raised intracranial pressure. Acta Neurochir Suppl. 2013;118:201-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
119.  Donkin JJ, Cernak I, Blumbergs PC, Vink R. A substance P antagonist reduces axonal injury and improves neurologic outcome when administered up to 12 hours after traumatic brain injury. J Neurotrauma. 2011;28:217-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 49]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
120.  Zheng J, Cai J, Li H, Zeng K, He L, Fu H, Zhang J, Chen L, Yao J, Zhang Y. Neutrophil to Lymphocyte Ratio and Platelet to Lymphocyte Ratio as Prognostic Predictors for Hepatocellular Carcinoma Patients with Various Treatments: a Meta-Analysis and Systematic Review. Cell Physiol Biochem. 2017;44:967-981.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 165]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
121.  An HJ, Jang JW, Bae SH, Choi JY, Yoon SK, Lee MA, You YK, Kim DG, Jung ES. Serum C-reactive protein is a useful biomarker for predicting outcomes after liver transplantation in patients with hepatocellular carcinoma. Liver Transpl. 2012;18:1406-1414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 61]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
122.  Mori S, Choi Y, Park MS, Kim H, Hong G, Yi NJ, Lee KW, Suh KS. Usefulness of preoperative C-reactive protein and alpha-fetoprotein levels for prognostication of patients with hepatocellular carcinoma after living donor liver transplantation. Hepatogastroenterology. 2014;61:2353-2358.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Harimoto N, Yoshizumi T, Shimagaki T, Nagatsu A, Motomura T, Harada N, Okabe H, Itoh S, Ikegami T, Uchiyama H. Inflammation-based Prognostic Score in Patients with Living Donor Liver Transplantation for Hepatocellular Carcinoma. Anticancer Res. 2016;36:5537-5542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
124.  Fujiki M, Takada Y, Ogura Y, Oike F, Kaido T, Teramukai S, Uemoto S. Significance of des-gamma-carboxy prothrombin in selection criteria for living donor liver transplantation for hepatocellular carcinoma. Am J Transplant. 2009;9:2362-2371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
125.  Taketomi A, Sanefuji K, Soejima Y, Yoshizumi T, Uhciyama H, Ikegami T, Harada N, Yamashita Y, Sugimachi K, Kayashima H. Impact of des-gamma-carboxy prothrombin and tumor size on the recurrence of hepatocellular carcinoma after living donor liver transplantation. Transplantation. 2009;87:531-537.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 140]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
126.  Shindoh J, Sugawara Y, Nagata R, Kaneko J, Tamura S, Aoki T, Sakamoto Y, Hasegawa K, Tanaka T, Kokudo N. Evaluation methods for pretransplant oncologic markers and their prognostic impacts in patient undergoing living donor liver transplantation for hepatocellular carcinoma. Transpl Int. 2014;27:391-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
127.  Lee JH, Cho Y, Kim HY, Cho EJ, Lee DH, Yu SJ, Lee JW, Yi NJ, Lee KW, Kim SH. Serum Tumor Markers Provide Refined Prognostication in Selecting Liver Transplantation Candidate for Hepatocellular Carcinoma Patients Beyond the Milan Criteria. Ann Surg. 2016;263:842-850.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 96]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
128.  Wang YL, Zhu ZJ, Teng DH, Yao Z, Gao W, Shen ZY. Glypican-3 expression and its relationship with recurrence of HCC after liver transplantation. World J Gastroenterol. 2012;18:2408-2414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 40]  [Cited by in F6Publishing: 40]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
129.  Jing JS, Ye W, Jiang YK, Ma J, Zhu MQ, Ma JM, Zhou H, Yu LQ, Yang YF, Wang SC. The Value of GPC3 and GP73 in Clinical Diagnosis of Hepatocellular Carcinoma. Clin Lab. 2017;63:1903-1909.  [PubMed]  [DOI]  [Cited in This Article: ]
130.  Oya H, Sato Y, Yamamoto S, Nakatsuka H, Kobayashi T, Hara Y, Waguri N, Suda T, Aoyagi Y, Hatakeyama K. Comparison between human-telomerase reverse transcriptase mRNA and alpha-fetoprotein mRNA as a predictive value for recurrence of hepatocellular carcinoma in living donor liver transplantation. Transplant Proc. 2006;38:3636-3639.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
131.  Sato Y, Yamamoto S, Oya H, Nakatsuka H, Kobayashi T, Takeishi T, Hirano K, Hara Y, Watanabe T, Waguri N. Preoperative human-telomerase reverse transcriptase mRNA in peripheral blood and tumor recurrence in living-related liver transplantation for hepatocellular carcinoma. Hepatogastroenterology. 2005;52:1325-1328.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Kim YD, Hwang S, Lee YJ, Kim KH, Ahn CS, Park KM, Moon DB, Ha TY, Song GW, Jung DH. Preoperative peripheral blood human telomerase reverse transcriptase mRNA concentration is not a prognostic factor for resection of hepatocellular carcinoma. Hepatogastroenterology. 2012;59:1512-1515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 5]  [Reference Citation Analysis (0)]
133.  Nart D, Yaman B, Yilmaz F, Zeytunlu M, Karasu Z, Kiliç M. Expression of matrix metalloproteinase-9 in predicting prognosis of hepatocellular carcinoma after liver transplantation. Liver Transpl. 2010;16:621-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
134.  Zhang Q, Chen X, Zhou J, Zhang L, Zhao Q, Chen G, Xu J, Qian F, Chen Z. CD147, MMP-2, MMP-9 and MVD-CD34 are significant predictors of recurrence after liver transplantation in hepatocellular carcinoma patients. Cancer Biol Ther. 2006;5:808-814.  [PubMed]  [DOI]  [Cited in This Article: ]
135.  Kuyvenhoven JP, Verspaget HW, Gao Q, Ringers J, Smit VT, Lamers CB, van Hoek B. Assessment of serum matrix metalloproteinases MMP-2 and MMP-9 after human liver transplantation: increased serum MMP-9 level in acute rejection. Transplantation. 2004;77:1646-1652.  [PubMed]  [DOI]  [Cited in This Article: ]
136.  Lempinen M, Lyytinen I, Nordin A, Tervahartiala T, Mäkisalo H, Sorsa T, Isoniemi H. Prognostic value of serum MMP-8, -9 and TIMP-1 in patients with hepatocellular carcinoma. Ann Med. 2013;45:482-487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
137.  Lorente L, Martín MM, Ramos L, Cáceres JJ, Solé-Violán J, Argueso M, Jiménez A, Borreguero-León JM, Orbe J, Rodríguez JA. Serum tissue inhibitor of matrix metalloproteinase-1 levels are associated with mortality in patients with malignant middle cerebral artery infarction. BMC Neurol. 2015;15:111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
138.  Lorente L, Martín MM, López P, Ramos L, Blanquer J, Cáceres JJ, Solé-Violán J, Solera J, Cabrera J, Argueso M. Association between serum tissue inhibitor of matrix metalloproteinase-1 levels and mortality in patients with severe brain trauma injury. PLoS One. 2014;9:e94370.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
139.  Lorente L, Martín MM, Solé-Violán J, Blanquer J, Labarta L, Díaz C, Borreguero-León JM, Orbe J, Rodríguez JA, Jiménez A. Association of sepsis-related mortality with early increase of TIMP-1/MMP-9 ratio. PLoS One. 2014;9:e94318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 51]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
140.  Lorente L, Martín MM, Labarta L, Díaz C, Solé-Violán J, Blanquer J, Orbe J, Rodríguez JA, Jiménez A, Borreguero-León JM. Matrix metalloproteinase-9, -10, and tissue inhibitor of matrix metalloproteinases-1 blood levels as biomarkers of severity and mortality in sepsis. Crit Care. 2009;13:R158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 89]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
141.  Soyama A, Eguchi S, Takatsuki M, Kawashita Y, Hidaka M, Tokai H, Nagayoshi S, Mochizuki S, Matsumoto S, Hamasaki K. Significance of the serum level of soluble E-cadherin in patients with HCC. Hepatogastroenterology. 2008;55:1390-1393.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Fiorentino M, Altimari A, Ravaioli M, Gruppioni E, Gabusi E, Corti B, Vivarelli M, Bringuier PP, Scoazec JY, Grigioni WF. Predictive value of biological markers for hepatocellular carcinoma patients treated with orthotopic liver transplantation. Clin Cancer Res. 2004;10:1789-1795.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  Ataide EC, Perales SR, Silva MG, Filho FC, Sparapani AC, Latuf Filho PF, Stucchi RSB, Vassallo J, Escanhoela CAF, Boin IFSF. Immunoexpression of Heat Shock Protein 70, Glypican 3, Glutamine Synthetase, and Beta-Catenin in Hepatocellular Carcinoma After Liver Transplantation: Association Between Positive Glypican 3 and Beta-Catenin With the Presence of Larger Nodules. Transplant Proc. 2017;49:858-862.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
144.  Hakeem AR, Young RS, Marangoni G, Lodge JP, Prasad KR. Systematic review: the prognostic role of alpha-fetoprotein following liver transplantation for hepatocellular carcinoma. Aliment Pharmacol Ther. 2012;35:987-999.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
145.  Pommergaard HC, Burcharth J, Rosenberg J, Rasmussen A. Serologic and molecular biomarkers for recurrence of hepatocellular carcinoma after liver transplantation: A systematic review and meta-analysis. Transplant Rev (Orlando). 2016;30:171-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
146.  Shen Q, Fan J, Yang XR, Tan Y, Zhao W, Xu Y, Wang N, Niu Y, Wu Z, Zhou J. Serum DKK1 as a protein biomarker for the diagnosis of hepatocellular carcinoma: a large-scale, multicentre study. Lancet Oncol. 2012;13:817-826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 265]  [Cited by in F6Publishing: 289]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
147.  Erdal H, Gül Utku Ö, Karatay E, Çelik B, Elbeg Ş, Doğan İ. Combination of DKK1 and AFP improves diagnostic accuracy of hepatocellular carcinoma compared with either marker alone. Turk J Gastroenterol. 2016;27:375-381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
148.  Kim SU, Park JH, Kim HS, Lee JM, Lee HG, Kim H, Choi SH, Baek S, Kim BK, Park JY. Serum Dickkopf-1 as a Biomarker for the Diagnosis of Hepatocellular Carcinoma. Yonsei Med J. 2015;56:1296-1306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
149.  Ge T, Shen Q, Wang N, Zhang Y, Ge Z, Chu W, Lv X, Zhao F, Zhao W, Fan J. Diagnostic values of alpha-fetoprotein, dickkopf-1, and osteopontin for hepatocellular carcinoma. Med Oncol. 2015;32:59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
150.  Fouad YM, Mohamed HI, Kamal EM, Rasek MA. Clinical significance and diagnostic value of serum dickkopf-1 in patients with hepatocellular carcinoma. Scand J Gastroenterol. 2016;51:1133-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
151.  Jang ES, Jeong SH, Kim JW, Choi YS, Leissner P, Brechot C. Diagnostic Performance of Alpha-Fetoprotein, Protein Induced by Vitamin K Absence, Osteopontin, Dickkopf-1 and Its Combinations for Hepatocellular Carcinoma. PLoS One. 2016;11:e0151069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
152.  Liu Y, Tang W, Xie L, Wang J, Deng Y, Peng Q, Zhai L, Li S, Qin X. Prognostic significance of dickkopf-1 overexpression in solid tumors: a meta-analysis. Tumour Biol. 2014;35:3145-3154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
153.  Sakabe T, Azumi J, Umekita Y, Toriguchi K, Hatano E, Hirooka Y, Shiota G. Expression of Cancer Stem Cell-associated DKK1 mRNA Serves as Prognostic Marker for Hepatocellular Carcinoma. Anticancer Res. 2017;37:4881-4888.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
154.  Huang Y, Yang X, Zhao F, Shen Q, Wang Z, Lv X, Hu B, Yu B, Fan J, Qin W. Overexpression of Dickkopf-1 predicts poor prognosis for patients with hepatocellular carcinoma after orthotopic liver transplantation by promoting cancer metastasis and recurrence. Med Oncol. 2014;31:966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
155.  Zhan P, Qian Q, Yu LK. Serum VEGF level is associated with the outcome of patients with hepatocellular carcinoma: a meta-analysis. Hepatobiliary Surg Nutr. 2013;2:209-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 26]  [Reference Citation Analysis (0)]
156.  Zhang W, Kim R, Quintini C, Hashimoto K, Fujiki M, Diago T, Eghtesad B, Miller C, Fung J, Tan A. Prognostic role of plasma vascular endothelial growth factor in patients with hepatocellular carcinoma undergoing liver transplantation. Liver Transpl. 2015;21:101-111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
157.  Chu Q, Jiang Y, Zhang W, Xu C, Du W, Tuguzbaeva G, Qin Y, Li A, Zhang L, Sun G. Pyroptosis is involved in the pathogenesis of human hepatocellular carcinoma. Oncotarget. 2016;7:84658-84665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 101]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
158.  Wan L, Yuan X, Liu M, Xue B. miRNA-223-3p regulates NLRP3 to promote apoptosis and inhibit proliferation of hep3B cells. Exp Ther Med. 2018;15:2429-2435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
159.  Kofahi HM, Taylor NG, Hirasawa K, Grant MD, Russell RS. Hepatitis C Virus Infection of Cultured Human Hepatoma Cells Causes Apoptosis and Pyroptosis in Both Infected and Bystander Cells. Sci Rep. 2016;6:37433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 63]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
160.  Sonohara F, Inokawa Y, Kanda M, Nishikawa Y, Yamada S, Fujii T, Sugimoto H, Kodera Y, Nomoto S. Association of Inflammasome Components in Background Liver with Poor Prognosis After Curatively-resected Hepatocellular Carcinoma. Anticancer Res. 2017;37:293-300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
161.  Scholz A, Plate KH, Reiss Y. Angiopoietin-2: a multifaceted cytokine that functions in both angiogenesis and inflammation. Ann N Y Acad Sci. 2015;1347:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 162]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
162.  Faillaci F, Marzi L, Critelli R, Milosa F, Schepis F, Turola E, Andreani S, Vandelli G, Bernabucci V, Lei B. Liver Angiopoietin-2 is a key predictor of de novo or recurrent hepatocellular cancer after HCV direct-acting antivirals. Hepatology. 2018;68:1010-1024.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 90]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
163.  Ogura Y, Hori T, Uemoto S. Intentional portal pressure control is key to improving the outcome of living donor liver transplantation: the Kyoto University Hospital experience. Clin Transpl. 2008;143-149.  [PubMed]  [DOI]  [Cited in This Article: ]
164.  Hidaka M, Takatsuki M, Soyama A, Tanaka T, Muraoka I, Hara T, Kuroki T, Kanematsu T, Eguchi S. Intraoperative portal venous pressure and long-term outcome after curative resection for hepatocellular carcinoma. Br J Surg. 2012;99:1284-1289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
165.  Hernandez-Gea V, Turon F, Berzigotti A, Villanueva A. Management of small hepatocellular carcinoma in cirrhosis: focus on portal hypertension. World J Gastroenterol. 2013;19:1193-1199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
166.  Waziry R, Hajarizadeh B, Grebely J, Amin J, Law M, Danta M, George J, Dore GJ. Hepatocellular carcinoma risk following direct-acting antiviral HCV therapy: A systematic review, meta-analyses, and meta-regression. J Hepatol. 2017;67:1204-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 343]  [Article Influence: 49.0]  [Reference Citation Analysis (0)]
167.  Cancer Genome Atlas Research Network. Cancer Genome Atlas Research Network. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell. 2017;169:1327-1341.e23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1175]  [Cited by in F6Publishing: 1608]  [Article Influence: 229.7]  [Reference Citation Analysis (1)]
168.  Chiang DY, Villanueva A. Progress towards molecular patient stratification of hepatocellular carcinoma: Lost in translation? J Hepatol. 2017;67:893-895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]