Skip to main content
Hepatic Oncology logoLink to Hepatic Oncology
. 2015 Jul 27;2(3):291–302. doi: 10.2217/hep.15.16

Current issues on genomic heterogeneity in hepatocellular carcinoma and its implication in clinical practice

Kornelius Schulze 1,1,2,2,3,3,4,4, Jessica Zucman-Rossi 1,1,2,2,3,3,4,4,5,5,*
PMCID: PMC6095162  PMID: 30191009

Abstract

Hepatocellular carcinoma (HCC) is a highly heterogeneous disease leading to a major diversity. Since staging systems are used in patient care, molecular and histopathological features remain to be incorporated in management algorithms. HCC, as other malignant solid tumors, exhibit a complex genetic diversity and genomic instability, driving tumorigenesis. The recent development of deep sequencing techniques has revealed different subgroups of tumors defined by specific patterns of genomic alterations that are related to clinical and histopathological diversity in HCC. Additionally, several genomic defects identified in HCC will be used in the future to develop clinical trial design for tumorized treatment.

KEYWORDS : ARID1A, CTNNB1, genomic heterogeneity, hepatocellular carcinoma, liver cancer, mutational signatures, next-generation sequencing, TERT, TP53, transcriptomic groups


Practice points.

  • Hepatocellular carcinoma (HCC) is a highly heterogeneous disease at the clinical level that can be linked to histological phenotypic and molecular diversity.

  • Immunohistochemical and molecular markers, related to prognosis and different pathways of carcinogenesis, have not found their way into treatment management.

  • Tumor initiation and progression result from accumulated alterations (copy-number alterations and mutations) in oncogenic drivers and pathways of carcinogenesis.

  • Ten mutational signatures (signatures 1A, 1B, 4, 5, 6, 12, 16, 17, 23, 24) are described in HCC.

  • Transcriptomic groups are associated with clinical and epi/genetic/genomic features.

  • Several molecular profiles have been introduced to potentially predict outcome and improve clinical trial design for targeted treatment.

  • The knowledge of molecular intratumor heterogeneity is limited and additional deep sequencing and transcriptome analyses of large HCC series are needed.

  • A biopsy-based integrative diagnostic approach including morphology, immunohistochemistry, transcriptomic data, mutational profiles, CNA and methylome analysis could be suitable in the future.

  • Multiple-site biopsies, considering intratumor heterogeneity, could refine this process and need to be discussed.

Hepatocellular carcinoma (HCC) represents the sixth most common cancer (fifth in men and ninth in women) with 782,000 new cases worldwide in 2012. It is the second leading cause of cancer-related mortality, and increases steadily in age-standardized death rates since 1990 [1,2]. HCC is a highly heterogeneous disease at the clinical level and this heterogeneity can be related to the histological phenotypic and molecular diversity (Figure 1). However, HCC is one of the only malignant tumors, for which in the vast majority of cases, no histological proof is mandatory for diagnosis and initiation of cancer-specific treatment, following the clinical practice guidelines of the EASL, AASLD and APASL to diagnose lesions in the background of cirrhosis or chronic hepatitis B virus (HBV) infection [3–6]. In spite of several histopathological, specific immunohistochemical and molecular marker, which have been identified as closely related to prognosis and different pathways of carcinogenesis, incorporation of these markers into treatment management has not taken place.

Figure 1. . Inter- and intra-tumor heterogeneity.

Figure 1. 

Hepatocellular carcinoma is a highly heterogeneous disease dependent on geographical origin, risk factors, underlying liver disease, and the disease stage. This clinical heterogeneity is associated to molecular diversity (CNA) leading in part to histopathological diversity. Additionally, intratumor heterogeneity is present and a biopsy-based integrative diagnostic approach including histopathology, transcriptomic classification, mutational profiling, CNA, epigenetic profiling and calculation of prognostic gene scores could improve patient management in terms of tumorized treatment.

CNA: Copy number alteration.

Adapted with permission from Globocan [2].

In times when new personalized treatment options hold the capacity to elevate the level of patient care, new techniques, such as next-generation sequencing (NGS) and large genomic databases (ICGC, TCGA), promise to link tumor genomic profiling with a more individualized management of patients [7–22]. In HCC, several clinical trials for novel molecular treatments have failed in recent years and this may be related to the vast molecular diversity of HCC and the lack of tumor sampling, leading to unselective patient enrollment. In contrast, clinical trial design integrating clinical, histopathological up to specific molecular information of HCC samples could pave the way to personalized treatment and tumor biopsies could lead to specific tumorized treatment.

Clinical-pathological heterogeneity

• Clinical heterogeneity & patient management

HCC is the most frequent primary cancer of the liver (85%) and predominantly effects men at a ratio of 2–4:1 (without cirrhosis approximately 1:1) [23]. At diagnosis, clinical characteristics, such as risk factors, age, local and regional tumor burden, liver function and clinical performance status, strongly vary in accordance to the geographical origin and to the socioeconomic status of patients (Figure 1). Age at diagnosis is indicated to peak at the age of 70 years and to increase up to 70–79 years in Japan, whereas in central Africa and China patients are younger [3,24–25]. The predominant risk factor in high-incidence regions such as China, Southeastern Asia and Sub-Saharan Africa is chronic HBV infection with or without exposure to AFB1 [23,26]. Chronic hepatitis C virus (HCV) infection is the major risk factor in Japan, where it is diagnosed in 80–90% of the cases [27]. In Europe and North America chronic HCV infection is accompanied by significant alcohol abuse as the most prevalent risk factors [28]. Of note, nonalcoholic steatohepatitis (NASH)-associated HCC is rapidly increasing in numbers in North America [29]. In general, liver cancer is mainly found in less developed regions where over 80% of cases occur [2]. Underlying chronic liver disease occurs in up to 90% and HCC is the most frequent cause of death in patients with cirrhosis [30–32]. Nevertheless, 10% of patients with HCC present without cirrhosis and/or unknown risk factors. To that regard, since cirrhosis is a common co-morbidity, the disease course and overall survival are co-determined by the level of liver impairment.

To summarize, HCC patients present a highly heterogenic clinical background at diagnosis and applying different staging systems, such as Barcelona Clinic Liver Cancer (BCLC) and the Hong Kong Liver Cancer (HKLC), lead to inconsistent prediction of treatment response, disease course and overall prognosis [33]. Furthermore, the implementation of staging systems in clinical practice and a different patient spectrum in Europe/North America (BCLC) and Asia (HKLC) will result in a heterogenic stratification of patients around the world. Profiling of tumor genomic heterogeneity, leading to histopathological heterogeneity and eventually determine the phenotypical heterogeneity, could therefore refine clinical management.

• Histopathological heterogeneity

Since the diagnosis of HCC is achieved by noninvasive criteria in the majority of cases, regular tumor biopsy is not recommended by international guidelines [3–6]. Therefore, 60–70% of nonresectable tumor samples are treated without HCC tissue confirmation leading to a general lack of tissue for precise characterization [34]. However, when imaging is atypical and aiming to distinguish HCC from benign tumors, three immunohistochemical markers, GPC3, HSP70 and GS are recommended by the international consensus group of hepatocellular neoplasia for tumor-confirmation [35–37]. Whereas some entities simply demonstrate differences in proliferation and differentiation, other HCC present distinct morphological types. For instance, fibrolamellar HCC (FLC) is a very specific morphological subtype of HCC [38–40]. Additional accepted histological patterns are scirrhous, sarcomatoid, inflammatory, or lympho-epithelial-like HCC. Furthermore, up to 5% of HCC samples contain distinct portions of intrahepatic cholangiocarcinoma (ICC) and are referred to as HCC/ICC [41].

Recently, several studies have investigated the clinical impact of immunohistochemical markers within multiple subtypes. Yamashita et al. have demonstrated that an EpCAM+/AFP+ subtype of HCC, representing a high proportion of liver tumor-initiating cells (TIC), exhibits increased tumorigeneity and higher invasiveness [42–44]. In the context of liver TIC, various associated immunohistochemical markers have been introduced such as CK19, CD13, CD24, CD44, CD90 and CD133 [45–53]. Histopathological features of microscopic vascular invasion are independent prognostic factors for progression-free and overall survival in patients with HCC [54,55]. Additionally, it is indicated that the tumor microenvironment could influence tumor characteristics and that the composition of tumor-infiltrating leukocytes, hepatic stellate cells, endothelial cells, hepatoma cells and cancer-associated fibroblasts (CAF) are linked to the histopathological heterogeneity of HCC [56,57].

Recently, further attention was attracted to identify intratumor histopathological heterogeneity [58]. Initially, intratumor heterogeneity in HCC was reported by Kenmochi et al. and An et al., who have demonstrated intratumor variations in differentiation and proliferation in two thirds and in up to half of HCC tissue samples, respectively [59,60]. Taking into account morphological and immunohistochemical features Friemel et al. demonstrated intratumor heterogeneity in at least one immunohistochemical characteristic in 87% of cases [58]. In addition, 50% of their HCC samples presented different expression patterns of CK7 and GS within one tumor [58].

Alongside the role of the tumor microenvironment, inter- and intratumor histopathological variability are linked to driver mutations in regards to clonal evolution and to the concept of stem cell-like TIC [56,61–63]. With recent advances in NGS light has been shed on the issue of cancer driver genes and associated oncogenic pathways.

Molecular heterogeneity

• Intertumor heterogeneity

Tumor initiation and progression result from accumulated alterations in oncogenic drivers and pathways finally leading through clonal evolution to intertumor molecular heterogeneity (Table 1) [7–13,15–17,22,64]. Several differences in alteration rates and activation of oncogenic pathways are assumably dependent on heterogeneous clinical characteristics (risk factors, underlying liver disease, or disease stage). Therefore, results of original papers depend very much on the tissue they have included in their studies.

Table 1. . Alterations in oncogenic drivers and major oncogenic pathways.

Study (year) Sample size Sequencing technique Most frequently altered cancer driver Most frequently altered oncogenic pathway Major risk factors Ref.
Fujimoto et al. (2012) 27 + 120 Whole genome + validation set TP53 (52%)   HCV (52%) [9]
      ATM (19%)   HBV (41%)  
      IGSF10 (15%)      
      CTNNB1 (11%)      
      ARID1A (11%)      

Guichard et al. (2012) 24 + 125 Whole exome + validation set CTNNB1 (33%) Wnt pathway (49%) Alcohol (37%) [8]
      TP53 (21%) p53 pathway (33%) HBV (24%)  
      ARID1A (17%) Chromatin remodeling (23%) HCV (19%)  
      AXIN1 (15%) PI3K/Ras pathway (13%) NASH (5%)  
      RPS6KA3 (10%) Oxidative stress (6%)    
      CDKN2A (7%)      

Huang et al. (2012) 10 + 100 Whole exome + validation set TP53 (27%)   HBV (100%) [7]
      ARID1A (13%)      
      SAMD9L (6%)      
      ARID2 (4%)      

Cleary et al. (2013) 87 Whole exome CTNNB1 (23%)   HBV (43%) [16]
      TP53 (20%)   HCV (21%)  
      CPA2 (9%)   Alcohol (11%)  
      IGSF3 (9%)      
      KEAP1 (8%)      

Kan et al. (2013) 88 Whole genome TP53 (35%) Wnt pathway (63%) HBV (92%) [15]
      CTNNB1 (16%) JAK/STAT pathway (46%)    
      LRP1B (11%) Apoptosis pathway (46%)    
      JAK1 (9%) p53 pathway (43%)    
      AXIN1 (5%)      

Ahn et al. (2014) 231 Whole exome TP53 (32%) p53 pathway (37%) HBV (72%) [64]
      CTNNB1 (23%) Wnt pathway (37%) HCV (10%)  
      RB1 (8%) Chromatin remodeling (34%)    
      AXIN1 (7%) Cell cycle (22%)    
      SELPLG (5%) PI3K/Ras pathway (12%)    
      FGF19 (5%)      

Totoki et al. (2014) 608 Whole genome + whole exome TERT (54%) p53 pathway (72%) HCV (42%) [11]
      CTNNB1 (31%) Telomere maintenance (68%) HBV (23%)  
      TP53 (31%) Chromatin remodeling (67%)    
      ARID1A (8%) Wnt pathway (66%)    
      AXIN1 (6%) PI3K/mTOR pathway (45%)    
      TSC2 (5%) Oxidative stress (19%)    

Schulze et al. (2015) 235 Whole exome TERT (60%) Telomere maintenance (60%) Alcohol (41%) [22]
      CTNNB1 (37%) Wnt pathway (54%) HCV (26%)  
      TP53 (24%) PI3K/mTOR pathway (51%) NASH (18%)  
      ARID1A (13%) p53 pathway (49%) HBV (14%)  
      ALB (13%) MAP kinase pathway (43%)    
      AXIN1 (11%) Hepatic differentiation (34%)    
      CDKN2A (9%) Epigenetic regulation (32%)    
        Chromatin remodeling (28%)    

HBV: Hepatitis B virus; HCV: Hepatitis C virus; NASH: Nonalcoholic steatohepatitis

Overall, activating TERT alterations (promoter mutation, focal amplification, or HBV insertion) are the most frequent alterations occurring in HCC [11,18–20,22]. Moreover, TERT promoter mutations are the earliest genetic alterations occurring during malignant transformation of cirrhotic nodules in HCC or during hepatic adenoma-carcinoma transition in association with CTNNB1 mutations [11,18–20]. Recurrent activating mutations of CTNNB1 (11–37%) are the second most common alterations observed in HCC (Table 1); more frequent in HBV- and alcohol-related tumors [8,11,15,22,64]. Additionally, recurrent inactivating mutations of β-catenin inhibitors, AXIN1, APC or ZNRF3, lead to an activating Wnt signaling in up to 66% of all liver cancers (Table 1) [11,22]. Inactivating mutations in TP53 are also highly frequent (20–52%), more predominantly in HBV-related HCC and associated with poor prognosis [8–9,11,15,22,64]. Beyond that, characteristic R249S somatic mutations in TP53 are linked to AFB1 exposure and are clustered in HBV infected patients in high endemic regions such as Sub-Saharan Africa [22]. Notably, alterations in CTNNB1 and TP53 are significantly exclusive from each other; they define two different clusters of HCC [22]. Several genes encoding chromatin remodeling factors are also frequently mutated in HCC. Among them, ARID1A and ARID2 are the genes most recurrently inactivated in HCC cohorts around the world (8–17%) [7–9,11,22]. Significant association to alcohol [22] or to non-HBV/non-HCV cases in Japan [11] is highlighted, implying an important tumor suppressor role of the SWI/SNF chromatin remodeling complex in nonviral HCC. Mutations in additional cancer drivers have been identified: ATM (up to 19%), ALB (up to 13%), RPS6KA3 (up to 10%), CDKN2A (up to 9%), IGSF3/10 (up to 9%), JAK1 (up to 9%), RB1 (up to 8%), KEAP1 (up to 8%), TSC2 (up to 5%), FGF19 (up to 5%) and ARID2 (up to 4%) (Table 1). From the clinical point of view, alcohol as a risk factor is further associated with alterations in SMARCA2, RB1 and homozygous deletions in the CDKN2A locus [22]. Alterations in IL6ST and less alterations in TERT are significantly found in cases without known etiologies [22].

Recurrent somatic copy number alterations (CNA), leading to chromosomal losses and gains, were identified at 1p, 4p-q, 6q, 8p, 13p-q, 16p-q, 17p, 21p-q, 22q and at 1q, 5p, 6p, 8q, 17q, 20q, Xq, respectively [8,11,15,22,64]. Moreover, frequent homozygous deletions inactivating AXIN1, CDKN2A/B, CFH, IRF2, MAP2K3, PTEN, PTPN3, RB1, RPS6KA3 were found, whereas recurrent focal amplifications were described associated with an overexpression of CCNE1, FGF3/4/19/CCND1, JAK3, MET, MYC, TERT and VEGFA [8,11,15,22,64]. Integrating CNA and frequent mutations in 161 putative cancer driver genes, pointed out frequent alterations of 11 major oncogenic pathways, including from the most to the least frequent, alterations in telomere maintenance, Wnt signaling, PI3K/mTOR pathway, p53 pathway, MAP kinase pathway, hepatic differentiation, epigenetic regulation, chromatin remodeling, oxidative stress, JAK/STAT pathway and TGF-β signaling (Table 1) [22]. Nevertheless, noticeable differences are observed among HCC cohorts, for example, alterations in the p53 pathway ranging from 33 to 72% and in PI3K/mTOR signaling ranging from 12 to 51%, reflecting heterogeneity related to geographical and risk factor distributions [11,22]. Along malignant transformation from dysplastic macronodules to poor prognosis HCC, we have found an increasing number of gene alterations and CNA per stage [22]. TERT promoter mutations were early events in hepatocarcinogenesis whereas focal amplifications of the FGF3/4/19/CCND1 locus were significantly associated with poor prognosis HCC [22]. Common alterations found in hepatocellular adenoma (HNF1a, IL6ST) have not been discovered in HCC in nonfibrotic liver tissue, implying that malignant transformation of hepatic adenomas is not frequent [22]. Nevertheless, the presence or absence of underlying liver disease may affect growth patterns.

Molecular diversity in HCC is also observed at the transcriptomic level, showing several subgroups of tumors characterized by a specific profile of gene expression [14,65–67]. Significant associations linked these transcriptomic subgroups with clinical and epi-/genetic/genomic features. The noteworthy integrative approach, including gene expression, genetic alterations and clinical features, remains to be performed in HCC related to risk-factor profiles. Moreover, thanks to multinational approaches HCC-subtypes have been molecularly classified to date, for example, three groups in a cohort of 78 FLC samples are reported as proliferative, inflammatory and unannotated subclass of FLC, taking into account multiple molecular technics [68].

Novel exciting genomic classifications are proposed by mutational signatures, linking exposure to risk factors to specific patterns of nucleotide mutations. As described by Alexandrov et al., a computational framework allows deciphering distinct patterns of somatic mutations in a set of cancer samples as mutational signatures based on 96 possible nucleotide triplets including the 3′ and 5′ nucleotides of substitution sites [14,69–71]. In a pancancer study, eight mutational signatures (signatures 1A, 1B, 4, 5, 6, 12, 16, 17) were described in HCC [70]. Associations for signatures 1A, 1B (age), 4 (tobacco) and 6 (defects in mismatch repair) are assumed, whereas for signatures 5, 12, 16 and 17 genotoxic causes are unknown. A recent study also found mutational signatures associated with ancestry groups (European, Japanese, and US Asian) without relation to specific risk factors (HBV, HCV, nonviral) [11]. Additional exome data demonstrated two novel mutational signatures (signatures 23 and 24) [22]. Signature 23 was identified in a woman presenting black deposition of mineral silica in the nontumor liver tissues suggesting exposition to an unidentified risk factor. Signature 24 was identified in patients from Africa, showing typical R249S mutation in TP53. Thus, this signature is characteristic of the AFB1 exposure [22].

• Intratumor heterogeneity

To understand the biological basis of intratumor heterogeneity, the evolutionary history of tumors has to be taken into account. Mutational signatures reflect the diversity of exogenous and endogenous impacts on genome stability occurring during cancer evolution, leading finally to the outgrowth of major clones and/or several subclones [70]. Moreover, intratumor heterogeneity can result of different modes of branched tumor evolution: during linear evolution mutagenic processes accumulate sequentially over time. In case of two genetically and geographically distinct minor clones within one tumor, branched cancer evolution can progress independently, synergistic, or antagonistic between subclones within one tumor [72].

Regarding HCC, our knowledge of molecular intratumor heterogeneity is limited [58,73] and we need additional data in large series of tumors analyzed using deep sequencing and transcriptome experiments. The knowledge of intratumor heterogeneity will be particularly important to interpret the results of resistance to treatment and identifying subclones selected by drugs.

Consequences for clinical practice

The fast growing knowledge about tumor genomic heterogeneity in HCC will lead to an individual molecular profiling. Therefore, like in other types of tumors, it is believed that precise molecular profiling will impact on clinical treatment decision for HCC patients.

• Molecular diversity according to prognosis

Several molecular signatures have been introduced to potentially predict outcome in patients with HCC (Table 2).

Table 2. . Molecular signatures potentially predicting outcome in patients with hepatocellular carcinoma.

Study (year) Signature Molecular feature Sample size Major risk factors Ref.
Villanueva et al. (2015) 36-DNA-methylation marker score Methylome 221 + 83 (validation) HBV, HCV, alcohol [74]

Wei et al. (2013) 30-miRNA signature miRNA 60 + 50 (validation) HBV [75]

Kim et al. (2012) 65-gene-based risk score mRNA 139 + 292 (validation) HBV [76]

Barry et al. (2012) 67-miRNA HCC recurrence signature miRNA 69 HCV, HBV, NASH [77]

Roessler et al. (2010) Metastasis gene signature mRNA 247 +139 (validation) HBV [78]

Lee et al. (2004) Proliferation cluster of genes mRNA 91 HBV, HCV, alcohol, hemochromatosis, Wilson disease [67]

Lee et al. (2006) Hepatic progenitor cell signature mRNA 139 HBV, HCV, alcohol [79]

Boyault et al. (2007) G1-6 transcriptome classification mRNA 120 HBV, HCV, alcohol [14]

Nault et al. (2013) Five-gene score mRNA 314 + 434 (validation) HBV, HCV, alcohol, hemochromatosis [21]

Yamashita et al. (2013) EpCAM+/CD90+ gene expression score mRNA 102 HBV, HCV, alcohol [80]

Yamashita et al. (2009) EpCAM+ gene expression score mRNA 156 HBV [42]

Hoshida et al. (2009) S1-3 expression-based subclasses mRNA 603 HBV, HCV [66]

Minguez et al. (2011) 35-gene signature of vascular invasion mRNA 79 + 135 (validation) HCV [81]

Toffanin et al. (2011) miRNA-based classification of HCC miRNA 89 HCV [82]

Kurokawa et al. (2004) 20-gene-based prediction score mRNA 100 HBV, HCV [83]

Iizuka et al. (2003) Statistical Pattern Recognition method mRNA 33 + 27 (validation) HCV, HBV [84]

Wang et al. (2007) 57-member molecular gene signature mRNA 25 HBV, HCV [85]

Woo et al. (2008) HCC recurrence signature mRNA 65 HBV [86]

van Malenstein et al. (2010) Seven-gene signature associated with hypoxia mRNA 272 + 91 (validation) HCV, HBV, alcohol, NASH, AIH, PBC [87]

Ye et al. (2003) Metastatic HCC gene signature mRNA 67 HBV [88]

Yoshioka et al. (2009) Prediction score of early recurrence mRNA 139 HCV, HBV [89]

Okamato et al. (2006) Gene signature for multicentric HCC mRNA 40 HCV [90]

Chiang et al. (2008) Molecular classification of HCV-associated HCC mRNA 109 HCV [91]

Jiang et al. (2008) miRNA prognostic signature miRNA 54 HCV, alcohol, HBV, hemochromatosis [92]

Budhu et al. (2008) 20-miRNA tumor signature miRNA 131 + 110 (validation) HBV [93]

Yamashita et al. demonstrated that gene expression profiles of stem cell-like TIC marker (EpCAM, CD90 and CD133) may determine the clinical outcome in patients with HCC [80]. Nault et al. introduced a five-gene mRNA (HN1, RAN, RAMP3, KRT19 and TAF9) score predicting disease-specific survival, independent of clinical or histopathological features, in 748 patients worldwide [21]. Macro- and micro-scopic vascular invasion are significant predictors for HCC recurrence after resection and it has been associated with a 35-gene-expression signature in 214 patients with HCC [81]. Ye et al. identified a molecular signature associated to intrahepatic metastases and poor outcome [88]. Another study investigated the role of gene signatures in development of metastases and identified a 161-gene signature that was validated in 386 HCC patients [78]. Finally, Okamato et al. introduced a 36-gene-expression profile predicting multicentric HCC in nontumor liver tissue with single or multiple tumor nodules [90].

Several groups analyzed miRNA patterns and their association to prognosis. Applying a 67-miRNA signature Barry et al. showed a significant association with HCC recurrence after liver transplantation in 64 patients [77]. Budhu et al. found a 20-miRNA tumor signature identifying patients who are at higher risk of developing metastases and predicting overall survival [93].

Recently, Villanueva et al. validated a 36-DNA-methylation signature and demonstrated accurate poor survival prediction in 304 patients with HCC [74]. Moreover, the 36-DNA-methylation signature is associated to an mRNA-based signature significantly indicating hepatic progenitor cells [74].

In conclusion, several promising signature models have been introduced and prospective studies have to validate the reproducibility in treatment stratification, for example, curative versus palliative approach.

• Targeted therapies adapted to the molecular diversity

Treatment options are limited in patients with HCC, particularly for patients in BCLC stage C, because until today sorafenib is the only approved systemic agent. In spite of a broad arsenal of small molecular inhibitors and monoclonal antibodies, multiple efforts of implementing new targeted treatments into clinical practice failed in recent years [94–101]. In part, inadequate patient selection and a ‘diluted treatment effect’ after unselective patient enrollment are thought to be the reasons. Hence, it is believed that future clinical trial designs need to implement the individual molecular profile to allocate patients to an adequate targeted treatment.

Recently, two large-scale NGS studies have schematized a broad panoramic view of putative cancer drivers and major pathways involved in hepatocarcinogenesis and pointed out potential therapeutic targets in accumulated 853 patients with HCC [11,22]. Molecular profiles (CNA and mutations) revealed 11 altered major pathways: telomere maintenance, WNT/β-catenin, PI3K/AKT/mTOR, TP53/cell cycle, MAPK, hepatic differentiation, epigenetic regulation, chromatin remodeling, oxidative stress, IL-6/JAK/STAT, and TGF-β. Within these pathways multiple oncogenes or tumor suppressor genes are potentially prone to targeted treatment. The French study reported that 28% of patients harbored at least one damaging alteration, potentially targetable by an FDA approved [22]. Moreover, 86% of the patients are potentially treatable by a drug, screened in clinical trials Phase I–III [22]. The most attractive target genes are involved in the PI3K/AKT/mTOR and MAPK pathways, since growth factor receptors and their ligands are well-suitable for tyrosine kinase inhibitors and antibodies. Only a small proportion of patients harbored alterations for a unique potential gene: FLTs (6%), FGF3/4/19 (4%), PDGFRs (3%), EPHA4 (3%), VEGFA (1%), HGF (3%), EGFR (1%), FGFRs (1%), KIT (1%), MET (1%), TEK (1%), ERBB2 (<1%), KDR (<1%) [22]. Hence, these findings underline the necessity of precise patient stratification for adequate clinical trial design. Despite frequent activating alterations of the PI3K/AKT/mTOR pathway (51%) and mutations of MTOR (2%), the mTOR inhibitor everolimus also failed in a Phase III trials as second line treatment [22,97]. Described by Sawey et al., focal amplification of the FGF19/3/4/CCND1 locus is an effective predictor for anti-FGF19 treatment in HCC [102]. Subsequent high-throughput sequencing studies found amplifications of FGF19 in up to 6% in human HCC samples promising targeted treatment in a subgroup of patients with HCC [11,22,64]. Of note, one study reported FGF19 amplifications are associated to poor prognosis patients, implying poor prognosis patients also for the control arm in clinical trial design [22]. At the end of the day, direct linkage of tumor genomic profiling with individualized patient management is far more complex. Extensive biological and pharmacological studies will be needed to understand how driver genes can be efficiently targeted and how to overcome resistance. Clinical trial design needs to integrate clinical, histopathological and specific molecular information of HCC samples to pave the way to personalized treatment. Moreover, taken into account intratumor heterogeneity, additional multiple-site biopsies could lead to specific tumorized treatment. Nevertheless, current data are from resection specimens. Therefore, further prospective studies, including tissue samples of biopsies, will answer these important questions in the future.

Conclusion & future perspective

Up until today, stratified molecular information has not been incorporated into staging systems utilized in HCC. A biopsy-based integrative diagnostic approach, including morphology, immunohistochemistry/-fluorescence, transcriptomic expression data, mutational profiles, CNA and methylome analyses, could be suitable in the future (Figure 1). Moreover, multiple-site biopsies, considering intratumor heterogeneity, could refine this process and need to be debated (Figure 1). Prospectively, this valuable knowledge could improve patient stratification for adequate enrollment in clinical trials.

Footnotes

Financial & competing interests disclosure

KS is supported by the Deutsche Forschungsgemeinschaft (DFG Grant Number: SCHU 2893/2-1). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as: • of interest; •• of considerable interest

  • 1.GBD 2013 Mortality and Causes of Death Collaborators. Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the global burden of disease study 2013. Lancet. 2014;385(9963):117–71. doi: 10.1016/S0140-6736(14)61682-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ferlay J, Soerjomataram I, Ervik M, et al. International Agency for Research on Cancer; Lyon, France: 2013. http://globocan.iarc.fr GLOBOCAN 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC CancerBase No. 11. [Google Scholar]
  • 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(4):908–943. doi: 10.1016/j.jhep.2011.12.001. [DOI] [PubMed] [Google Scholar]
  • 4.Bruix J, Sherman M. Practice Guidelines Committee, American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma. Hepatology. 2005;42(5):1208–1236. doi: 10.1002/hep.20933. [DOI] [PubMed] [Google Scholar]
  • 5.Bruix J, Sherman M. American Association for the Study of Liver Diseases. Management of hepatocellular carcinoma: an update. Hepatology. 2011;53(3):1020–1022. doi: 10.1002/hep.24199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Omata M, Lesmana LA, Tateishi R, et al. Asian Pacific Association for the Study of the Liver Consensus Recommendations on Hepatocellular Carcinoma. Hepatol. Int. 2010;4(2):439–474. doi: 10.1007/s12072-010-9165-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Huang J, Deng Q, Wang Q, et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat. Genet. 2012;44(10):1117–1121. doi: 10.1038/ng.2391. [DOI] [PubMed] [Google Scholar]
  • 8.Guichard C, Amaddeo G, Imbeaud S, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 2012;44(6):694–698. doi: 10.1038/ng.2256. [DOI] [PMC free article] [PubMed] [Google Scholar]; • Emphasizes important cancer drivers recurrently altered in hepatocellular carcinoma and highlights major pathways involved in hepatocarcinogenesis.
  • 9.Fujimoto A, Totoki Y, Abe T, et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 2012;44(7):760–764. doi: 10.1038/ng.2291. [DOI] [PubMed] [Google Scholar]
  • 10.Totoki Y, Tatsuno K, Yamamoto S, et al. High-resolution characterization of a hepatocellular carcinoma genome. Nat. Genet. 2011;43(5):464–469. doi: 10.1038/ng.804. [DOI] [PubMed] [Google Scholar]
  • 11.Totoki Y, Tatsuno K, Covington KR, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 2014;46(12):1267–1273. doi: 10.1038/ng.3126. [DOI] [PubMed] [Google Scholar]; •• This is a large-scale study introducing mutational signatures associated to ancestry groups without relation to risk factors (HBV, HCV, nonviral).
  • 12.Nault JC, Zucman-Rossi J. Genetics of hepatobiliary carcinogenesis. Semin. Liver Dis. 2011;31(2):173–187. doi: 10.1055/s-0031-1276646. [DOI] [PubMed] [Google Scholar]
  • 13.Nault JC, Zucman-Rossi J. Genetics of hepatocellular carcinoma: the next generation. J. Hepatol. 2014;60(1):224–226. doi: 10.1016/j.jhep.2013.08.019. [DOI] [PubMed] [Google Scholar]
  • 14.Boyault S, Rickman DS, De Reynies A, et al. Transcriptome classification of HCC is related to gene alterations and to new therapeutic targets. Hepatology. 2007;45(1):42–52. doi: 10.1002/hep.21467. [DOI] [PubMed] [Google Scholar]; • Introduces six transcriptomic subgroups significantly associated with clinical and genetic/genomic features.
  • 15.Kan Z, Zheng H, Liu X, et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res. 2013;23(9):1422–1433. doi: 10.1101/gr.154492.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cleary SP, Jeck WR, Zhao X, et al. Identification of driver genes in hepatocellular carcinoma by exome sequencing. Hepatology. 2013;58(5):1693–1702. doi: 10.1002/hep.26540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sung WK, Zheng H, Li S, et al. Genome-wide survey of recurrent HBV integration in hepatocellular carcinoma. Nat. Genet. 2012;44(7):765–769. doi: 10.1038/ng.2295. [DOI] [PubMed] [Google Scholar]
  • 18.Pilati C, Letouze E, Nault JC, et al. Genomic profiling of hepatocellular adenomas reveals recurrent frk-activating mutations and the mechanisms of malignant transformation. Cancer Cell. 2014;25(4):428–441. doi: 10.1016/j.ccr.2014.03.005. [DOI] [PubMed] [Google Scholar]
  • 19.Nault JC, Mallet M, Pilati C, et al. High frequency of telomerase reverse-transcriptase promoter somatic mutations in hepatocellular carcinoma and preneoplastic lesions. Nat. Commun. 2013;4:2218. doi: 10.1038/ncomms3218. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This is the first study to demonstrate that TERT promoter mutations are the most frequent and earliest somatic alterations during malignant transformation in hepatocellular carcinoma.
  • 20.Nault JC, Calderaro J, Di Tommaso L, et al. Telomerase reverse transcriptase promoter mutation is an early somatic genetic alteration in the transformation of premalignant nodules in hepatocellular carcinoma on cirrhosis. Hepatology. 2014;60(6):1983–1992. doi: 10.1002/hep.27372. [DOI] [PubMed] [Google Scholar]; • Emphasizes that TERT promoter mutations are an early step in hepatocarcinogenesis.
  • 21.Nault JC, De Reynies A, Villanueva A, et al. A hepatocellular carcinoma 5-gene score associated with survival of patients after liver resection. Gastroenterology. 2013;145(1):176–187. doi: 10.1053/j.gastro.2013.03.051. [DOI] [PubMed] [Google Scholar]; •• Introduces the five-gene mRNA score, which accurately predicts the prognosis after liver resection.
  • 22.Schulze K, Imbeaud S, Letouzé E, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 2015;47(5):505–511. doi: 10.1038/ng.3252. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This is a large-scale study identifying novel mutational signatures and schematizing a panoramic view of putative cancer drivers and major pathways involved in hepatocarcinogenesis, and points out potential therapeutic targets.
  • 23.El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132(7):2557–2576. doi: 10.1053/j.gastro.2007.04.061. [DOI] [PubMed] [Google Scholar]
  • 24.El-Serag HB. Hepatocellular carcinoma. N. Engl. J. Med. 2011;365(12):1118–1127. doi: 10.1056/NEJMra1001683. [DOI] [PubMed] [Google Scholar]
  • 25.Tanaka H, Imai Y, Hiramatsu N, et al. Declining incidence of hepatocellular carcinoma in Osaka, Japan, from 1990 to 2003. Ann. Intern. Med. 2008;148(11):820–826. doi: 10.7326/0003-4819-148-11-200806030-00004. [DOI] [PubMed] [Google Scholar]
  • 26.Chen JG, Zhang SW. Liver cancer epidemic in China: past, present and future. Semin. Cancer Biol. 2011;21(1):59–69. doi: 10.1016/j.semcancer.2010.11.002. [DOI] [PubMed] [Google Scholar]
  • 27.Nordenstedt H, White DL, El-Serag HB. The changing pattern of epidemiology in hepatocellular carcinoma. Dig. Liver Dis. 2010;42(Suppl. 3):S206–S214. doi: 10.1016/S1590-8658(10)60507-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Forner A, Llovet JM, Bruix J. Hepatocellular carcinoma. Lancet. 2012;379(9822):1245–1255. doi: 10.1016/S0140-6736(11)61347-0. [DOI] [PubMed] [Google Scholar]
  • 29.Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 2013;10(11):656–665. doi: 10.1038/nrgastro.2013.183. [DOI] [PubMed] [Google Scholar]
  • 30.Nault JC. Pathogenesis of hepatocellular carcinoma according to aetiology. Best Pract. Res. Clin. Gastroenterol. 2014;28(5):937–947. doi: 10.1016/j.bpg.2014.08.006. [DOI] [PubMed] [Google Scholar]
  • 31.Alazawi W, Cunningham M, Dearden J, Foster GR. Systematic review: outcome of compensated cirrhosis due to chronic hepatitis c infection. Aliment. Pharmacol. Ther. 2010;32(3):344–355. doi: 10.1111/j.1365-2036.2010.04370.x. [DOI] [PubMed] [Google Scholar]
  • 32.Sherman M. Hepatocellular carcinoma: epidemiology, surveillance, and diagnosis. Semin. Liver Dis. 2010;30(1):3–16. doi: 10.1055/s-0030-1247128. [DOI] [PubMed] [Google Scholar]
  • 33.Yau T, Tang VY, Yao TJ, Fan ST, Lo CM, Poon RT. Development of Hong Kong liver cancer staging system with treatment stratification for patients with hepatocellular carcinoma. Gastroenterology. 2014;146(7):1691–1700. e1693. doi: 10.1053/j.gastro.2014.02.032. [DOI] [PubMed] [Google Scholar]
  • 34.Torbenson M, Schirmacher P. Liver cancer biopsy – back to the future?! Hepatology. 2015;61(2):431–433. doi: 10.1002/hep.27545. [DOI] [PubMed] [Google Scholar]
  • 35.International Consensus Group for Hepatocellular Neoplasiathe International Consensus Group for Hepatocellular N. Pathologic diagnosis of early hepatocellular carcinoma: a report of the International Consensus Group for Hepatocellular Neoplasia. Hepatology. 2009;49(2):658–664. doi: 10.1002/hep.22709. [DOI] [PubMed] [Google Scholar]
  • 36.Di Tommaso L, Franchi G, Park YN, et al. Diagnostic value of HSP70, glypican 3, and glutamine synthetase in hepatocellular nodules in cirrhosis. Hepatology. 2007;45(3):725–734. doi: 10.1002/hep.21531. [DOI] [PubMed] [Google Scholar]
  • 37.Di Tommaso L, Destro A, Seok JY, et al. The application of markers (HSP70 GPC3 and GS) in liver biopsies is useful for detection of hepatocellular carcinoma. J. Hepatol. 2009;50(4):746–754. doi: 10.1016/j.jhep.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 38.Torbenson M. Fibrolamellar carcinoma: 2012 update. Scientifica (Cairo) 2012:743790. doi: 10.6064/2012/743790. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Edmondson HA. Differential diagnosis of tumors and tumor-like lesions of liver in infancy and childhood. AMA J. Dis. Child. 1956;91(2):168–186. doi: 10.1001/archpedi.1956.02060020170015. [DOI] [PubMed] [Google Scholar]
  • 40.Torbenson M. Review of the clinicopathologic features of fibrolamellar carcinoma. Adv. Anat. Pathol. 2007;14(3):217–223. doi: 10.1097/PAP.0b013e3180504913. [DOI] [PubMed] [Google Scholar]
  • 41.Tickoo SK, Zee SY, Obiekwe S, et al. Combined hepatocellular-cholangiocarcinoma: a histopathologic, immunohistochemical, and in situ hybridization study. Am. J. Surg. Pathol. 2002;26(8):989–997. doi: 10.1097/00000478-200208000-00003. [DOI] [PubMed] [Google Scholar]
  • 42.Yamashita T, Ji J, Budhu A, et al. Epcam-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features. Gastroenterology. 2009;136(3):1012–1024. doi: 10.1053/j.gastro.2008.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yamashita T, Forgues M, Wang W, et al. Epcam and alpha-fetoprotein expression defines novel prognostic subtypes of hepatocellular carcinoma. Cancer Res. 2008;68(5):1451–1461. doi: 10.1158/0008-5472.CAN-07-6013. [DOI] [PubMed] [Google Scholar]
  • 44.Zeng SS, Yamashita T, Kondo M, et al. The transcription factor SALL4 regulates stemness of epcam-positive hepatocellular carcinoma. J. Hepatol. 2014;60(1):127–134. doi: 10.1016/j.jhep.2013.08.024. [DOI] [PubMed] [Google Scholar]
  • 45.Kim H, Park YN. Hepatocellular carcinomas expressing ‘stemness’-related markers: clinicopathological characteristics. Dig. Dis. 2014;32(6):778–785. doi: 10.1159/000368021. [DOI] [PubMed] [Google Scholar]
  • 46.Hou Y, Zou Q, Ge R, Shen F, Wang Y. The critical role of CD133(+)CD44(+/high) tumor cells in hematogenous metastasis of liver cancers. Cell Res. 2012;22(1):259–272. doi: 10.1038/cr.2011.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tang KH, Ma S, Lee TK, et al. CD133(+) liver tumor-initiating cells promote tumor angiogenesis, growth, and self-renewal through neurotensin/interleukin-8/CXCl1 signaling. Hepatology. 2012;55(3):807–820. doi: 10.1002/hep.24739. [DOI] [PubMed] [Google Scholar]
  • 48.Lee TK, Castilho A, Cheung VC, Tang KH, Ma S, Ng IO. CD24(+) liver tumor-initiating cells drive self-renewal and tumor initiation through STAT3-mediated NANOG regulation. Cell Stem Cell. 2011;9(1):50–63. doi: 10.1016/j.stem.2011.06.005. [DOI] [PubMed] [Google Scholar]
  • 49.Zhu Z, Hao X, Yan M, et al. Cancer stem/progenitor cells are highly enriched in CD133+CD44+ population in hepatocellular carcinoma. Int. J. Cancer. 2010;126(9):2067–2078. doi: 10.1002/ijc.24868. [DOI] [PubMed] [Google Scholar]
  • 50.Ma S, Chan KW, Hu L, et al. Identification and characterization of tumorigenic liver cancer stem/progenitor cells. Gastroenterology. 2007;132(7):2542–2556. doi: 10.1053/j.gastro.2007.04.025. [DOI] [PubMed] [Google Scholar]
  • 51.Yang ZF, Ho DW, Ng MN, et al. Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell. 2008;13(2):153–166. doi: 10.1016/j.ccr.2008.01.013. [DOI] [PubMed] [Google Scholar]
  • 52.Yang ZF, Ngai P, Ho DW, et al. Identification of local and circulating cancer stem cells in human liver cancer. Hepatology. 2008;47(3):919–928. doi: 10.1002/hep.22082. [DOI] [PubMed] [Google Scholar]
  • 53.Haraguchi N, Ishii H, Mimori K, et al. CD13 is a therapeutic target in human liver cancer stem cells. J. Clin. Invest. 2010;120(9):3326–3339. doi: 10.1172/JCI42550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Du M, Chen L, Zhao J, et al. Microvascular invasion (MVI) is a poorer prognostic predictor for small hepatocellular carcinoma. BMC Cancer. 2014;14:38. doi: 10.1186/1471-2407-14-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Roayaie S, Blume IN, Thung SN, et al. A system of classifying microvascular invasion to predict outcome after resection in patients with hepatocellular carcinoma. Gastroenterology. 2009;137(3):850–855. doi: 10.1053/j.gastro.2009.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hernandez-Gea V, Toffanin S, Friedman SL, Llovet JM. Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology. 2013;144(3):512–527. doi: 10.1053/j.gastro.2013.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Leonardi GC, Candido S, Cervello M, et al. The tumor microenvironment in hepatocellular carcinoma (review) Int. J. Oncol. 2012;40(6):1733–1747. doi: 10.3892/ijo.2012.1408. [DOI] [PubMed] [Google Scholar]
  • 58.Friemel J, Rechsteiner MP, Frick L, et al. Intratumor heterogeneity in hepatocellular carcinoma. Clin. Cancer Res. 2015;21(8):1951–1961. doi: 10.1158/1078-0432.CCR-14-0122. [DOI] [PubMed] [Google Scholar]
  • 59.Kenmochi K, Sugihara S, Kojiro M. Relationship of histologic grade of hepatocellular carcinoma (HCC) to tumor size, and demonstration of tumor cells of multiple different grades in single small HCC. Liver. 1987;7(1):18–26. doi: 10.1111/j.1600-0676.1987.tb00310.x. [DOI] [PubMed] [Google Scholar]
  • 60.An FQ, Matsuda M, Fujii H, et al. Tumor heterogeneity in small hepatocellular carcinoma: analysis of tumor cell proliferation, expression and mutation of p53 and beta-catenin. Int. J. Cancer. 2001;93(4):468–474. doi: 10.1002/ijc.1367. [DOI] [PubMed] [Google Scholar]
  • 61.Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat. Rev. Cancer. 2012;12(5):323–334. doi: 10.1038/nrc3261. [DOI] [PubMed] [Google Scholar]
  • 62.Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012;366(10):883–892. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fidler IJ. Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Res. 1978;38(9):2651–2660. [PubMed] [Google Scholar]
  • 64.Ahn SM, Jang SJ, Shim JH, et al. Genomic portrait of resectable hepatocellular carcinomas: implications of RB1 and FGF19 aberrations for patient stratification. Hepatology. 2014;60(6):1972–1982. doi: 10.1002/hep.27198. [DOI] [PubMed] [Google Scholar]
  • 65.Budhu A, Roessler S, Zhao X, et al. Integrated metabolite and gene expression profiles identify lipid biomarkers associated with progression of hepatocellular carcinoma and patient outcomes. Gastroenterology. 2013;144(5):1066–1075. e1061. doi: 10.1053/j.gastro.2013.01.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hoshida Y, Nijman SM, Kobayashi M, et al. Integrative transcriptome analysis reveals common molecular subclasses of human hepatocellular carcinoma. Cancer Res. 2009;69(18):7385–7392. doi: 10.1158/0008-5472.CAN-09-1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lee JS, Chu IS, Heo J, et al. Classification and prediction of survival in hepatocellular carcinoma by gene expression profiling. Hepatology. 2004;40(3):667–676. doi: 10.1002/hep.20375. [DOI] [PubMed] [Google Scholar]
  • 68.Cornella H, Alsinet C, Sayols S, et al. Unique genomic profile of fibrolamellar hepatocellular carcinoma. Gastroenterology. 2014;148(4) doi: 10.1053/j.gastro.2014.12.028. 806–18e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Alexandrov LB, Stratton MR. Mutational signatures: the patterns of somatic mutations hidden in cancer genomes. Curr. Opin. Genet. Dev. 2014;24:52–60. doi: 10.1016/j.gde.2013.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Alexandrov LB, Nik-Zainal S, Wedge DC, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This is the major study introducing mutational signatures in over 7000 cancers.
  • 71.Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ, Stratton MR. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 2013;3(1):246–259. doi: 10.1016/j.celrep.2012.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mcgranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015;27(1):15–26. doi: 10.1016/j.ccell.2014.12.001. [DOI] [PubMed] [Google Scholar]
  • 73.Saeki R, Nagai H, Kaneko S, et al. Intratumoral genomic heterogeneity in human hepatocellular carcinoma detected by restriction landmark genomic scanning. J. Hepatol. 2000;33(1):99–105. doi: 10.1016/s0168-8278(00)80165-8. [DOI] [PubMed] [Google Scholar]
  • 74.Villanueva A, Portela A, Sayols S, et al. DNA methylation-based prognosis and epidrivers in hepatocellular carcinoma. Hepatology. 2015 doi: 10.1002/hep.27732. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wei R, Huang GL, Zhang MY, et al. Clinical significance and prognostic value of microrna expression signatures in hepatocellular carcinoma. Clin. Cancer. Res. 2013;19(17):4780–4791. doi: 10.1158/1078-0432.CCR-12-2728. [DOI] [PubMed] [Google Scholar]
  • 76.Kim SM, Leem SH, Chu IS, et al. Sixty-five gene-based risk score classifier predicts overall survival in hepatocellular carcinoma. Hepatology. 2012;55(5):1443–1452. doi: 10.1002/hep.24813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Barry CT, D'souza M, Mccall M, et al. Micro rna expression profiles as adjunctive data to assess the risk of hepatocellular carcinoma recurrence after liver transplantation. Am. J. Transplant. 2012;12(2):428–437. doi: 10.1111/j.1600-6143.2011.03788.x. [DOI] [PubMed] [Google Scholar]
  • 78.Roessler S, Jia HL, Budhu A, et al. A unique metastasis gene signature enables prediction of tumor relapse in early-stage hepatocellular carcinoma patients. Cancer Res. 2010;70(24):10202–10212. doi: 10.1158/0008-5472.CAN-10-2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee JS, Heo J, Libbrecht L, et al. A novel prognostic subtype of human hepatocellular carcinoma derived from hepatic progenitor cells. Nat. Med. 2006;12(4):410–416. doi: 10.1038/nm1377. [DOI] [PubMed] [Google Scholar]
  • 80.Yamashita T, Honda M, Nakamoto Y, et al. Discrete nature of EpCAM+ and CD90+ cancer stem cells in human hepatocellular carcinoma. Hepatology. 2013;57(4):1484–1497. doi: 10.1002/hep.26168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Minguez B, Hoshida Y, Villanueva A, et al. Gene-expression signature of vascular invasion in hepatocellular carcinoma. J. Hepatol. 2011;55(6):1325–1331. doi: 10.1016/j.jhep.2011.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Toffanin S, Hoshida Y, Lachenmayer A, et al. Microrna-based classification of hepatocellular carcinoma and oncogenic role of miR-517a. Gastroenterology. 2011;140(5):1618–1628. e1616. doi: 10.1053/j.gastro.2011.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kurokawa Y, Matoba R, Takemasa I, et al. Molecular-based prediction of early recurrence in hepatocellular carcinoma. J. Hepatol. 2004;41(2):284–291. doi: 10.1016/j.jhep.2004.04.031. [DOI] [PubMed] [Google Scholar]
  • 84.Iizuka N, Oka M, Yamada-Okabe H, et al. Oligonucleotide microarray for prediction of early intrahepatic recurrence of hepatocellular carcinoma after curative resection. Lancet. 2003;361(9361):923–929. doi: 10.1016/S0140-6736(03)12775-4. [DOI] [PubMed] [Google Scholar]
  • 85.Wang SM, Ooi LL, Hui KM. Identification and validation of a novel gene signature associated with the recurrence of human hepatocellular carcinoma. Clin. Cancer. Res. 2007;13(21):6275–6283. doi: 10.1158/1078-0432.CCR-06-2236. [DOI] [PubMed] [Google Scholar]
  • 86.Woo HG, Park ES, Cheon JH, et al. Gene expression-based recurrence prediction of hepatitis B virus-related human hepatocellular carcinoma. Clin. Cancer. Res. 2008;14(7):2056–2064. doi: 10.1158/1078-0432.CCR-07-1473. [DOI] [PubMed] [Google Scholar]
  • 87.Van Malenstein H, Gevaert O, Libbrecht L, et al. A seven-gene set associated with chronic hypoxia of prognostic importance in hepatocellular carcinoma. Clin. Cancer. Res. 2010;16(16):4278–4288. doi: 10.1158/1078-0432.CCR-09-3274. [DOI] [PubMed] [Google Scholar]
  • 88.Ye QH, Qin LX, Forgues M, et al. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profiling and supervised machine learning. Nat. Med. 2003;9(4):416–423. doi: 10.1038/nm843. [DOI] [PubMed] [Google Scholar]
  • 89.Yoshioka S, Takemasa I, Nagano H, et al. Molecular prediction of early recurrence after resection of hepatocellular carcinoma. Eur. J. Cancer. 2009;45(5):881–889. doi: 10.1016/j.ejca.2008.12.019. [DOI] [PubMed] [Google Scholar]
  • 90.Okamoto M, Utsunomiya T, Wakiyama S, et al. Specific gene-expression profiles of noncancerous liver tissue predict the risk for multicentric occurrence of hepatocellular carcinoma in hepatitis C virus-positive patients. Ann. Surg. Oncol. 2006;13(7):947–954. doi: 10.1245/ASO.2006.07.018. [DOI] [PubMed] [Google Scholar]
  • 91.Chiang DY, Villanueva A, Hoshida Y, et al. Focal gains of VEGFa and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68(16):6779–6788. doi: 10.1158/0008-5472.CAN-08-0742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Jiang J, Gusev Y, Aderca I, et al. Association of microrna expression in hepatocellular carcinomas with hepatitis infection, cirrhosis, and patient survival. Clin. Cancer Res. 2008;14(2):419–427. doi: 10.1158/1078-0432.CCR-07-0523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Budhu A, Jia HL, Forgues M, et al. Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology. 2008;47(3):897–907. doi: 10.1002/hep.22160. [DOI] [PubMed] [Google Scholar]
  • 94.Cheng AL, Kang YK, Lin DY, et al. Sunitinib versus sorafenib in advanced hepatocellular cancer: results of a randomized Phase III trial. J. Clin. Oncol. 2013;31(32):4067–4075. doi: 10.1200/JCO.2012.45.8372. [DOI] [PubMed] [Google Scholar]
  • 95.Llovet JM, Hernandez-Gea V. Hepatocellular carcinoma: reasons for Phase III failure and novel perspectives on trial design. Clin. Cancer Res. 2014;20(8):2072–2079. doi: 10.1158/1078-0432.CCR-13-0547. [DOI] [PubMed] [Google Scholar]
  • 96.Villanueva A, Llovet JM. Targeted therapies for hepatocellular carcinoma. Gastroenterology. 2011;140(5):1410–1426. doi: 10.1053/j.gastro.2011.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Llovet JM. Liver cancer: time to evolve trial design after everolimus failure. Nat. Rev. Clin. Oncol. 2014;11(9):506–507. doi: 10.1038/nrclinonc.2014.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kudo M, Han G, Finn RS, et al. Brivanib as adjuvant therapy to transarterial chemoembolization in patients with hepatocellular carcinoma: a randomized Ohase III trial. Hepatology. 2014;60(5):1697–1707. doi: 10.1002/hep.27290. [DOI] [PubMed] [Google Scholar]
  • 99.Llovet JM, Decaens T, Raoul JL, et al. Brivanib in patients with advanced hepatocellular carcinoma who were intolerant to sorafenib or for whom sorafenib failed: results from the randomized Phase III BRISK-PS study. J. Clin. Oncol. 2013;31(28):3509–3516. doi: 10.1200/JCO.2012.47.3009. [DOI] [PubMed] [Google Scholar]
  • 100.Johnson PJ, Qin S, Park JW, et al. Brivanib versus sorafenib as first-line therapy in patients with unresectable, advanced hepatocellular carcinoma: results from the randomized Phase III brisk-fl study. J. Clin. Oncol. 2013;31(28):3517–3524. doi: 10.1200/JCO.2012.48.4410. [DOI] [PubMed] [Google Scholar]
  • 101.Zhu AX, Kudo M, Assenat E, et al. Effect of everolimus on survival in advanced hepatocellular carcinoma after failure of sorafenib: the EVOLVE-1 randomized clinical trial. JAMA. 2014;312(1):57–67. doi: 10.1001/jama.2014.7189. [DOI] [PubMed] [Google Scholar]
  • 102.Sawey ET, Chanrion M, Cai C, et al. Identification of a therapeutic strategy targeting amplified FGF19 in liver cancer by oncogenomic screening. Cancer Cell. 2011;19(3):347–358. doi: 10.1016/j.ccr.2011.01.040. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Hepatic Oncology are provided here courtesy of Taylor & Francis

RESOURCES