Abstract
Purpose of review
Intrahepatic cholangiocarcinoma (ICC) is a treatment-refractory disease with a dismal outcome. Limited success in the clinical management and a persistent increase in the incidence world-wide have made ICC one of the most lethal and fastest growing malignancies. However, recent advancements in genome-wide technologies combined with the application of integrative multidimensional analytical approaches have begun to provide both detailed insight into the underlying biological traits of ICC and identified new therapeutic opportunities.
Recent findings
In comparison with other cancers genomic studies of ICC have been limited. We and others have recently procured large cohorts of ICC patients intended for genome-wide analyses. In our study samples from ICC patients were obtained from three cancer centers and subjected to integrated genetic and genomic analyses. We provided new insights into both pathogenesis and optimal treatment options demonstrating the presence of unique subclasses of patients, based partly on KRAS mutations and increased levels of receptor tyrosine kinase signaling. The group of patients with the worst prognosis was characterized by transcriptional enrichment of genes regulating inflammation and proteasome activities, suggesting a combination of tyrosine kinase inhibitors and anti-inflammatory drugs as a new therapeutic option for these patients.
Summary
We have critically examined the progress in genome-wide studies of ICC including genetic profiling, transcriptomics and epigenomics. Current limitations in applying these technologies to archival samples and the insufficient access to fresh-frozen material are partly the cause of the delayed implementation of the omics-based investigations of ICC compared to other hepatobiliary diseases. Thus, selected candidate single gene studies will also be discussed.
Keywords: Cholangiocarcinoma (CCA), intrahepatic cholangiocarcinoma (ICC), extrahepatic cholangiocarcinoma (ECC)
INTRODUCTION
Cholangiocarcinoma (CCA) is a heterogeneous malignancy which arises in the epithelia of the intra- and extrahepatic biliary ducts. ICC is classified as a peripheral tumor of interlobular bile ducts whereas tumors designated hilar are generally considered extrahepatic and originate from the main hepatic ducts or at the bifurcation of the common hepatic duct. The main difference between peripheral and hilar tumors is in the clinical presentation and gross appearance which is likely a result of late diagnosis. Recent genomic data suggests that these tumor subtypes are less distinct than the underlying biological variances which classify patients according to overall survival and early tumor recurrence [1**]. Although recent molecular insight into ICC has improved, the understanding of genetic mechanisms involved in the development of ICC is still insufficient.
GENETIC ALTERATIONS IN INTRAHEPATIC CHOLANGIOCARCINOMA
Genetic analyses of ICC are still few and often limited to select genes. A number of studies analyzing combined hepatocellular-cholangiocarcinoma have attempted to assess the provocative hypothesis that ICC is, similar to hepatocellular carcinoma (HCC), derived from a common precursor cell [2–5]. Comparison of chromosomal imbalance, i.e. copy number gains and losses, suggests that ICC and HCC are closely related and that they share major genomic aberrations and abnormalities in molecular pathways involved in neoplastic transformation.
1. Chromosomal instability – Copy number variations
Few comparative genomics hybridization (CGH) studies on ICC were performed during the past decade [6–10*]. Unfortunately, in several studies where the authors have investigated copy number variations (CNVs) in biliary tract cancers, the analyses were merged between subtypes such as intrahepatic and extrahepatic tumors [11] or even including gallbladders [12*–14], making an accurate interpretation difficult. We have reviewed 5 studies where data for 98 typical ICCs were investigated separately [6–10*]. In our meta-analysis of ICC, CGH revealed frequent copy number losses on chromosomal arm 1p, 4q, 8p, 9p, 17p and 18q while copy number gains were found on chromosome 1q, 5p, 7p, 8q, 17q and 20q in at least 3 studies with greater than 20% overall change (Table 1). Although we did identify common CNVs, the complexity of the karyotype revealed by the degree of genomic imbalance differed substantially among the 5 studies. Whereas the rigid parameters of our meta-analysis clearly do limit the data, the common CNVs identified for ICC may be of significance. It is also noteworthy to indicate that recent advancements in genomics, the limited number of cases analyzed in each study and the ethnic diversity (3 studies were performed in Korea [7–9*], one in Hong Kong [10*] and one in Germany [6*]) do warrant a more detailed analysis. In contrast, a meta-analysis of 31 CGH studies performed in 2005 examined a total of 785 cases of HCC [15]. Interestingly, 7 of 9 loci identified with significant CNV in HCC showed similar gains on 1q, 8q and 17q and losses on 4q, 8p, 13q and 17p common with ICC. These results suggest that HCC and ICC are genomically closely related.
Table 1.
Chromosome arm | ICC | Studies (no) | Koo et al. [7] (Korea) | Wong et al. [10] (Hong Kong) | Lee et al. [8] (Korea) | Uhm et al. [9] (Korea) | Homayounfar et al. [6] (Germany) | Moinzadeh et al. [15] HCC (31 studies) |
---|---|---|---|---|---|---|---|---|
Cases (no.) | 98 | 5 | 11 | 13 | 33 | 19 | 22 | 785 |
1p- | 28 | 3 | 21 | 37 | 27 | |||
3p- | 53 | 2 | 38 | 68 | ||||
3q- | 23 | 1 | 23 | |||||
4p- | 36 | 1 | 36 | |||||
4q- | 35 | 4 | 46 | 21 | 32 | 41 | 34 | |
5q- | 26 | 1 | 26 | |||||
6q- | 69 | 2 | 46 | 91 | ||||
8p- | 33 | 3 | 31 | 26 | 41 | 38 | ||
9p- | 40 | 3 | 38 | 26 | 55 | |||
9q- | 36 | 1 | 36 | |||||
11p- | 23 | 1 | 23 | |||||
11q- | 23 | 1 | 23 | |||||
12q- | 23 | 1 | 23 | |||||
13q- | 45 | 1 | 45 | 26 | ||||
14q- | 43 | 2 | 31 | 55 | ||||
16q- | 0 | 0 | 36 | |||||
17p- | 40 | 3 | 55 | 38 | 26 | 32 | ||
18q- | 31 | 3 | 38 | 32 | 23 | |||
19p- | 32 | 1 | 32 | |||||
21q- | 23 | 1 | 23 | |||||
1p+ | 23 | 1 | 23 | |||||
1q+ | 40 | 3 | 54 | 24 | 41 | 57 | ||
2q+ | 26 | 1 | 26 | |||||
3q+ | 37 | 2 | 46 | 27 | ||||
5p+ | 28 | 3 | 27 | 24 | 32 | |||
5q+ | 27 | 2 | 31 | 23 | ||||
6p+ | 0 | 0 | 22 | |||||
7p+ | 28 | 3 | 27 | 26 | 32 | |||
7q+ | 36 | 1 | 36 | |||||
8p+ | 39 | 1 | 39 | |||||
8q+ | 45 | 4 | 36 | 54 | 58 | 32 | 47 | |
11q+ | 37 | 2 | 31 | 42 | ||||
12p+ | 27 | 1 | 27 | |||||
12q+ | 23 | 1 | 23 | |||||
13q+ | 45 | 1 | 45 | |||||
15q+ | 37 | 2 | 38 | 36 | ||||
16p+ | 36 | 1 | 36 | |||||
17q+ | 42 | 3 | 38 | 61 | 26 | 22 | ||
18p+ | 39 | 1 | 39 | |||||
18q+ | 27 | 2 | 27 | 26 | ||||
19q+ | 26 | 1 | 26 | |||||
20q+ | 45 | 3 | 36 | 31 | 67 |
2. Prevalent mutations identified in single gene studies
The first high-coverage genome sequencing project of ICC, e.g. whole genome or exome analyses, has not been performed. Most of the studies investigating genetic variation in ICC have focused on a specific gene, or at most a few genes. A project of this magnitude promises to identify and achieve a detailed map of the common and rare genetic variants to attain an understanding of the underlying genetic traits which result in the development of CCA. Insight into the role of genetic variants may improve the selection of therapy and, thus, avoid drug-resistance. A few common mutations in genes such as KRAS, BRAF, EGFR, PIK3CA and TP53 were investigated in CCA; however, clinical outcome of the disease has not improved.
Among the pathways involved in the pathogenesis of ICC, the ErbB-family of receptors are perhaps affected the most [16]. The most notably are the aberrant regulation of ErbB2 and the epidermal growth factor receptor (EGFR) signaling. Whereas no mutations have been reported in ERBB2, increased expression and DNA amplification was reported ranging from 0 to 73% in tumors [16–18]. EGFR-targeted therapies may show promise in the treatment of a subclass of ICCs. Mutations in EGFR are rare events and we did not detect any in our cohort, however, in the subclass of patients with poor survival we observe a significant overexpression of EGFR and its downstream pathways [1**]. Activating mutations in EGFR were described in two small studies in 3/22 (13.6%) and 3/15 (20%) of cases [19–20]. Activation of EGFR triggers the activation of downstream RAS/RAF/MEK/ERK and PI3K/PTEN/AKT, two major cell survival pathways. Hotspot activating missense mutations in genes downstream to EGFR, such as, BRAF (0–22%) [21–22] and PIK3CA (9–32%) [23–24] are rarely described in ICC.
Gain of function mutations in KRAS downstream of EGFR represents one of the most frequent mutations found in ICC (8–54%) [25–28] as compared to 25% in our cohort of peripheral and hilar-type tumors [1**]. When classified by tumor site, 17% of peripheral-type CCAs were positive for mutations in KRAS with the most frequent alteration in codon12. Importantly, the incidence of mutations was higher in the hilar-type tumors (53%) [1**, 29]. It is noteworthy that KRAS mutations were detected at a higher incidence with increasing tumor stage (stage I, 8%; stage II, 15%; stage III, 31%; stage IV, 46%) [24], however, confirmation in an independent cohort is needed. Mutations in KRAS were detected in 30% of bile from patients with primary sclerosing cholangitis, suggesting that it is an early event contributing to the malignant transformation of cholangiocytes [30]. Although KRAS is established as a causative somatic mutation, we were unable to determine KRAS as an independent prognostic factor within our cohort [1**]. Integrating the KRAS mutational status with our prognostic gene classifier grouped all patients with mutated KRAS amid patients with poor prognosis.
Another pathway involved in ICC carcinogenesis which is frequently inactivated and associated with loss of heterozygosity is the central cell cycle regulator p53. More than 90 different mutations have been described in TP53 of which most are associated with exposure to chemical carcinogens [31], such as thorotrast [32]. A review of 10 studies which included 229 cases found a total of 21% (49 patients) with mutations in TP53, ranging from 23% in Asia, 14% in Europe and 26% in the USA [33]. However, there is a great deal of variability in the reported cases for most genetic alterations. TP53 is no exception with 14–77% of cases positive reported in single studies [31, 33]. Other genes such as CTNNB1 (8%), APC (13%), AXIN1 (41%) and CDH1 (11%) have occasionally been reported [34–35].
ALTERATIONS IN THE EPIGENOME IN THE PROCESS OF CHOLANGIOCARCINOGENESIS
Recent technological advances have brought epigenetics into the omics-age by the introduction of array-based and deep sequencing methods, highlighting the importance of the epigenome, including DNA CpG methylation, histone modifications and non-coding RNA species, in the process of human carcinogenesis. Only limited comprehensive epigenomic studies have been published on ICC and data on aberrant CpG promoter methylation in the regulation of ICC has largely been centered on individual genes (reviewed in [36*–39*]). This review provides an overview of select key genes frequently hypermethylated and microRNAs identified by miR-profiles in ICC.
1. Promoter hypermethylation in cholangiocarcinoma
Aberrant epigenetic regulation such as promoter hypermethylation was demonstrated in numerous important cancer-associated genes in ICC (a comprehensive list can be found in [36*–37*]). Studies profiling these modifications to established prognostic and predictive gene signatures attempting to predict the therapeutic potential of agents that target the cancer epigenome [40*] have not yet been investigated in ICC. Recently, a study in a large ICC cohort (n=102) associated with liver fluke infection was published demonstrating promoter hypermethylation in a handful of target genes compared to adjacent tissue [41]. Unfortunately, the authors only investigate 26 selected loci using traditional methodology rather than applying readily available genome-wide array-based technology.
Epigenetic silencing by promoter hypermethylation is common in tumor suppressor genes: p16INK4a/CDKN2 (17–83%) [42–45]; SOCS3 (62%) [46]; RASSF1A (31–69%) [10, 42, 45]; p15 (54%) [45]; CDH1 (17–49%) [42–43, 45]; hMLH1 (19–45%) [45]; APC (27–47%) [42–43, 45]; p14ARF (19–30%) [42, 44–45] and GSTP1 (15–31%) [42, 45] are some of the most frequent events reported in ICC.
ICC often arises in the background of chronic inflammation such as primary sclerosing cholangitis (PSC) which provides survival signals to the tumor (the contribution of the inflammatory microenvironment and epigenetics is reviewed in [47]). The cyclin-dependent kinase inhibitor p16 is frequently silenced by epigenetic modifications and was proposed as a prognostic marker associated with dismal outcome in patients with PSC [48]. Also, inflammatory signals (such as AKT) can trigger cytokine pathways (i.e. JAK/STAT) which in turn were shown to be involved in cholangiocarcinogenesis [49–50]. Indeed, a common target in ICC is the suppressor of cytokine signaling 3 (SOCS3) which is silenced by promoter hypermethylation [46]. Inhibition of SOCS3 results in a self-enhancing constitutive activation of the oncogenic pathway IL6/STAT3 with a subsequent autocrine feedback and paracrine signaling to the tumor environment [46, 49–51].
The frequency by which these targets are methylated in ICC is variable and the ability to utilize any or a group of silenced genes as predictors for outcome remains to be critically investigated. Since inconsistencies can be a result of limited study size, heterogeneous cohorts (i.e. mixed ICC and ECC cases), etiology and methodology, future studies applying epigenome-wide technology to a standardized ICC cohort will be needed to enhance our understanding of both the underlying biology and importance of epigenetic changes in ICC pathogenesis.
2. Profiling of non-coding RNAs
Non-coding RNAs include both large and small RNA species. A major group and the best characterized non-coding RNAs in ICC are microRNAs (miRs) which are involved in the epigenetic regulation of gene expression, transcriptional stability and translation. A number of studies have concentrated on investigating the biological significance and aberrant expression of miRs in ICC cell lines [52*–57], demonstrating a link between tumor growth, response to therapy and expression of inflammatory cytokines (reviewed in [38*]). Recently, Kawahigashi, Y. and colleagues [58] performed a comprehensive study by sequencing two ICC cell lines (HuCCT1 and MEC) and a normal intrahepatic bile duct epithelial cell line (HIBEpiC), providing the first profile of differently expressed miRs in ICC derived cells. A unique 27 miRs ICC signature was identified that included down-regulation of 8 miRs (i.e. miR-22, miR-125a, miR-127, miR-199a, miR-199a*, miR-214, miR-376a and miR-424) specific for normal bile duct epithelium. The authors suggested that these miRs may serve as biomarkers for ICC, however the functional and clinicopathological significance of this miR signature remains to be evaluated in clinical samples. In two recent studies miRs were profiled in ICC [53**, 59**]. Selaru and colleagues [59**] showed that expression level of miR-21 could distinguish ICC from normal bile ducts. Furthermore, increasing expression of miR-21 correlated with a decreased expression of programmed cell death 4 (PDCD4) and tissue inhibitor of metalloproteinase 3 (TIMP3), suggesting that miR-21 may act as an oncomir in ICC. However, 9 of the 23 samples in this study were of distal extrahepatic origin, making the interpretation of the data difficult. Another study profiling 27 ICCs reported 38 miRs differentially expressed in the tumors compared to 10 normal cholangiocytes [53**]. Interestingly, hierarchical clustering grouped the clinical samples into two clusters, distinguishing the patients according to the level of CA19-9, a serum secreted mucin-type glycoprotein which is commonly used as a biomarker for CCA and was shown to be associated with poor prognosis after surgical resection of ICC [60]. In addition, the status of vascular invasion was found to be significantly correlated with the two clusters. However, the potential prognostic importance of the 38 miRs signature remains to be evaluated in an independent cohort.
TRANSCRIPTOMIC PROFILING OF ICC
Tumor infiltrating stroma is often a prominent characteristic of ICC, indicating that to capture the gene expression profile of the tumor epithelial, laser microdissection may be needed. The first comprehensive transcriptomic study applied this approach to 25 ICCs and identified a total of 473 differentially expressed genes between the tumor and non-malignant biliary epithelial cells [61]. Even though the authors established a clean gene signature in the tumor epithelial compartment the stromal compartment was not investigated. Few studies have since been published analyzing the gene expression profiles in ICCs [62–63], associated with liver fluke infection [64–65] or anatomical site of the tumor [12]. We recently published a genetic and genomic study in a large cohort of 104 peripheral- and hilar-type tumors and 59 matched surrounding liver samples from Australia, Europe and USA, accompanied by a detailed analysis of the epithelial and stromal compartments from 23 laser-capture microdissected tumors [1**]. Our study provided new insights into the ICC pathogenesis and identified two patient categories by a 238-gene prognostic classifier which stratified the patients according to overall survival and early recurrence. We minimized the classifier to 36 genes which in combination with other molecular predictors (i.e. mutations, co-activation of multiple oncogenic pathways), improved the molecular classification and outcome prediction in the cohort. A detailed class comparison identified a total of four patient subclasses based on dependence of receptor tyrosine kinases (EGFR, ERBB2 and MET), activation of the mTOR pathway, increased proliferation and overrepresentation of gene involved in proteasomal activity and inflammation. These data suggested a novel therapeutic potential for dual-target tyrosine kinase inhibitors (e.g. lapatinib), either alone or in combination with proteasome inhibitors to improve therapeutic response of this currently treatment-refractory malignancy.
CONCLUSION
ICC is a rare malignancy, diagnosed at a frequency less than 1% of primary hepatic tumors. Genome-wide technologies have provided a detailed characterization of genetic variation, genomics, and epigenomics in ICC. This unprecedented capacity to study the underlying traits of a human disease such as ICC at the molecular level and to integrate these data at a multidimensional scale suggests that future advancements in clinical management may come from systems-based translational genomics. The realization of this goal, at least for ICC, may require extensive collaborative team efforts across institutions.
KEY POINTS.
CCA is a rare disease with poor outcome, and increasing incidence worldwide. A limited number of genomic studies on CCA have been published during the past decade. Inadequate sample size and heterogeneous anatomical locations of CCA (intrahepatic, extrahepatic (hilar) and distal extrahepatic) frequently complicate interpretation of the data.
Receptor tyrosine kinases have been shown to be potential drug-able targets in CCA. Detailed translational approaches are needed to study the efficacy of tyrosine kinase inhibitors in CCA either alone or in combination with other chemotherapeutics. This may warrant small proof of concept clinical trials.
Application of translational system biology approaches, and utilizing multidimensional integrative techniques including whole genome sequencing promises to yield both deeper insights into the mechanism of CCA pathogenesis as well as providing novel therapeutic options for CCA patients.
Footnotes
Conflicts of interest
No potential conflict of interest related to this study was reported. The study was supported by the Intramural Research Program of the Center for Cancer Research, NCI, NIH.
REFERENCES AND RECOMMENDED READING
- 1**.Andersen JB, Spee B, Blechacz BR, et al. Genomic and Genetic Characterization of Cholangiocarcinoma Identifies Therapeutic Targets for Tyrosine Kinase Inhibitors. Gastroenterology. 2011 doi: 10.1053/j.gastro.2011.12.005. In press. The first detailed molecular characterization of a large cohort. The study identified four subclasses of CCA patients identifying the potential efficacy of TKIs in patients with poor clinical prognosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cazals-Hatem D, Rebouissou S, Bioulac-Sage P, et al. Clinical and molecular analysis of combined hepatocellular-cholangiocarcinomas. J Hepatol. 2004;41:292–8. doi: 10.1016/j.jhep.2004.04.030. [DOI] [PubMed] [Google Scholar]
- 3.Fujii H, Zhu XG, Matsumoto T, et al. Genetic classification of combined hepatocellular-cholangiocarcinoma. Hum Pathol. 2000;31:1011–7. doi: 10.1053/hupa.2000.9782. [DOI] [PubMed] [Google Scholar]
- 4.Imai Y, Oda H, Arai M, et al. Mutational analysis of the p53 and K-ras genes and allelotype study of the Rb-1 gene for investigating the pathogenesis of combined hapatocellular-cholangiocellular carcinomas. Jpn J Cancer Res. 1996;87:1056–62. doi: 10.1111/j.1349-7006.1996.tb03110.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Murata M, Miyoshi Y, Iwao K, et al. Combined hepatocellular/cholangiocellular carcinoma with sarcomatoid features: genetic analysis for histogenesis. Hepatol Res. 2001;21:220–227. doi: 10.1016/s1386-6346(01)00100-0. [DOI] [PubMed] [Google Scholar]
- 6*.Homayounfar K, Gunawan B, Cameron S, et al. Pattern of chromosomal aberrations in primary liver cancers identified by comparative genomic hybridization. Hum Pathol. 2009;40:834–42. doi: 10.1016/j.humpath.2008.11.005. Analysis of CNVs in ICC. [DOI] [PubMed] [Google Scholar]
- 7*.Koo SH, Ihm CH, Kwon KC, et al. Genetic alterations in hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Cancer Genet Cytogenet. 2001;130:22–8. doi: 10.1016/s0165-4608(01)00460-5. The first study to analyze ICC for potential copy number gains or losses. [DOI] [PubMed] [Google Scholar]
- 8*.Lee JY, Park YN, Uhm KO, et al. Genetic alterations in intrahepatic cholangiocarcinoma as revealed by degenerate oligonucleotide primed PCR-comparative genomic hybridization. J Korean Med Sci. 2004;19:682–7. doi: 10.3346/jkms.2004.19.5.682. Analysis of CNVs in ICC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9*.Uhm KO, Park YN, Lee JY, et al. Chromosomal imbalances in Korean intrahepatic cholangiocarcinoma by comparative genomic hybridization. Cancer Genet Cytogenet. 2005;157:37–41. doi: 10.1016/j.cancergencyto.2004.05.007. Analysis of CNVs in ICC. [DOI] [PubMed] [Google Scholar]
- 10*.Wong N, Li L, Tsang K, et al. Frequent loss of chromosome 3p and hypermethylation of RASSF1A in cholangiocarcinoma. J Hepatol. 2002;37:633–9. doi: 10.1016/s0168-8278(02)00269-6. Analysis of CNVs in ICC. [DOI] [PubMed] [Google Scholar]
- 11.McKay SC, Unger K, Pericleous S, et al. Array comparative genomic hybridization identifies novel potential therapeutic targets in cholangiocarcinoma. HPB (Oxford) 2011;13:309–19. doi: 10.1111/j.1477-2574.2010.00286.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12*.Miller G, Socci ND, Dhall D, et al. Genome wide analysis and clinical correlation of chromosomal and transcriptional mutations in cancers of the biliary tract. J Exp Clin Cancer Res. 2009;28:62. doi: 10.1186/1756-9966-28-62. This study is the first to analyze changes in gene expression and DNA copy number in the same samples. The study is further more a comparison between intra-, extrahepatic and gallbladder tumors. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Shiraishi K, Okita K, Harada T, et al. Comparative genomic hybridization analysis of genetic aberrations associated with development and progression of biliary tract carcinomas. Cancer. 2001;91:570–7. doi: 10.1002/1097-0142(20010201)91:3<570::aid-cncr1036>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 14.Shiraishi K, Okita K, Kusano N, et al. A comparison of DNA copy number changes detected by comparative genomic hybridization in malignancies of the liver, biliary tract and pancreas. Oncology. 2001;60:151–61. doi: 10.1159/000055313. [DOI] [PubMed] [Google Scholar]
- 15.Moinzadeh P, Breuhahn K, Stutzer H, et al. Chromosome alterations in human hepatocellular carcinomas correlate with aetiology and histological grade--results of an explorative CGH meta-analysis. Br J Cancer. 2005;92:935–41. doi: 10.1038/sj.bjc.6602448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sirica AE. Role of ErbB family receptor tyrosine kinases in intrahepatic cholangiocarcinoma. World J Gastroenterol. 2008;14:7033–58. doi: 10.3748/wjg.14.7033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Endo K, Yoon BI, Pairojkul C, et al. ERBB-2 overexpression and cyclooxygenase-2 up-regulation in human cholangiocarcinoma and risk conditions. Hepatology. 2002;36:439–50. doi: 10.1053/jhep.2002.34435. [DOI] [PubMed] [Google Scholar]
- 18.Sirica AE, Lai GH, Endo K, et al. Cyclooxygenase-2 and ERBB-2 in cholangiocarcinoma: potential therapeutic targets. Semin Liver Dis. 2002;22:303–13. doi: 10.1055/s-2002-34507. [DOI] [PubMed] [Google Scholar]
- 19.Gwak GY, Yoon JH, Shin CM, et al. Detection of response-predicting mutations in the kinase domain of the epidermal growth factor receptor gene in cholangiocarcinomas. J Cancer Res Clin Oncol. 2005;131:649–52. doi: 10.1007/s00432-005-0016-1. [DOI] [PubMed] [Google Scholar]
- 20.Leone F, Cavalloni G, Pignochino Y, et al. Somatic mutations of epidermal growth factor receptor in bile duct and gallbladder carcinoma. Clin Cancer Res. 2006;12:1680–5. doi: 10.1158/1078-0432.CCR-05-1692. [DOI] [PubMed] [Google Scholar]
- 21.Goldenberg D, Rosenbaum E, Argani P, et al. The V599E BRAF mutation is uncommon in biliary tract cancers. Mod Pathol. 2004;17:1386–91. doi: 10.1038/modpathol.3800204. [DOI] [PubMed] [Google Scholar]
- 22.Tannapfel A, Sommerer F, Benicke M, et al. Mutations of the BRAF gene in cholangiocarcinoma but not in hepatocellular carcinoma. Gut. 2003;52:706–12. doi: 10.1136/gut.52.5.706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Riener MO, Bawohl M, Clavien PA, et al. Rare PIK3CA hotspot mutations in carcinomas of the biliary tract. Genes Chromosomes Cancer. 2008;47:363–7. doi: 10.1002/gcc.20540. [DOI] [PubMed] [Google Scholar]
- 24.Xu RF, Sun JP, Zhang SR, et al. KRAS and PIK3CA but not BRAF genes are frequently mutated in Chinese cholangiocarcinoma patients. Biomed Pharmacother. 2011;65:22–6. doi: 10.1016/j.biopha.2010.06.009. [DOI] [PubMed] [Google Scholar]
- 25.Isa T, Tomita S, Nakachi A, et al. Analysis of microsatellite instability, K-ras gene mutation and p53 protein overexpression in intrahepatic cholangiocarcinoma. Hepatogastroenterology. 2002;49:604–8. [PubMed] [Google Scholar]
- 26.Kiba T, Tsuda H, Pairojkul C, et al. Mutations of the p53 tumor suppressor gene and the ras gene family in intrahepatic cholangiocellular carcinomas in Japan and Thailand. Mol Carcinog. 1993;8:312–8. doi: 10.1002/mc.2940080415. [DOI] [PubMed] [Google Scholar]
- 27.Ohashi K, Nakajima Y, Kanehiro H, et al. Ki-ras mutations and p53 protein expressions in intrahepatic cholangiocarcinomas: relation to gross tumor morphology. Gastroenterology. 1995;109:1612–7. doi: 10.1016/0016-5085(95)90650-9. [DOI] [PubMed] [Google Scholar]
- 28.Tannapfel A, Benicke M, Katalinic A, et al. Frequency of p16(INK4A) alterations and K-ras mutations in intrahepatic cholangiocarcinoma of the liver. Gut. 2000;47:721–7. doi: 10.1136/gut.47.5.721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tada M, Omata M, Ohto M. High incidence of ras gene mutation in intrahepatic cholangiocarcinoma. Cancer. 1992;69:1115–8. doi: 10.1002/cncr.2820690509. [DOI] [PubMed] [Google Scholar]
- 30.Kubicka S, Kuhnel F, Flemming P, et al. K-ras mutations in the bile of patients with primary sclerosing cholangitis. Gut. 2001;48:403–8. doi: 10.1136/gut.48.3.403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Khan SA, Taylor-Robinson SD, Carmichael PL, et al. Analysis of p53 mutations for a mutational signature in human intrahepatic cholangiocarcinoma. Int J Oncol. 2006;28:1269–77. [PubMed] [Google Scholar]
- 32.Kamikawa T, Amenomori M, Itoh T, et al. Analysis of genetic changes in intrahepatic cholangiocarcinoma induced by thorotrast. Radiat Res. 1999;152:S118–24. [PubMed] [Google Scholar]
- 33.Khan SA, Thomas HC, Toledano MB, et al. p53 Mutations in human cholangiocarcinoma: a review. Liver Int. 2005;25:704–16. doi: 10.1111/j.1478-3231.2005.01106.x. [DOI] [PubMed] [Google Scholar]
- 34.Endo K, Ashida K, Miyake N, et al. E-cadherin gene mutations in human intrahepatic cholangiocarcinoma. J Pathol. 2001;193:310–7. doi: 10.1002/1096-9896(2000)9999:9999<::AID-PATH816>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- 35.Tokumoto N, Ikeda S, Ishizaki Y, et al. Immunohistochemical and mutational analyses of Wnt signaling components and target genes in intrahepatic cholangiocarcinomas. Int J Oncol. 2005;27:973–80. [PubMed] [Google Scholar]
- 36*.Isomoto H. Epigenetic alterations associated with cholangiocarcinoma (review) Oncol Rep. 2009;22:227–32. Review giving an overview of epigenetic alterations in CCA. [PubMed] [Google Scholar]
- 37*.Sandhu DS, Shire AM, Roberts LR. Epigenetic DNA hypermethylation in cholangiocarcinoma: potential roles in pathogenesis, diagnosis and identification of treatment targets. Liver Int. 2008;28:12–27. doi: 10.1111/j.1478-3231.2007.01624.x. Excellent detailed review discussing single genes shown to be affected by DNA methylation in CCA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38*.Stutes M, Tran S, DeMorrow S. Genetic and epigenetic changes associated with cholangiocarcinoma: from DNA methylation to microRNAs. World J Gastroenterol. 2007;13:6465–9. doi: 10.3748/wjg.v13.i48.6465. Review giving a short overview of the involvement of DNA methylation and miRs in CCA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tischoff I, Wittekind C, Tannapfel A. Role of epigenetic alterations in cholangiocarcinoma. J Hepatobiliary Pancreat Surg. 2006;13:274–9. doi: 10.1007/s00534-005-1055-3. Review describing epigenetic alteration in CCA. [DOI] [PubMed] [Google Scholar]
- 40*.Andersen JB, Factor VM, Marquardt JU, et al. An integrated genomic and epigenomic approach predicts therapeutic response to zebularine in human liver cancer. Sci Transl Med. 2010;2:54ra77. doi: 10.1126/scitranslmed.3001338. A genomic and epigenomic prediction of response to epigenetic therapy in primary liver cancer. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sriraksa R, Zeller C, El-Bahrawy MA, et al. CpG-island methylation study of liver fluke-related cholangiocarcinoma. Br J Cancer. 2011;104:1313–8. doi: 10.1038/bjc.2011.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kim BH, Cho NY, Choi M, et al. Methylation profiles of multiple CpG island loci in extrahepatic cholangiocarcinoma versus those of intrahepatic cholangiocarcinomas. Arch Pathol Lab Med. 2007;131:923–30. doi: 10.5858/2007-131-923-MPOMCI. [DOI] [PubMed] [Google Scholar]
- 43.Lee S, Kim WH, Jung HY, et al. Aberrant CpG island methylation of multiple genes in intrahepatic cholangiocarcinoma. Am J Pathol. 2002;161:1015–22. doi: 10.1016/S0002-9440(10)64262-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Tannapfel A, Sommerer F, Benicke M, et al. Genetic and epigenetic alterations of the INK4a-ARF pathway in cholangiocarcinoma. J Pathol. 2002;197:624–31. doi: 10.1002/path.1139. [DOI] [PubMed] [Google Scholar]
- 45.Yang B, House MG, Guo M, et al. Promoter methylation profiles of tumor suppressor genes in intrahepatic and extrahepatic cholangiocarcinoma. Mod Pathol. 2005;18:412–20. doi: 10.1038/modpathol.3800287. [DOI] [PubMed] [Google Scholar]
- 46.Isomoto H, Mott JL, Kobayashi S, et al. Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing. Gastroenterology. 2007;132:384–96. doi: 10.1053/j.gastro.2006.10.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Huang L, Frampton G, Liang LJ, et al. Aberrant DNA methylation profile in cholangiocarcinoma. World J Gastrointest Pathophysiol. 2010;1:23–9. doi: 10.4291/wjgp.v1.i2.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ahrendt SA, Eisenberger CF, Yip L, et al. Chromosome 9p21 loss and p16 inactivation in primary sclerosing cholangitis-associated cholangiocarcinoma. J Surg Res. 1999;84:88–93. doi: 10.1006/jsre.1999.5615. [DOI] [PubMed] [Google Scholar]
- 49.Isomoto H, Kobayashi S, Werneburg NW, et al. Interleukin 6 upregulates myeloid cell leukemia-1 expression through a STAT3 pathway in cholangiocarcinoma cells. Hepatology. 2005;42:1329–38. doi: 10.1002/hep.20966. [DOI] [PubMed] [Google Scholar]
- 50.Kobayashi S, Werneburg NW, Bronk SF, et al. Interleukin-6 contributes to Mcl-1 up-regulation and TRAIL resistance via an Akt-signaling pathway in cholangiocarcinoma cells. Gastroenterology. 2005;128:2054–65. doi: 10.1053/j.gastro.2005.03.010. [DOI] [PubMed] [Google Scholar]
- 51.Wehbe H, Henson R, Meng F, et al. Interleukin-6 contributes to growth in cholangiocarcinoma cells by aberrant promoter methylation and gene expression. Cancer Res. 2006;66:10517–24. doi: 10.1158/0008-5472.CAN-06-2130. [DOI] [PubMed] [Google Scholar]
- 52*.Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology. 2010;51:881–90. doi: 10.1002/hep.23381. Interesting single gene study providing evidence for a link between mir expression, maintainance of DNA methylation and cytokine expression in the regulation of cholangiocarcinoma. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53**.Chen L, Yan HX, Yang W, et al. The role of microRNA expression pattern in human intrahepatic cholangiocarcinoma. J Hepatol. 2009;50:358–69. doi: 10.1016/j.jhep.2008.09.015. In this study the authors have used a combination of laser microdissection and TaqMan MicroRNA Array assay to profile 27 ICCs. Two miRs were selected (mir-204 and mir-320) for further study demonstrating a negative regulation of Mcl-1 and Bcl-2. [DOI] [PubMed] [Google Scholar]
- 54.Meng F, Henson R, Lang M, et al. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology. 2006;130:2113–29. doi: 10.1053/j.gastro.2006.02.057. [DOI] [PubMed] [Google Scholar]
- 55.Meng F, Henson R, Wehbe-Janek H, et al. The MicroRNA let-7a modulates interleukin-6-dependent STAT-3 survival signaling in malignant human cholangiocytes. J Biol Chem. 2007;282:8256–64. doi: 10.1074/jbc.M607712200. [DOI] [PubMed] [Google Scholar]
- 56.Meng F, Wehbe-Janek H, Henson R, et al. Epigenetic regulation of microRNA-370 by interleukin-6 in malignant human cholangiocytes. Oncogene. 2008;27:378–86. doi: 10.1038/sj.onc.1210648. [DOI] [PubMed] [Google Scholar]
- 57.Mott JL, Kobayashi S, Bronk SF, et al. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 2007;26:6133–40. doi: 10.1038/sj.onc.1210436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kawahigashi Y, Mishima T, Mizuguchi Y, et al. MicroRNA profiling of human intrahepatic cholangiocarcinoma cell lines reveals biliary epithelial cell-specific microRNAs. J Nihon Med Sch. 2009;76:188–97. doi: 10.1272/jnms.76.188. [DOI] [PubMed] [Google Scholar]
- 59**.Selaru FM, Olaru AV, Kan T, et al. MicroRNA-21 is overexpressed in human cholangiocarcinoma and regulates programmed cell death 4 and tissue inhibitor of metalloproteinase 3. Hepatology. 2009;49:1595–601. doi: 10.1002/hep.22838. Though this paper is not a profiling study and therefore not directly the topic of this review, it is an excellent analysis of mir-21. The study suggests that mir-21 is an oncomir in part by inhibiting PDCD4 and TIMP3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Huang JL, Biehl TR, Lee FT, et al. Outcomes after resection of cholangiocellular carcinoma. Am J Surg. 2004;187:612–7. doi: 10.1016/j.amjsurg.2004.01.007. [DOI] [PubMed] [Google Scholar]
- 61.Obama K, Ura K, Li M, et al. Genome-wide analysis of gene expression in human intrahepatic cholangiocarcinoma. Hepatology. 2005;41:1339–48. doi: 10.1002/hep.20718. [DOI] [PubMed] [Google Scholar]
- 62.Seol MA, Chu IS, Lee MJ, et al. Genome-wide expression patterns associated with oncogenesis and sarcomatous transdifferentation of cholangiocarcinoma. BMC Cancer. 2011;11:78. doi: 10.1186/1471-2407-11-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang AG, Yoon SY, Oh JH, et al. Identification of intrahepatic cholangiocarcinoma related genes by comparison with normal liver tissues using expressed sequence tags. Biochem Biophys Res Commun. 2006;345:1022–32. doi: 10.1016/j.bbrc.2006.04.175. [DOI] [PubMed] [Google Scholar]
- 64.Jinawath N, Chamgramol Y, Furukawa Y, et al. Comparison of gene expression profiles between Opisthorchis viverrini and non-Opisthorchis viverrini associated human intrahepatic cholangiocarcinoma. Hepatology. 2006;44:1025–38. doi: 10.1002/hep.21330. [DOI] [PubMed] [Google Scholar]
- 65.Pak JH, Kim DW, Moon JH, et al. Differential gene expression profiling in human cholangiocarcinoma cells treated with Clonorchis sinensis excretory-secretory products. Parasitol Res. 2009;104:1035–46. doi: 10.1007/s00436-008-1286-8. [DOI] [PubMed] [Google Scholar]