Skip to main content
American Journal of Cancer Research logoLink to American Journal of Cancer Research
. 2016 Feb 15;6(3):577–586.

Genetic profiling of intrahepatic cholangiocarcinoma and its clinical implication in targeted therapy

Diyang Xie 1, Zhenggang Ren 1, Jia Fan 1,2, Qiang Gao 1
PMCID: PMC4851838  PMID: 27152236

Abstract

Intrahepatic cholangiocarcinoma (iCCA) is a treatment-refractory primary liver cancer with an increasing incidence and mortality worldwide in recent years. Lack of a stereotyped genetic signature and limited understanding of genomic landscape make the development of effective targeted therapies challenging. Recent application of advanced technologies such as next-generation sequencing (NGS) has broadened our understanding of genetic heterogeneity in iCCA and many potentially actionable genetic alterations have been identified. This review explores the recent advances in defining genetic alterations in iCCAs, which may present potent therapeutic targets. Chromatin remodeling genes and genes encoding isocitrate dehydrogenase and tyrosine kinase receptors as well as their downstream effectors are among the most frequently altered genes. Clinical trials testing the effect of new targeted agents on iCCA patients, especially those with the above genetic markers are under way. However, the complex interplay of environmental and evolutionary factors contributing to the genetic variability in iCCA calls for a more cautionary use of NGS in tailoring targeted regimen to the patients. Next-generation functional testing may complement NGS to execute precision medicine in future.

Keywords: Cholangiocarcinoma, genetic heterogeneity, targeted therapy

Introduction

Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer, accounting for up to 15% of all primary liver cancers [1]. The incidence and mortality of iCCA have increased worldwide despite geographic variations during the past decades [2]. Surgical resection is the only potentially curative approach to iCCA. However, approximately 70% of patients are diagnosed with inoperable iCCA at advanced stage and are refractory to conventional chemotherapy [3]. The 5-year survival rate for these inoperable patients is lower than 10% [4]. Limited molecular understanding of iCCA has hindered the development of effective targeted therapies. Previously, genetic analyses of iCCA are very few and often restricted to selected oncogenes and tumor suppressors. The introduction of next-generation sequencing (NGS) to oncology practice has enabled better understanding of genetic landscapes and promoted identification of potential actionable genetic alterations in iCCA. iCCA is a distinct hepatobiliary malignancy that differs from hepatocellular carcinoma (HCC), hilar and distal bile duct cholangiocarcinoma in clinicopathologic and molecular features [4], and thus requires a distinct investigation. Herein, we briefly explored NGS-based genetic characterizations in iCCA as well as their impacts on patient prognosis and molecular targeted therapy.

Cancers often harbor multiple genetic alterations including somatic gene mutations, translocations and copy number variations (CNVs), but only one or few are responsible for induction and maintenance of tumorigenesis. These are referred to as “driver mutations”, while the remaining “passenger mutations” may have a more nuanced impact. Molecular pathogenesis of iCCA is a highly complex process involving multiple genetic alterations and signaling pathway changes. Although it is not fully understood, the genomic landscape of iCCA revealed by NGS has facilitated the identification of novel driver mutations and certification of previously-recognized ones.

Potential driver somatic mutations in iCCA

The potential driver mutations were listed in Table 1 after analyzing the results of NGS on approximately 1,000 iCCA cases [5-19]. Mutations in common oncogenes and oncosuppressors previously reported in various tumors including KRAS, BRAF, EGFR, PI3CA, PTEN, TP53 have also been identified in iCCA. KRAS and TP53 are among the most frequent mutated genes in iCCA. Michael et al. have successfully developed a genetically engineered mouse model of iCCA by incorporating tissue-specific activation of Kras G12D and deletion of p53 [20]. The prevalence of KRAS mutations in patients with iCCA varied considerably ranging from 4 to 24% [5-16]. Activating mutations of KRAS were associated with a worse outcome in iCCA [6,7]. EGFR is one of the major receptors that mediate activation of RAS/RAF/MEK/ERK signaling pathway. EGFR mutations occurred relatively rarely in iCCA with a frequency of 0-7.4% [5-9]. An addition of an orally active tyrosine-kinase inhibitor of EGFR erlotinib to traditional chemotherapy has proved to significantly prolong median progression-free survival in patients with cholangiocarcinoma including iCCA [21]. Rapid and robust anti-tumor activity of erlotinib has also been reported in an iCCA patient with somatic nonsense mutation in ERBB receptor feedback inhibitor 1 (ERRFI1), a direct negative regulator of EGFR activation [18]. However, KRAS mutations have been identified as a predictor for poor response to anti-EGFR therapy in colorectal cancer [22,23]. Fortunately, various KRAS-driven cancers have been reported to respond to MEK inhibitors [24-27], and similar effect may also present in iCCA. Elumetinib, a small-molecule kinase inhibitor against MEK1/2, is currently under investigation about its therapeutic effect on patients with iCCA (NCT00553332). Mutations in genes coding for other components of RAS/RAF/MEK/ERK pathway including NRAS, BRAF, ARAF, MAP3K1, MAP2K1 have also been described [5-11,16,18,19]. Ras-induced oncogenesis could be promoted by interaction between a unique MAPK p38γ (also known as MAPK12) and its specific phosphatase PTPN3 (also known as PTPH1) [28,29]. We have previously identified new PTPN3 mutations in 51 iCCA patients after sequencing a cohort of 124 Chinese iCCA patients [15]. Gain-of-function mutations in PTPN3 promoted proliferation and migration of iCCA cells in vitro, and correlated with increased postoperative recurrence in iCCA patients. More recently, the reciprocal allosteric regulation of p38γ and PTPN3 involving a PDZ domain-modulated complex has been successfully defined, which will promote the development of structure-based anticancer drug targeting the PTPN3-p38γ interaction [30]. We have also reported that 8.06% of iCCA patients from the same cohort harbored mutations in MAP3K4, mostly inactivating mutations [31]. Interestingly, MAP3K4 served as a putative tumor suppressor and its deficiency promoted invasiveness of iCCA via induction of epithelial-mesenchymal transition (EMT) mediated by p38/NF-κB/snail signaling pathway. Accordingly, MAPK could serve as a novel marker for risk stratification of iCCA. PI3K/AKT/mTOR signaling pathway is another pathway frequently activated by EGFR and mutations in genes coding for its components have also been reported [5-12,18,19]. The therapeutic effect of mTOR inhibitor everolimus, which has been approved for use in renal carcinoma, on advanced cholangiocarcinoma is being studied in a Phase II clinical trial (NCT00973713). In addition, AKT inhibitor MK2206 applied to treat patients with advanced refractory biliary cancer including iCCA is also currently under investigation (NCT01425879).

Table 1.

Recurrent mutations in iCCAs based on next generation sequencing

Pathway Target Prevalence of alterations (%)* Reference (s)
P53 TP53 159/1015 (15.7) [5-13,15,16,18,19]
Ras/Raf/MEK/ERK pathway EGFR 11/492 (2.2) [5-9]
KRAS 122/1283(9.5) [5-16,18,19]
NRAS 21/576 (3.6) [5-11,18,19]
BRAF 24/723 (3.3) [5-11,16,18,19]
P38/MAPK pathway PTPN3 51/124 (41.1) [15]
MAP3K4 10/124 (8.1) [31]
PI3K/mTOR pathway PTEN 26/597 (4.4) [5-12,18,19]
PIK3CA 26/597 (4.4) [5-12,18,19]
Metabolic pathway IDH1 121/1094 (11.0) [5-8,10-14,17-19]
IDH2 53/1094 (4.8) [5-8,10-14,17-19]
Chromatin remodeling ARID1A 72/501 (14) [5-8,11-13,18,19]
PBRM1 34/501 (6.8) [5-8,11-13,18,19]
BAP1 44/501 (8.8) [5-8,11-13,18,19]
*

The percentage has been calculated by considering the number of samples presenting the somatic mutation over the total number of samples analyzed in all cohorts (discovery and prevalence set of samples).

Abbreviations: TP53, tumor protein P53; MEK, mitogen-activated protein kinase/extracellular signal-regulated kinase kinase; ERK, extracellular regulated protein kinase; EGFR, epidermal growth factor receptor; MPAK, mitogen-activated protein kinase; PTPN3, tyrosine phosphatase non-receptor type 3; MAP3K4, mitogen-activated protein kinase kinase kinase 4; PI3K, phosphatidylinositol 3-kinase; mTOR, mammalian target of rapamycin; AKT, protein kinase B; PTEN, phosphatase and tensin homolog deleted on chromosome ten; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; IDH1, isocitrate dehydrogenase 1; IDH2, isocitrate dehydrogenase 2; ARID1A, AT-rich interactive domain-containing protein 1A; PBRM1, Protein polybromo-1; BAP1, BRCA1 associated protein-1.

IDH1 and IDH2 encoding the NADP+-dependent isocitrate dehydrogenases represent the most frequently mutated metabolic genes in human cancers [32-35]. Mutations in IDH1/2 occurred to approximately 10-20% of iCCA cases, which had not been identified in HCC [5-8,10-14,17-19]. Recurrent mutations in IDH1/2 resulted in the conversion of normal metabolite α-ketoglutarate (α-KG) to oncogenic counterpart R(-)-2-hydroxyglutarate (2HG), which inhibited the activity of multiple α-KG-dependent dioxygenases [36-38]. According to the study from Saha et al. [39], mutant IDH blocked liver progenitor cells from undergoing hepatocyte differentiation through accumulation of 2HG and suppression of HNF-4α, and promoted the development of iCCA with cooperative function of activated Kras. IDH1/2 mutations were associated with longer overall survival and were independent factors for longer time to tumor recurrence after surgical resection of iCCAs, suggesting a molecular sub-class with a better prognosis [14]. Circulating 2HG, a potential surrogate biomarker in iCCA, has found to be directly correlated with the tumor burden of iCCA [40]. Serum level of 2HG ≥170 ng/ml could predict the presence of IDH1/2 mutation in iCCA with high sensitivity and specificity [40]. Specific IDH1 inhibitor AG-120 has shown a promising therapeutic effect on acute myeloid leukemia (AML) harboring IDH1 mutation in a phase I clinical trial. Whether patients with iCCA can gain parallel benefit from specific inhibitors to IDH1 (AG-120) and IDH2 (AG-221) are currently being investigated in a phase I (NCT02073994) and a phase I/II (NCT02273739) clinical trial respectively.

Chromatin remodeling genes regulate the organization of DNA into chromatin, and disruption of their expression can interfere with the control of gene expression. Approximately 25-50% of iCCAs had genetic alterations involving at least one of the chromatin modulating genes (i.e. BAP1, ARID1A and PBRM1) [5-8,11-13]. ARID1A encodes the AT-rich interactive domain-containing protein 1A, which is a member of the SWI/SNF chromatin-remodeling complexes. PBRM1 also encodes component of the SWI/SNF-B (PBAF) chromatin-remodeling complexes whereas BAP1 encodes a nuclear deubiquitinase. ARID1A, PBRM1 and BAP1 functioned as tumor suppressors and were enriched in inactivating mutations in iCCA [12]. Subjects with mutations in any one of these genes tended to have shorter survival time than subjects in whom all three genes were wild type [12]. Besides, BAP1 mutations were also associated with an increased risk of early postoperative tumor recurrence [6,11]. These mutations may qualify additional therapies for iCCA, as drugs targeting chromatin remodeling like histone deacetylase (HDAC) inhibitors may be of therapeutic benefit [41]. Mutations in genes involved in DNA repair pathway like MSH6, BRCA1, ATM, MLH1 and other novel genes like GNAS, SMAD4 have also been identified [5,13].

Actionable gene fusions in iCCA

Somatic gene fusions can drive the development of human cancers and function as druggable targets, though their translational relevance has been mostly limited to hematological malignancies. Recently, various studies have revealed that more than 10% of iCCAs featured fibroblast growth factor receptor 2 (FGFR2) fusions including FGFR2-PPHLN, FGFR2-AHCYL1, FGFR2-BICC1, FGFR2-KIAA1598, FGFR2-TACC3, FGFR2-NOL4, FGFR2-PARK2, FGFR2-MGEA5 [6,7,18,19]. FGFR2 encodes a member of the fibroblast growth factor receptor family. Constitutive activation of FGFR2 and downstream MAP kinase ERK1/2 by FGFR2-AHCYL1, FGFR2-BHCC1 and FGFR2-PPHLN1 fusions have been proven in vitro and in vivo [19]. FGFR2 fusions were detected exclusively in iCCA, rarely in colorectal cancer (1/149 in Arai et al.) and HCC (0/100 in Daniela S et al. and 1/96 in Arai et al.) and none in gastric cancer (0/212 in Arai et al.), indicating a relative specificity of FGFR2 fusions in iCCA [19,42]. In contrast, the majority of HCC cases involved in aberrant FGF signaling were characteristic of FGF19 amplification, which drove carcinogenesis via persistent activation of FGF19/FGFR4 signaling rather than FGFR2 signaling [43]. Selective FGFR kinase inhibitors (BGJ398 and PD173074) successfully inhibited the oncogenic activity of FGFR2 gene fusions in vitro and might represent as a potential therapeutic target in iCCA [19]. A multi-center, open label, single arm phase II study evaluating anti-tumor activity of BGJ398 in advanced or metastatic cholangiocarcinoma patients with FGFR genetic alterations is ongoing (NCT02150967). A phase I/II study of ARQ 087, which preferentially inhibits FGFR family, has also been launched in advanced solid tumors with FGFR alterations (NCT01752920). Regorafenib, which inhibits multiple kinases including FGFR2, has been approved for treatment of partial metastatic colorectal carcinoma and its clinical implication in iCCAs has been under way (NCT02115542).

Besides FGFR2 fusions, c-ros oncogene 1 receptor tyrosine kinase (ROS1) fusions have been identified in 8.7% of iCCAs [44]. ROS1 encodes an orphan receptor tyrosine kinase related to anaplastic lymphoma kinase (ALK) and can be continuously activated by chromosomal rearrangement. FIG-ROS identified in iCCA patients was validated as a potent oncogene in an orthotopic allograft mouse iCCA model, and could accelerate tumor onset and form an aggressive and metastatic subtype with cooperation of Kras G12D and mutant p53 [45]. Crizotinib, a small-molecule tyrosine kinase inhibitor of ALK, ROS1 and hepatocyte growth factor receptor (HGFR, also known as c-MET), has showed marked antitumor activity in patients with advanced ROS1-rearranged non-small-cell lung cancer (NSCLC) regardless of the type of ROS1 rearrangement [46]. Ceritinib (LDK378) with similar mechanism to crizotinib is being investigated about its therapeutic effect on ROS1 and/or ALK over-expressed advanced iCCAs (NCT02374489) as well as tumors with ROS1 or ALK aberrations (NCT02186821). Novel gene fusions including RABGAP1L-NTRK1 have also been reported with clinical implication currently undefined [7].

Copy number variations in iCCA

Recurrent focal amplifications or deletions in iCCA based on NGS were summarized in Table 2 [6,7,16]. Daniela el al. identified 5 regions with high-level amplification which affected 23% (30/128) of iCCAs [16]. One such amplification at 11q13.2 including 6 genes, for example, CCND1, FGF family members, and oral cancer overexpressed 1 (ORAOV1), was identified in 4% of iCCAs [16]. Notably, overexpression of ORAOV1 was also found in HCC with 11q13 high-level amplification, suggesting that this oncogene could possibly be a common driver in liver carcinogenesis [47]. MET and FGF amplifications have also been described in iCCA, wherein MET amplification has been associated with poor clinical outcome [6,7,48]. A phase II study of cabozantinib, which blocks c-MET and vascular endothelial growth factor receptor 2 (VEGFR2) signaling pathways, has been under investigation about its effect on patients with advanced iCCA after progression on systemic therapy (NCT01954745). Patients with FGF or MET amplifications may predict sensitivity to their inhibitors respectively, which may serve as biomarkers for precision medicine accordingly. Copy number alterations in chromosomal regions encoding genes related to MAPK, Wnt, TGF-β pathway were also noted via integrative analysis of transcriptional regulatory network (TRN) and CNV [49]. Focal deletions at 9p21.3, which harbored tumor suppressors CDKN2A and CDKN2B, were observed in 7%-18% of iCCAs [6,7,16]. Tumors with loss of CDKN2A/CDKN2B locus or with CDK6 amplification may respond to Ckd4/6 inhibitors. Indeed, one of these agents palbociclib has presented a favorable outcome in patients with hormone-receptor-positive advanced breast cancer and deserves further study in patients with iCCA [50]. Other copy number variations including copy number gain for chromosome 1 q, 7 p and loss for 6 q, 9 p, 3 p, 13 q, 14 q and 8 p were also identified in more than 20% of iCCAs [16].

Table 2.

Recurrent focal amplifications or deletions in iCCAs based on next generation sequencing

Pathway Target Prevalence of alteration (%)* Reference (s)
High-level amplification
HGF signaling c-MET 2/28 (3.6) [7]
FGF signaling FGF 19 6/202 (3.0) [6,16]
Cell cycle control CKD6 3/102 (2.9) [6,7]
CCND1 6/202 (3.0) [6,16]
Homozygous deletion
Cell cycle control CDKN2A 31/202 (15.3) [6,7,16]
CDKN2B 28/202 (13.9) [6,7,16]
*

The percentage has been calculated by considering the number of samples presenting the copy number variation over the total number of samples analyzed in all cohorts (discovery and prevalence set of samples).

Abbreviations: HGF, hepatocyte growth factor; c-MET, hepatocyte growth factor receptor; FGF, fibroblast growth factor; CKD6, cyclin-dependent kinase 6; CCND1, cyclin D1; CDKN2A, cyclin-dependent kinase inhibitor 2A; CDKN2B, cyclin-dependent kinase inhibitor 2B.

Genetic heterogeneity in iCCA

Although agents targeting the above genetic alterations (summarized in Table 3 and Figure 1) may bring out promising results, the genetic characteristics of iCCA have further been complicated by prominent genetic heterogeneity. In fact, the prevalence of the genetic alterations varied widely among studies, partly reflecting interpatient tumor heterogeneity, also known as population heterogeneity. This might be related to the specific context where a tumor grows as well as other host factors like germ line variants. Compared with iCCAs with normal liver, iCCAs with chronic advanced liver diseases (viral, alcoholic, autoimmune and cryptogenic pre-cirrhosis or cirrhosis) featured significantly lower overall mutation rates and lower mutation rates of KRAS, MLH1, GNAS but higher EGFR mutation rate [9]. And mutations in PIK3CA, PTEN, CDKN2A and TP53 harbored exclusively in iCCAs with normal liver. Liver fluke infection by O. viverrini contributes to a major risk factor for iCCAs in endemic regions. Whole-exome sequencing analysis of mutation patterns in O. Viverrini-related and non-O. Viverrini-related iCCAs identified more frequent mutations of BAP1, IDH1/2 in non-O-viverrini-related iCCAs, whereas TP53, KRAS, SMAD4 were more frequently mutated in O. viverrini-related iCCAs [13]. Although approximately 70% of iCCA patients harbored at least one actionable alterations, no single gene was mutated in >25% of resected iCCA tissues, suggesting a need for broad-based mutational profiling in these patients for targeted therapy [5,7,19].

Table 3.

New target therapies under evaluation in clinical trials in iCCA*

Drug Phase Target Trial enrichment Primary endpoint
Elumetinib II MEK1/2 No DCR
Everolimus II mTOR No DCR
MK2206 II AKT No ORR
AG-120 I IDH1 IDH1 mutation MTD
AG-221 I/II IDH2 IDH2 mutation MTD, TTP
BGJ398 II FGFR 1-4 FGFR alterations ORR
ARQ 087 I/II FGFR 1-4 FGFR alterations Safety, TTP
Regorafenib II VEGFR1-3, TIE-2, MAPK, FGFR-1 No OS
Crizotinib II ALK, ROS1, MET ROS1 or ALK over-expression/genetic alterations DCR
Cabozantinib II c-MET, VEGFR2 No PFS
*

The data were accessed in November 2015 on the ClinicalTrials.gov online database.

Abbreviations: DCR, disease control rate; ORR, overall response rate; MTD, maximum-tolerated dose; TTP, time to progression; OS, overall survival; PFS, progression free survival.

Figure 1.

Figure 1

Recurrent genetic alterations and targeted therapies under investigation in iCCA. Genetic alterations in tyrosine kinase receptors and downstream RAS/MAPK, PI3K/AKT/mTOR effector casades result in aberrant modulation of proliferation, apoptosis and cell cycle. Newly identified mutations in MAP3K4 lead to EMT process in iCCA through p38/NF-kB signaling pathway. Mutated IDH1/IDH2 result in the accumulation of oncogenic metabolite 2HG. Potential targeted therapies against the above alterations are currently under clinical trials. Recurrently mutated chromatin remodeling genes also present as druggable targets.

Despite marked population heterogeneity, intratumor subclonal heterogeneity makes the targeted therapy for iCCA even more complex [51-55]. Tumor progression is an evolutionary process, which leads to another two types of tumor heterogeneity -- spatial and temporal heterogeneity. Spatial heterogeneity exists among subclones separated across the different topograohic regions of the primary tumor (intratumoral heterogeneity) and the same metastasis (intrametastatic heteriogeneity) as well as among metastases (intermetastatic heterogeneity). A snapshot of a small subset of neoplastic cells at a given moment by single tumor biopsy results in an underestimation of the tumor genomic landscape and a possibly biased selection of targeted medicine [53]. Sequencing of multiple tumor sites at different time points and matching the therapy to the identified actionable targets will probably represent the best approach in iCCA as this malignancy is characterized by multifocal lesions even at diagnosis. Recently, Shi et al. applied multiregional whole-exome sequencing in a patient with synchronous two HCC and one iCCA as well as two postoperative recurrent tumors, which successfully delineated the clonality and heterogeneity [56]. Genotyping circulating tumor DNA (ctDNA) with the same genetic alterations to the tumor itself, also known as “liquid biopsy”, may represent an effective and less invasive strategy to track clonal evolution and monitor drug resistance during tumor progression [57].

Future prospects

The translation of NGS into oncology practice has promoted the development of precision medicine, which tailors anticancer therapy to each individual based on the genetic landscape. Compared with the traditional therapy focused on the primary site of origin of a tumor, precision medicine endeavors to identify actionable genetic alterations. There also comes a new and evolving form of clinical trial design, the so-called “basket trials” which assign patients to specific treatment arm based on the genetic findings of NGS regardless of histologic subtypes [58]. As to iCCA individuals, selection of an effective targeted therapy is much more challenging because of the presence of extremely marked genetic heterogeneity. Fortunately, NGS has offered an opportunity to identify candidate actionable mutations. But whether iCCA patients can benefit from tailored targeted therapy needs further clarification by clinical trials, especially those designed to provide treatment based on genetic findings. More recently, Adam et al. have proposed a new strategy for precision medicine, which additionally applies next-generation functional diagnostics, such as measuring tumor cell death from biopsies after ex vivo drug exposure [59]. As a result, functional testing by providing the response of patient cells without a prior knowledge of the mechanism of drug activity can complement NGS in identification of responders to the targeted therapy. However, its clinical application needs further studies to assess the validity. Besides genetic heterogeneity, environmental risk factors, epithelial-to-stromal cross-talk, epigenetic alterations also contribute to the pathogenesis of iCCA and should be integrated to form a more individualized target approach.

Acknowledgements

This study was supported in part by National Natural Science Foundation of China. Grant Number: 81372648, 81522036, and 81572292.

Declaration of conflict of interest

None.

References

  • 1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
  • 2.Yang JD, Kim B, Sanderson SO, Sauver JS, Yawn BP, Larson JJ, Therneau TM, Roberts LR, Gores GJ, Kim WR. Biliary tract cancers in Olmsted County, Minnesota, 1976-2008. Am J Gastroenterol. 2012;107:1256–1262. doi: 10.1038/ajg.2012.173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tan JC, Coburn NG, Baxter NN, Kiss A, Law CH. Surgical management of intrahepatic cholangiocarcinoma--a population-based study. Ann Surg Oncol. 2008;15:600–608. doi: 10.1245/s10434-007-9627-x. [DOI] [PubMed] [Google Scholar]
  • 4.Khan SA, Davidson BR, Goldin RD, Heaton N, Karani J, Pereira SP, Rosenberg WM, Tait P, Taylor-Robinson SD, Thillainayagam AV, Thomas HC, Wasan H British Society of Gastroenterology. Guidelines for the diagnosis and treatment of cholangiocarcinoma: an update. Gut. 2012;61:1657–69. doi: 10.1136/gutjnl-2011-301748. [DOI] [PubMed] [Google Scholar]
  • 5.Simbolo M, Fassan M, Ruzzenente A, Mafficini A, Wood LD, Corbo V, Melisi D, Malleo G, Vicentini C, Malpeli G, Antonello D, Sperandio N, Capelli P, Tomezzoli A, Iacono C, Lawlor RT, Bassi C, Hruban RH, Guglielmi A, Tortora G, de Braud F, Scarpa A. Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget. 2014;5:2839–2852. doi: 10.18632/oncotarget.1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, Zinner R, Hong D, Meric-Bernstam F, Janku F, Crane CH, Mishra L, Vauthey JN, Wolff RA, Mills G, Javle M. Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PLoS One. 2014;9:e115383. doi: 10.1371/journal.pone.0115383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ross JS, Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, Lee HJ, Sheehan CE, Otto GA, Palmer G, Yelensky R, Lipson D, Morosini D, Hawryluk M, Catenacci DV, Miller VA, Churi C, Ali S, Stephens PJ. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist. 2014;19:235–242. doi: 10.1634/theoncologist.2013-0352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fujimoto A, Furuta M, Shiraishi Y, Gotoh K, Kawakami Y, Arihiro K, Nakamura T, Ueno M, Ariizumi S, Nguyen HH, Shigemizu D, Abe T, Boroevich KA, Nakano K, Sasaki A, Kitada R, Maejima K, Yamamoto Y, Tanaka H, Shibuya T, Shibata T, Ojima H, Shimada K, Hayami S, Shigekawa Y, Aikata H, Ohdan H, Marubashi S, Yamada T, Kubo M, Hirano S, Ishikawa O, Yamamoto M, Yamaue H, Chayama K, Miyano S, Tsunoda T, Nakagawa H. Whole-genome mutational landscape of liver cancers displaying biliary phenotype reveals hepatitis impact and molecular diversity. Nat Commun. 2015;6:6120. doi: 10.1038/ncomms7120. [DOI] [PubMed] [Google Scholar]
  • 9.Jang S, Chun SM, Hong SM, Sung CO, Park H, Kang HJ, Kim KP, Lee YJ, Yu E. High throughput molecular profiling reveals differential mutation patterns in intrahepatic cholangiocarcinomas arising in chronic advanced liver diseases. Mod Pathol. 2014;27:731–739. doi: 10.1038/modpathol.2013.194. [DOI] [PubMed] [Google Scholar]
  • 10.Zhu AX, Borger DR, Kim Y, Cosgrove D, Ejaz A, Alexandrescu S, Groeschl RT, Deshpande V, Lindberg JM, Ferrone C, Sempoux C, Yau T, Poon R, Popescu I, Bauer TW, Gamblin TC, Gigot JF, Anders RA, Pawlik TM. Genomic profiling of intrahepatic cholangiocarcinoma: refining prognosis and identifying therapeutic targets. Ann Surg Oncol. 2014;21:3827–3834. doi: 10.1245/s10434-014-3828-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jiao Y, Pawlik TM, Anders RA, Selaru FM, Streppel MM, Lucas DJ, Niknafs N, Guthrie VB, Maitra A, Argani P, Offerhaus GJ, Roa JC, Roberts LR, Gores GJ, Popescu I, Alexandrescu ST, Dima S, Fassan M, Simbolo M, Mafficini A, Capelli P, Lawlor RT, Ruzzenente A, Guglielmi A, Tortora G, de Braud F, Scarpa A, Jarnagin W, Klimstra D, Karchin R, Velculescu VE, Hruban RH, Vogelstein B, Kinzler KW, Papadopoulos N, Wood LD. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet. 2013;45:1470–1473. doi: 10.1038/ng.2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zou S, Li J, Zhou H, Frech C, Jiang X, Chu JS, Zhao X, Li Y, Li Q, Wang H, Hu J, Kong G, Wu M, Ding C, Chen N, Hu H. Mutational landscape of intrahepatic cholangiocarcinoma. Nat Commun. 2014;5:5696. doi: 10.1038/ncomms6696. [DOI] [PubMed] [Google Scholar]
  • 13.Chan-On W, Nairismagi ML, Ong CK, Lim WK, Dima S, Pairojkul C, Lim KH, McPherson JR, Cutcutache I, Heng HL, Ooi L, Chung A, Chow P, Cheow PC, Lee SY, Choo SP, Tan IB, Duda D, Nastase A, Myint SS, Wong BH, Gan A, Rajasegaran V, Ng CC, Nagarajan S, Jusakul A, Zhang S, Vohra P, Yu W, Huang D, Sithithaworn P, Yongvanit P, Wongkham S, Khuntikeo N, Bhudhisawasdi V, Popescu I, Rozen SG, Tan P, Teh BT. Exome sequencing identifies distinct mutational patterns in liver fluke-related and non-infection-related bile duct cancers. Nat Genet. 2013;45:1474–1478. doi: 10.1038/ng.2806. [DOI] [PubMed] [Google Scholar]
  • 14.Wang P, Dong Q, Zhang C, Kuan PF, Liu Y, Jeck WR, Andersen JB, Jiang W, Savich GL, Tan TX, Auman JT, Hoskins JM, Misher AD, Moser CD, Yourstone SM, Kim JW, Cibulskis K, Getz G, Hunt HV, Thorgeirsson SS, Roberts LR, Ye D, Guan KL, Xiong Y, Qin LX, Chiang DY. Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas. Oncogene. 2013;32:3091–3100. doi: 10.1038/onc.2012.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gao Q, Zhao YJ, Wang XY, Guo WJ, Gao S, Wei L, Shi JY, Shi GM, Wang ZC, Zhang YN, Shi YH, Ding J, Ding ZB, Ke AW, Dai Z, Wu FZ, Wang H, Qiu ZP, Chen ZA, Zhang ZF, Qiu SJ, Zhou J, He XH, Fan J. Activating mutations in PTPN3 promote cholangiocarcinoma cell proliferation and migration and are associated with tumor recurrence in patients. Gastroenterology. 2014;146:1397–1407. doi: 10.1053/j.gastro.2014.01.062. [DOI] [PubMed] [Google Scholar]
  • 16.Sia D, Hoshida Y, Villanueva A, Roayaie S, Ferrer J, Tabak B, Peix J, Sole M, Tovar V, Alsinet C, Cornella H, Klotzle B, Fan JB, Cotsoglou C, Thung SN, Fuster J, Waxman S, Garcia-Valdecasas JC, Bruix J, Schwartz ME, Beroukhim R, Mazzaferro V, Llovet JM. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes. Gastroenterology. 2013;144:829–840. doi: 10.1053/j.gastro.2013.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kipp BR, Voss JS, Kerr SE, Barr Fritcher EG, Graham RP, Zhang L, Highsmith WE, Zhang J, Roberts LR, Gores GJ, Halling KC. Isocitrate dehydrogenase 1 and 2 mutations in cholangiocarcinoma. Hum Pathol. 2012;43:1552–1558. doi: 10.1016/j.humpath.2011.12.007. [DOI] [PubMed] [Google Scholar]
  • 18.Borad MJ, Champion MD, Egan JB, Liang WS, Fonseca R, Bryce AH, McCullough AE, Barrett MT, Hunt K, Patel MD, Young SW, Collins JM, Silva AC, Condjella RM, Block M, McWilliams RR, Lazaridis KN, Klee EW, Bible KC, Harris P, Oliver GR, Bhavsar JD, Nair AA, Middha S, Asmann Y, Kocher JP, Schahl K, Kipp BR, Barr Fritcher EG, Baker A, Aldrich J, Kurdoglu A, Izatt T, Christoforides A, Cherni I, Nasser S, Reiman R, Phillips L, McDonald J, Adkins J, Mastrian SD, Placek P, Watanabe AT, Lobello J, Han H, Von Hoff D, Craig DW, Stewart AK, Carpten JD. Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma. PLoS Genet. 2014;10:e1004135. doi: 10.1371/journal.pgen.1004135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sia D, Losic B, Moeini A, Cabellos L, Hao K, Revill K, Bonal D, Miltiadous O, Zhang Z, Hoshida Y, Cornella H, Castillo-Martin M, Pinyol R, Kasai Y, Roayaie S, Thung SN, Fuster J, Schwartz ME, Waxman S, Cordon-Cardo C, Schadt E, Mazzaferro V, Llovet JM. Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nat Commun. 2015;6:6087. doi: 10.1038/ncomms7087. [DOI] [PubMed] [Google Scholar]
  • 20.O’Dell MR, Huang JL, Whitney-Miller CL, Deshpande V, Rothberg P, Grose V, Rossi RM, Zhu AX, Land H, Bardeesy N, Hezel AF. Kras(G12D) and p53 mutation cause primary intrahepatic cholangiocarcinoma. Cancer Res. 2012;72:1557–1567. doi: 10.1158/0008-5472.CAN-11-3596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lee J, Park SH, Chang HM, Kim JS, Choi HJ, Lee MA, Jang JS, Jeung HC, Kang JH, Lee HW, Shin DB, Kang HJ, Sun JM, Park JO, Park YS, Kang WK, Lim HY. Gemcitabine and oxaliplatin with or without erlotinib in advanced biliarytract cancer: a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2012;13:181–188. doi: 10.1016/S1470-2045(11)70301-1. [DOI] [PubMed] [Google Scholar]
  • 22.Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, Valtorta E, Schiavo R, Buscarino M, Siravegna G, Bencardino K, Cercek A, Chen CT, Veronese S, Zanon C, Sartore-Bianchi A, Gambacorta M, Gallicchio M, Vakiani E, Boscaro V, Medico E, Weiser M, Siena S, Di Nicolantonio F, Solit D, Bardelli A. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532–536. doi: 10.1038/nature11156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, Kinzler KW, Oliner KS, Vogelstein B. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540. doi: 10.1038/nature11219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R, Hewitt JF, Zak M, Peck A, Orr C, Merchant M, Hoeflich KP, Chan J, Luoh SM, Anderson DJ, Ludlam MJ, Wiesmann C, Ultsch M, Friedman LS, Malek S, Belvin M. Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers. Nature. 2013;501:232–236. doi: 10.1038/nature12441. [DOI] [PubMed] [Google Scholar]
  • 25.Hamidi H, Lu M, Chau K, Anderson L, Fejzo M, Ginther C, Linnartz R, Zubel A, Slamon DJ, Finn RS. KRAS mutational subtype and copy number predict in vitro response of human pancreatic cancer cell lines to MEK inhibition. Br J Cancer. 2014;111:1788–1801. doi: 10.1038/bjc.2014.475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Irving J, Matheson E, Minto L, Blair H, Case M, Halsey C, Swidenbank I, Ponthan F, Kirschner-Schwabe R, Groeneveld-Krentz S, Hof J, Allan J, Harrison C, Vormoor J, von Stackelberg A, Eckert C. Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition. Blood. 2014;124:3420–3430. doi: 10.1182/blood-2014-04-531871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, Maira M, McNamara K, Perera SA, Song Y, Chirieac LR, Kaur R, Lightbown A, Simendinger J, Li T, Padera RF, Garcia-Echeverria C, Weissleder R, Mahmood U, Cantley LC, Wong KK. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14:1351–1356. doi: 10.1038/nm.1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hou S, Suresh PS, Qi X, Lepp A, Mirza SP, Chen G. p38gamma Mitogen-activated protein kinase signals through phosphorylating its phosphatase PTPH1 in regulating ras protein oncogenesis and stress response. J Biol Chem. 2012;287:27895–27905. doi: 10.1074/jbc.M111.335794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hou SW, Zhi HY, Pohl N, Loesch M, Qi XM, Li RS, Basir Z, Chen G. PTPH1 dephosphorylates and cooperates with p38gamma MAPK to increase ras oncogenesis through PDZmediated interaction. Cancer Res. 2010;70:2901–2910. doi: 10.1158/0008-5472.CAN-09-3229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen KE, Lin SY, Wu MJ, Ho MR, Santhanam A, Chou CC, Meng TC, Wang AH. Reciprocal allosteric regulation of p38gamma and PTPN3 involves a PDZ domain-modulated complex formation. Sci Signal. 2014;7:ra98. doi: 10.1126/scisignal.2005722. [DOI] [PubMed] [Google Scholar]
  • 31.Yang LX, Gao Q, Shi JY, Wang ZC, Zhang Y, Gao PT, Wang XY, Shi YH, Ke AW, Shi GM, Cai JB, Liu WR, Duan M, Zhao YJ, Ji Y, Gao DM, Zhu K, Zhou J, Qiu SJ, Cao Y, Tang QQ, Fan J. Mitogen-activated protein kinase kinase kinase 4 deficiency in intrahepatic cholangiocarcinoma leads to invasive growth and epithelial-mesenchymal transition. Hepatology. 2015;62:1804–1816. doi: 10.1002/hep.28149. [DOI] [PubMed] [Google Scholar]
  • 32.Amary MF, Bacsi K, Maggiani F, Damato S, Halai D, Berisha F, Pollock R, O’Donnell P, Grigoriadis A, Diss T, Eskandarpour M, Presneau N, Hogendoorn PC, Futreal A, Tirabosco R, Flanagan AM. IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol. 2011;224:334–343. doi: 10.1002/path.2913. [DOI] [PubMed] [Google Scholar]
  • 33.Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, Friedman H, Friedman A, Reardon D, Herndon J, Kinzler KW, Velculescu VE, Vogelstein B, Bigner DD. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360:765–773. doi: 10.1056/NEJMoa0808710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shibata T, Kokubu A, Miyamoto M, Sasajima Y, Yamazaki N. Mutant IDH1 confers an in vivo growth in a melanoma cell line with BRAF mutation. Am J Pathol. 2011;178:1395–1402. doi: 10.1016/j.ajpath.2010.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrozek K, Margeson D, Holland KB, Whitman SP, Becker H, Schwind S, Metzeler KH, Powell BL, Carter TH, Kolitz JE, Wetzler M, Carroll AJ, Baer MR, Caligiuri MA, Larson RA, Bloomfield CD. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J. Clin. Oncol. 2010;28:2348–2355. doi: 10.1200/JCO.2009.27.3730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, Edwards CR, Khanin R, Figueroa ME, Melnick A, Wellen KE, O’Rourke DM, Berger SL, Chan TA, Levine RL, Mellinghoff IK, Thompson CB. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–478. doi: 10.1038/nature10860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sasaki M, Knobbe CB, Munger JC, Lind EF, Brenner D, Brustle A, Harris IS, Holmes R, Wakeham A, Haight J, You-Ten A, Li WY, Schalm S, Su SM, Virtanen C, Reifenberger G, Ohashi PS, Barber DL, Figueroa ME, Melnick A, Zuniga-Pflucker JC, Mak TW. IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics. Nature. 2012;488:656–659. doi: 10.1038/nature11323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Losman JA, Kaelin WG Jr. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev. 2013;27:836–852. doi: 10.1101/gad.217406.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Saha SK, Parachoniak CA, Ghanta KS, Fitamant J, Ross KN, Najem MS, Gurumurthy S, Akbay EA, Sia D, Cornella H, Miltiadous O, Walesky C, Deshpande V, Zhu AX, Hezel AF, Yen KE, Straley KS, Travins J, Popovici-Muller J, Gliser C, Ferrone CR, Apte U, Llovet JM, Wong KK, Ramaswamy S, Bardeesy N. Mutant IDH inhibits HNF-4alpha to block hepatocyte differentiation and promote biliary cancer. Nature. 2014;513:110–114. doi: 10.1038/nature13441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Borger DR, Goyal L, Yau T, Poon RT, Ancukiewicz M, Deshpande V, Christiani DC, Liebman HM, Yang H, Kim H, Yen K, Faris JE, Iafrate AJ, Kwak EL, Clark JW, Allen JN, Blaszkowsky LS, Murphy JE, Saha SK, Hong TS, Wo JY, Ferrone CR, Tanabe KK, Bardeesy N, Straley KS, Agresta S, Schenkein DP, Ellisen LW, Ryan DP, Zhu AX. Circulating oncometabolite 2-hydroxyglutarate is a potential surrogate biomarker in patients with isocitrate dehydrogenase-mutant intrahepatic cholangiocarcinoma. Clin Cancer Res. 2014;20:1884–1890. doi: 10.1158/1078-0432.CCR-13-2649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ma X, Ezzeldin HH, Diasio RB. Histone deacetylase inhibitors: current status and overview of recent clinical trials. Drugs. 2009;69:1911–1934. doi: 10.2165/11315680-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 42.Arai Y, Totoki Y, Hosoda F, Shirota T, Hama N, Nakamura H, Ojima H, Furuta K, Shimada K, Okusaka T, Kosuge T, Shibata T. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology. 2014;59:1427–1434. doi: 10.1002/hep.26890. [DOI] [PubMed] [Google Scholar]
  • 43.Han ZG. Functional genomic studies: insights into the pathogenesis of liver cancer. Annu Rev Genomics Hum Genet. 2012;13:171–205. doi: 10.1146/annurev-genom-090711-163752. [DOI] [PubMed] [Google Scholar]
  • 44.Gu TL, Deng X, Huang F, Tucker M, Crosby K, Rimkunas V, Wang Y, Deng G, Zhu L, Tan Z, Hu Y, Wu C, Nardone J, MacNeill J, Ren J, Reeves C, Innocenti G, Norris B, Yuan J, Yu J, Haack H, Shen B, Peng C, Li H, Zhou X, Liu X, Rush J, Comb MJ. Survey of tyrosine kinase signaling reveals ROS kinase fusions in human cholangiocarcinoma. PLoS One. 2011;6:e15640. doi: 10.1371/journal.pone.0015640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Saborowski A, Saborowski M, Davare MA, Druker BJ, Klimstra DS, Lowe SW. Mouse model of intrahepatic cholangiocarcinoma validates FIG-ROS as a potent fusion oncogene and therapeutic target. Proc Natl Acad Sci U S A. 2013;110:19513–19518. doi: 10.1073/pnas.1311707110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shaw AT, Solomon BJ. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med. 2015;372:683–684. doi: 10.1056/NEJMc1415359. [DOI] [PubMed] [Google Scholar]
  • 47.Chiang DY, Villanueva A, Hoshida Y, Peix J, Newell P, Minguez B, LeBlanc AC, Donovan DJ, Thung SN, Sole M, Tovar V, Alsinet C, Ramos AH, Barretina J, Roayaie S, Schwartz M, Waxman S, Bruix J, Mazzaferro V, Ligon AH, Najfeld V, Friedman SL, Sellers WR, Meyerson M, Llovet JM. Focal gains of VEGFA and molecular classification of hepatocellular carcinoma. Cancer Res. 2008;68:6779–6788. doi: 10.1158/0008-5472.CAN-08-0742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Miyamoto M, Ojima H, Iwasaki M, Shimizu H, Kokubu A, Hiraoka N, Kosuge T, Yoshikawa D, Kono T, Furukawa H, Shibata T. Prognostic significance of overexpression of c-Met oncoprotein in cholangiocarcinoma. Br J Cancer. 2011;105:131–138. doi: 10.1038/bjc.2011.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Li L, Lian B, Li C, Li W, Li J, Zhang Y, He X, Li Y, Xie L. Integrative analysis of transcriptional regulatory network and copy number variation in intrahepatic cholangiocarcinoma. PLoS One. 2014;9:e98653. doi: 10.1371/journal.pone.0098653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Turner NC, Huang Bartlett C, Cristofanilli M. Palbociclib in Hormone-Receptor-Positive Advanced Breast Cancer. N Engl J Med. 2015;373:1672–1673. doi: 10.1056/NEJMc1510345. [DOI] [PubMed] [Google Scholar]
  • 51.Greaves M, Maley CC. Clonal evolution in cancer. Nature. 2012;481:306–313. doi: 10.1038/nature10762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Marusyk A, Almendro V, Polyak K. Intra-tumour heterogeneity: a looking glass for cancer? Nat Rev Cancer. 2012;12:323–334. doi: 10.1038/nrc3261. [DOI] [PubMed] [Google Scholar]
  • 53.Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501:338–345. doi: 10.1038/nature12625. [DOI] [PubMed] [Google Scholar]
  • 55.Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science. 2013;339:1546–1558. doi: 10.1126/science.1235122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Shi JY, Xing Q, Duan M, Wang ZC, Yang LX, Zhao YJ, Wang XY, Liu Y, Deng M, Ding ZB, Ke AW, Zhou J, Fan J, Cao Y, Wang J, Xi R, Gao Q. Inferring the progression of multifocal liver cancer from spatial and temporal genomic heterogeneity. Oncotarget. 2016;7:2867–77. doi: 10.18632/oncotarget.6558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Diaz LA Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J. Clin. Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Redig AJ, Janne PA. Basket trials and the evolution of clinical trial design in an era of genomic medicine. J. Clin. Oncol. 2015;33:975–977. doi: 10.1200/JCO.2014.59.8433. [DOI] [PubMed] [Google Scholar]
  • 59.Friedman AA, Letai A, Fisher DE, Flaherty KT. Precision medicine for cancer with nextgeneration functional diagnostics. Nat Rev Cancer. 2015;15:747–756. doi: 10.1038/nrc4015. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Cancer Research are provided here courtesy of e-Century Publishing Corporation

RESOURCES