Biliary tract cancers (BTC), including cholangiocarcinoma and gallbladder cancer, are poor-prognosis and low-incidence cancers, although the incidence of intrahepatic cholangiocarcinoma is rising. A minority of patients present with resectable disease but relapse rates are high; benefit from adjuvant capecitabine chemotherapy has been demonstrated. Cisplatin/gemcitabine combination chemotherapy has emerged as the reference first-line treatment regimen; there is no standard second-line therapy. Selected patients may be suitable for liver-directed therapy (e.g., radioembolization or external beam radiation), pending confirmation of benefit in randomized studies. Initial trials targeting the epithelial growth factor receptor and angiogenesis pathways have failed to deliver new treatments. Emerging data from next-generation sequencing analyses have identified actionable mutations (e.g., FGFR fusion rearrangements and IDH1 and IDH2 mutations), with several targeted drugs entering clinical development with encouraging results. The role of systemic therapies, including targeted therapies and immunotherapy for BTC, is rapidly evolving and is the subject of this review.

Significance: The authors address genetic drivers and molecular biology from a translational perspective, in an intent to offer a clear view of the recent past, present, and future of BTC. The review describes a state-of-the-art update of the current status and future directions of research and therapy in advanced BTC. Cancer Discov; 7(9); 943–62. ©2017 AACR.

Biliary tract cancers (BTC), including cholangiocarcinoma [both intrahepatic (ICC) and extrahepatic (ECC)] and gallbladder cancer, are low-incidence cancers carrying a poor prognosis (1). BTCs account for approximately 3% of all adult cancers (2). Incidence and mortality are increasing, largely due to a rise in ICC (3–5). Most patients (>65%) are diagnosed with nonresectable disease (1), and there is a high relapse rate in the minority of patients who undergo potentially curative surgery (6, 7). The five-year survival rate is around 5% to 15% when considering all patients (8, 9); estimated five-year survival rate varies with stage: 50% for American Joint Committee on Cancer (AJCC) stage I, 30% for stage II, 10% for stage III, and 0% for stage IV (6, 10).

It is widely accepted by the BTC community that BTC malignancies are not one unique disease only, but a group of different diseases with distinct demographics, molecular characteristics, and treatment options (Fig. 1). Such differences are worth taking into account at time of treatment planning, research, and clinical trial design. BTCs are more frequent in patients between ages 50 and 70 years, with a male preponderance for cholangiocarcinoma and female for gallbladder cancers (2); >90% are adenocarcinomas (1). Several risk factors, mainly associated with chronic gallbladder or biliary tract inflammation, have been identified (11–13). Opistorchis viverrini is one of the three major liver trematodes (flukes) that infect humans. It is endemic in Thailand, Vietnam, Cambodia, and Laos and accounts for a global “hotspot” of intrahepatic cholangiocarcinoma in this region. Adult flukes can remain in the bile ducts for years, stimulating a host immune response, leading to chronic biliary tract inflammation. This results in an up to 15-fold increase in the risk of developing intra/extrahepatic cholangiocarcinoma (14). There are also differences in risk factors (15–17) and symptoms at presentation between the different BTCs [gallbladder patients (in advanced stages) are less likely to present with jaundice and usually present with abdominal pain; refs. 15, 18].

Figure 1.

BTCs are a group of different diseases which includes ICC, ECC, and gallbladder cancer. They differ in many aspects, such as anatomical location, demographics, clinical presentations, and treatment options.

Figure 1.

BTCs are a group of different diseases which includes ICC, ECC, and gallbladder cancer. They differ in many aspects, such as anatomical location, demographics, clinical presentations, and treatment options.

Close modal

Patients with tumors arising in proximity to the bile ducts present with biliary obstruction, due to local infiltration of the biliary tract. A minority of patients will be diagnosed with early (resectable) disease, in which case treatment will be surgical with curative intent. For patients diagnosed with advanced disease (often presenting with nonspecific, nonbiliary obstructive symptoms), treatment options are noncurative and mainly chemotherapy-based.

Despite potentially curative resection for localized disease, relapse rates are high (19), highlighting the need to optimize adjuvant strategies. The role of adjuvant treatment for BTC has been unclear for many years (20). A systematic review and meta-analysis found that adjuvant treatment did not improve survival when compared with surgery alone when considering all patients (21). However, there appeared to be benefit for patients with microscopically involved margins (R1-resection) versus clear resection margins [R0-resection; odds ratio (OR), 0.36; 95% confidence interval (CI), 0.19–0.68] and lymph node–positive disease (OR, 0.49; 95% CI, 0.30–0.80). Two randomized phase III clinical trials exploring adjuvant chemotherapy were reported in 2017. First, the results from the PRODIGE-12/ACCORD-18 clinical trial assessing the benefit of adjuvant combination chemotherapy compared with observation alone were reported in January 2017 (22). This multicenter phase III trial randomized 196 patients within 3 months of resection of a localized BTC (intrahepatic, perihilar, or extrahepatic cholangiocarcinoma, or gallbladder cancer), to receive either adjuvant gemcitabine and oxaliplatin or surveillance; there was no significant difference in relapse-free survival between the arms [HR of 0.83 (95% CI, 0.58–1.19); P = 0.31]. Second, the BilCap clinical trial exploring the role of adjuvant capecitabine compared with observation alone was reported at ASCO 2017. A total of 447 patients with BTC were randomized to capecitabine (n = 223) or observation (n = 224; ref. 23). Sensitivity analyses by intention-to-treat were adjusted to nodal status, grade of disease, and gender (447 patients). This trial demonstrated benefit from capecitabine in terms of overall survival [OS; HR, 0.71 (95% CI, 0.55–0.92); P < 0.01; median OS 51 months (95% CI, 35–59) and 36 months (95% CI, 30–45) for capecitabine and observation arms, respectively]. There was also benefit from adjuvant capecitabine in terms of relapse-free survival [median 25 months (95% CI, 19–37) and 18 months (95% CI, 13–28) for capecitabine and observation arms, respectively]. Based on these results, adjuvant capecitabine is likely to be considered standard of care following surgery for BTC.

Around 60% to 70% of patients will be diagnosed with advanced disease, which is defined as inoperable or metastatic disease. For these patients, palliative treatment, usually in the form of systemic chemotherapy, is the only treatment option. Selected patients with liver-predominant disease may benefit from liver-directed therapies such as external beam radiation (24, 25) or radioembolization (26, 27). Unfortunately, data suggesting such benefit are based on retrospective series or small phase II trials rather than randomized studies; further data are awaited to confirm the incremental benefit of approaches involving liver radioembolization. Liver transplant has been suggested as a potential treatment option for patients with small perihilar cholangiocarcinoma (28, 29). However, prospective studies are needed to ensure the most suitable patient selection and benefit due to challenges for organ allocation and living donation policies; currently, liver transplant remains controversial in this setting. New options for systemic treatment are an urgent area of unmet need for this patient population.

First-Line Chemotherapy

The most active cytotoxic chemotherapy agents in the management of BTCs are gemcitabine and platinum agents (30, 31). The first study to suggest that palliative chemotherapy could improve survival and quality of life was reported in 1996 and established gemcitabine as a treatment option for patients with advanced disease (32). These results increased the interest in the treatment of BTCs, and over the past 20 years many studies have been performed (30) that have been negative or lacked the statistical power and rigor to change clinical practice.

In 2010 we showed, in the 410-patient, randomized, phase III, ABC-02 study, a benefit from cisplatin/gemcitabine chemotherapy over single-agent gemcitabine; the doublet conferred an advantage in OS over gemcitabine alone (11.7 vs. 8.1 months; HR, 0.64; 95% CI, 0.52–0.80; P < 0.001) (33). These results were replicated in the Japanese randomized phase II (BT22) study (34). Based on these results, cisplatin/gemcitabine has become the recognized reference regimen for first-line treatment of patients with advanced BTC. Although patients with jaundice were excluded from the ABC-02 study, we have since shown that the doublet may safely be used in patients with jaundice secondary to obstructing endoluminal disease (although not the case in patients with jaundice due to parenchymal replacement by metastatic disease; ref. 35). The lack of further practice-changing trials since these studies highlights the desperate need for new therapies for patients with advanced BTC.

Second-Line Chemotherapy

Patients progressing on first-line chemotherapy often have a rapidly worsening performance status, and only a small number of patients may be suitable for further treatment. In addition, patients often have the inherent problems of biliary obstruction and sepsis associated with BTC, which may preclude further treatment.

Currently, no quality evidence is available supporting the use of second-line chemotherapy (36). For most patients, active symptom control (ASC; e.g., biliary stenting and antibiotics, as appropriate) is considered the standard of care after progression on first-line chemotherapy. However, small prospective and retrospective studies have shown signals of potential benefit in selected patients (37–41). Based on the previously reported benefit from 5-fluorouracil (5-FU) in BTCs (42, 43), the ongoing UK ABC-06 study is a randomized phase III study comparing oxaliplatin and 5-FU (FOLFOX regimen) with ASC versus ASC alone following progression on or after first-line cisplatin/gemcitabine; recruitment is ongoing (NCT01926236; ref. 44).

In summary, robust (phase III) evidence is available for the use of first-line chemotherapy in patients presenting with advanced disease. Cisplatin and gemcitabine have become the reference regimen; other regimens are sometimes considered by individual clinicians based on phase II studies. The role of second-line therapy is unclear; no single regimen has emerged. Ongoing trials are trying to address this lack of treatment options, highlighting the need for development of novel targeted therapy approaches.

Tumor Sequencing Data

There has been a great effort to apply new parallel sequencing technologies to gather more insight about the molecular biology of these malignancies. As with other studies with next-generation sequencing (NGS) in cancer, the starting questions were if these malignancies share common anomalies with other cancers and what is the idiosyncratic pattern of anomalies associated with BTCs.

Main New Findings for ICC and ECC

Several studies have identified different genetic alterations that occur in cholangiocarcinoma using various approaches, from whole-exome sequencing (WES; refs. 45–47) to a focused approach on specific pathways (48–53). A WES study of 8 cases of Opisthorchis viverrini–related cholangiocarcinoma revealed mutations in TP53 (44.4%), KRAS (16.7%), and SMAD4 (16.7%; ref. 45). Loss-of-function mutations of tumor suppressor genes have been reported in cholangiocarcinoma with an overall frequency of 15% (54–59). Activating mutations of KRAS (22%, range 5%–57%), mainly located in codon 12 hotspots, have been associated with a worse prognosis after radical surgery. The tumor suppressor gene SMAD4, located in the long arm of chromosome 18q 21.1, encodes a nuclear transcription factor of TGFβ (60, 61) and is usually inactivated when mutated. In the infection-driven cases, mutations in TP53 and SMAD4 were more common, 39.8% and 19.4% versus 9.3% and 5.8%, respectively. Mutations in CDKN2A/B (p16) were identified in 7% of patients with ICC (52).

In a sequencing study of 102 patients with ICC from China, Zou and colleagues found TP53 mutations are more likely to be HBsAg-seropositive, whereas KRAS mutations are nearly exclusively found in HBsAg-seronegative patients with ICC (62).

Interesting findings include inactivating mutations in MLL3 (14.8%), ROBO2 (9.3%), RNF43 (9.3%), and PEG3 (5.6%), and activating mutations in GNAS (9.3%; ref. 45). RNF43 (encoding a RING domain E3 ubiquitin ligase) interacts with p53 and suppresses p53-mediated apoptosis (63). In this study, RNF43 was an independent prognostic factor in the multivariable analysis. Interestingly, RNF43 is also a key molecule in the WNT–β-catenin pathway and can inhibit WNT signaling by interacting with the WNT receptors of the Frizzled family (64). RNF43 mutations may predict sensitivity to porcupine inhibitors (65). PEG3 is an imprinted gene that regulates apoptosis; when it is inhibited, it blocks p53-induced apoptosis (66). Loss of PEG3 activates WNT signaling, leading to chromosomal instability (67). MLL3 encodes a histone–lysine N-methyltransferase that is one of the histone modifiers implicated in numerous cancers such as pancreatic cancer (68–70). Most of the tumors harboring a mutation in MLL3 did not contain mutations in TP53, KRAS, or SMAD4, despite the fact that these three genes were mutated together in 57% of cases. This finding suggested the possibility of a particular subtype of cholangiocarcinoma where alterations in chromatin packaging have driven the development of the disease.

ROBO2 is a receptor protein involved in activating the SLIT–ROBO pathway. These proteins are key components of the axon guidance signaling and have been recently implicated in pancreatic adenocarcinoma (71).

GNAS encodes a guanine nucleotide-binding protein alpha subunit (72), which is frequently mutated in intraductal papillary mucinous neoplasms of the pancreas and villous adenoma of the colon (73, 74).

IDH Alterations

Mutations in the isocitrate dehydrogenase genes (IDH1 and IDH2; Fig. 2; refs. 75, 76) were found more often in noninfectious cholangiocarcinomas (76). IDH1 and IDH2 mutations were also found (19%) by the Johns Hopkins group. These mutations were clustered in previously identified hotspots (codons 132 and 172, respectively; ref. 77, 78) and were associated with a worse prognosis in contrast to the previously published report (47). These differences in prognosis may be accounted for by the differing sample size and baseline characteristics of the two studies. In a Chinese study, only five (4.9%) patients with ICC harbored IDH1 mutations (62).

Figure 2.

IDH1 and IDH2 are metabolic enzymes found in the cytoplasm and mitochondria, respectively, and catalyze the decarboxylation of isocitrate to alpha-ketoglutarate (α-KG), resulting in the reduction of NADP+ to NADPH. The oncometabolite 2-hydroxyglutarate (2-HG) can competitively inhibit one or more members of the family of over 60 dioxygenases which require α-KG as a cofactor. The dioxygenases include the JmjC family of histone demethylases and the TET family of methylcytosine dioxygenase enzymes that catalyze the demethylation of DNA. IDH and KRAS mutations can cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic ICC. Agents targeting IDH1 and IDH2 are under development. m, mutant.

Figure 2.

IDH1 and IDH2 are metabolic enzymes found in the cytoplasm and mitochondria, respectively, and catalyze the decarboxylation of isocitrate to alpha-ketoglutarate (α-KG), resulting in the reduction of NADP+ to NADPH. The oncometabolite 2-hydroxyglutarate (2-HG) can competitively inhibit one or more members of the family of over 60 dioxygenases which require α-KG as a cofactor. The dioxygenases include the JmjC family of histone demethylases and the TET family of methylcytosine dioxygenase enzymes that catalyze the demethylation of DNA. IDH and KRAS mutations can cooperate to drive the expansion of liver progenitor cells, development of premalignant biliary lesions, and progression to metastatic ICC. Agents targeting IDH1 and IDH2 are under development. m, mutant.

Close modal

FGFR Pathway

Genome-wide structural analyses showed recurrent translocation events involving the FGFR2 locus (48). Wu and colleagues published the first report of FGFR fusions in ICC in 2013 with a description of 2 cases of FGFR2–BICC1 fusions (79). BICC is a negative regulator of WNT signaling (80). Tumor profiling studies of ICC have reported multiple additional fusion partners with FGFR2, including AHCYL1, TACC1, MGEA5, and PPHLN (48, 81–83), all of which fuse at a consistent breakpoint within the FGFR2 gene on chromosome 10 (48). The mechanism by which FGFR2 fusions drive oncogenesis is under active investigation. Arai and colleagues showed that in clones expressing FGFR2–BICC1 and FGFR2–AHCYL1, the MAPK pathway was activated but not AKT or STAT, suggesting that FGFR2 fusion proteins activate canonical signaling events downstream of FGFR (81). A mutation in ERRFI1, a negative regulator of the EGFR family, was found that was not present in cases having alterations of FGFR (81). ERBB receptor inhibitor-1 has a role as a negative regulator of the EGFR family of receptors (84–87). Thus, patients harboring this mutation may be suitable for an anti-EGFR treatment approach.

Another cooperative effort of sequencing intrahepatic cholangiocarcinoma tumor samples confirmed the presence of FGFR2 fusions in 3 of 28 tumor samples (10.7%). The Johns Hopkins study also found four somatic mutations in FGFR2 (13%; ref. 47).

Nakamura and colleagues conducted a comprehensive genomic analysis of 260 BTCs and found that 40% of cases harbored targetable genetic alterations (88). They found that gene fusions involving FGFR2 and PRKACA or PRKACB preferentially occurred in ICC and ECC, respectively.

Chromatin Modifiers

The Singapore group subsequently analyzed cases of infection-related versus non–infection-related cholangiocarcinoma (46). A new set of 15 non–infection-related cases were sequenced identifying mutations in chromatin-remodeling genes: ARID1A (19%) and BAP1 (25%). ARID1A encodes an accessory subunit of the SWI/SNF chromatin remodeling complex. In cell lines, silencing ARID1A results in a significant increase in the proliferation of cholangiocarcinoma-derived cell lines compared with cells expressing the wild-type form (46), showing mechanistically the involvement of this alteration in cholangiocarcinoma proliferation. BAP1 is a deubiquitinase protein of the ubiquitin C-terminal hydrolase (UCH) family (89–91). Increased proliferation was shown after BAP1 knockdown using an RNA-interference approach.

The Johns Hopkins group identified mutations in several of the chromatin-remodeling genes, including the ones described by Chan-On and colleagues (46), ARID1A and BAP1. They also found alterations in PBRM1 (17%), a gene that encodes a subunit of the ATP-dependent SWI/SNF chromatin-remodeling complexes. These findings reinforced the idea of a major role of the chromatin remodeling process in the carcinogenesis of this tumor (Fig. 3; ref. 92).

Figure 3.

Chromatin remodeling complex: DNA is packaged in chromatin to allow the 1.8 m–long human genome to fit in a single cell of the body. SWI/SNF complexes are evolutionarily conserved, ATP-dependent, molecular machines that alter local chromatin structure. ARID1A encodes an accessory subunit of the SWI/SNF chromatin remodeling complex. ARID1A, AT-rich interactive domain-containing protein 1A; BAF, BRG1 associated factor; BRD, Bromo domain containing protein; SMARC, SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamily A, also known as BRG: Brahma related gene; BAP1, BRCA1-associated protein-1 ASXL: additional sex combs-like; OGT, UDP-glucose-dependent O-glucosyltransferase; HCF1, host cell factor 1; YY1, Ying Yang 1; FOXK, Forkhead box protein K.

Figure 3.

Chromatin remodeling complex: DNA is packaged in chromatin to allow the 1.8 m–long human genome to fit in a single cell of the body. SWI/SNF complexes are evolutionarily conserved, ATP-dependent, molecular machines that alter local chromatin structure. ARID1A encodes an accessory subunit of the SWI/SNF chromatin remodeling complex. ARID1A, AT-rich interactive domain-containing protein 1A; BAF, BRG1 associated factor; BRD, Bromo domain containing protein; SMARC, SWI/SNF Related, Matrix Associated, Actin Dependent Regulator of Chromatin, Subfamily A, also known as BRG: Brahma related gene; BAP1, BRCA1-associated protein-1 ASXL: additional sex combs-like; OGT, UDP-glucose-dependent O-glucosyltransferase; HCF1, host cell factor 1; YY1, Ying Yang 1; FOXK, Forkhead box protein K.

Close modal

New Gene Fusions: NTRK

New gene fusions have also been identified in ICC, such as RABGAP1LNTRK1 (52). NTRK1 encodes a protein of the neurotrophic tyrosine kinase receptor (NTKR) family; this kinase is a membrane-bound receptor that, on neurotrophin binding, phosphorylates itself and members of the MAPK pathway (93). Gene fusions involving the NTRK1, NTRK2, and NTRK3 genes result in constitutively active TRKA, TRKB, and TRKC kinases. The presence of these kinases leads to cell differentiation and may play a role in specifying sensory neuron subtypes. The TRK inhibitor LOXO-101 has shown early promise in a phase I trial of patients with advanced solid tumors where 5 out of 6 (83%) patients evaluable for response and harboring NTRK fusions achieved a partial response (although no patients had a diagnosis of cholangiocarcinoma/gallbladder cancer; ref. 94). Other compounds targeting an NTRK1/2/3, ROS1, or ALK gene rearrangement have shown positive responses in the selected population (95). TRK inhibition is being explored in cholangiocarcinoma in a selected cohort in the STARTRK-2 phase II basket study of entrectinib (RXDX-101) in patients with solid tumors harboring an NTRK1/2/3, ROS1, or ALK gene rearrangement (NCT0256867). Sequencing studies have identified the presence of NTRK fusions in 1 of 28 (3.5%) of patients diagnosed with ICC (52). ROS1 and ALK fusions are also rare targets ICC, with a frequency of 0% to 8.7% (96, 97) and 2.6% (98), respectively.

Protein Tyrosine Phosphatases

Protein tyrosine phosphatases (PTP) are a structurally diverse family of tightly regulated enzymes. Gao and colleagues (49) found frequent mutations in PTPN3 in ICC that were significantly correlated with tumor recurrence.

Gene Profiling

Gene expression profiles, high-density single-nucleotide polymorphism arrays, and mutation analyses using formalin-fixed ICC samples from patients diagnosed with ICC identified two main biological classes of ICC (54). The first group, the inflammation class (38% of ICCs), was characterized by activation of inflammatory signaling pathways, overexpression of cytokines, and STAT3 activation. In contrast, the proliferation class (62% of ICCs) was characterized by activation of oncogenic signaling pathways and was associated with shorter survival. Andersen and colleagues also characterized ICC based on the genomic as well as transcriptomic signatures and were able to classify ICC into subclasses with different prognoses (99). Recently The Cancer Genome Atlas (TCGA) study in ICC was published, and this integrated analysis of somatic mutations, RNA expression, copy number, and DNA methylation also led to a molecular classification scheme and identified an IDH mutant–enriched subtype with distinct molecular features, including low expression of chromatin modifiers, elevated expression of mitochondrial genes, and increased mitochondrial DNA copy number (100).

Gallbladder Subset Findings

A later study sequenced gallbladder carcinoma and cholangiocarcinoma separately. The analysis of 57 tumor–normal pairs with a double approach using WES and ultra-deep sequencing of a 283-gene panel gave a striking result; mutations in the ERBB family of proteins (including their downstream genes) were found in 35.8%, and, in the multivariable analysis, these cases had a worse outcome (P = 0.001). Among the 11.8% of mutations in ERBB3, the majority of mutations were found in a hotspot in codon 104 (101). This pattern is not shared with cholangiocarcinoma, suggesting that although both tumors originate from the biliary epithelium, they are genetically distinct. No IDH mutations have been identified in gallbladder cancer (102). Regarding the classic PI3K–AKT–mTOR pathway, activating mutations in PIK3CA have been identified (12.5%; ref. 103).

In summary, the recent targeted and WES genomic analyses have enriched our understanding of the genetic landscape of BTCs and informed us on the most actionable signatures (Fig. 4). They have highlighted that (i) the genomic spectra vary significantly in different subtypes of BTCs; (ii) IDH mutations and FGFR2 fusions are the most common genetic alterations in ICC; (iii) frequent mutations occur in chromatin-remodeling genes: ARID1A, BAP1, and PBRM1; and (iv) mutation frequency in different genes varies by etiology and geographic regions.

Figure 4.

Genetic landscape of BTC. Most frequent genetic aberrations in targetable pathways of interest in BTC. The mutation is quoted as the highest to lowest with range from different reports on each mutation. Those without range come from single reports. Extracted from Desphande et al. BMC Cancer 2011 (103), Borger et al. The Oncologist 2012 (102), Voss et al. Human Pathology 2013 (204), Ross et al. The Oncologist 2014 (52), Ong et al. Nature Genetics 2012 (45), Graham et al. Human Pathology 2014 (123), Arai et al. Hepatology 2014 (81), Sia et al. Nature Communications 2015 (82), Javle et al. Cancer 2016 (53), Zou et al. Nature Communications 2014 (62), Li et al. Nature Genetics 2014 (101), Zhu et al. Annals of Surgical Oncology 2014 (116), Sia Gastroenterology 2013 (54), Jiao et al. Nature Genetics 2013 (47), Chan-On et al. Nature Genetics 2013 (46), Wang et al. Oncogene 2013 (115), Wu et al. Cancer Discovery 2013 (79), Ross et al. Journal of Clinical Oncology 2015 (51), Nakamura et al. Nature Genetics 2015 (89), Borad et al. PLoS Genetics 2014 (48), Randall et al. Journal of Clinical Oncology 2015 (50), Galdy et al. Cancer and Metastases Reviews 2016 (162), Churi et al. PlosOne 2014 (83), Turner et al. Nature Reviews in Cancer 2010 (124), Pai et al. European Journal of Cancer Prevention 2011 (199), Riener et al. Genes, Chromosomes and Cancer 2008 (208). GBCA, gallbladder cancer.

Figure 4.

Genetic landscape of BTC. Most frequent genetic aberrations in targetable pathways of interest in BTC. The mutation is quoted as the highest to lowest with range from different reports on each mutation. Those without range come from single reports. Extracted from Desphande et al. BMC Cancer 2011 (103), Borger et al. The Oncologist 2012 (102), Voss et al. Human Pathology 2013 (204), Ross et al. The Oncologist 2014 (52), Ong et al. Nature Genetics 2012 (45), Graham et al. Human Pathology 2014 (123), Arai et al. Hepatology 2014 (81), Sia et al. Nature Communications 2015 (82), Javle et al. Cancer 2016 (53), Zou et al. Nature Communications 2014 (62), Li et al. Nature Genetics 2014 (101), Zhu et al. Annals of Surgical Oncology 2014 (116), Sia Gastroenterology 2013 (54), Jiao et al. Nature Genetics 2013 (47), Chan-On et al. Nature Genetics 2013 (46), Wang et al. Oncogene 2013 (115), Wu et al. Cancer Discovery 2013 (79), Ross et al. Journal of Clinical Oncology 2015 (51), Nakamura et al. Nature Genetics 2015 (89), Borad et al. PLoS Genetics 2014 (48), Randall et al. Journal of Clinical Oncology 2015 (50), Galdy et al. Cancer and Metastases Reviews 2016 (162), Churi et al. PlosOne 2014 (83), Turner et al. Nature Reviews in Cancer 2010 (124), Pai et al. European Journal of Cancer Prevention 2011 (199), Riener et al. Genes, Chromosomes and Cancer 2008 (208). GBCA, gallbladder cancer.

Close modal

Animal Models

Some of the efforts to generate animal models were primarily focused on well-known oncogenes such as KRAS and TP53 (104). The Notch and IDH pathways seem to have an oncogenic role in cholangiocellular malignancies. A transgenic mouse model (NotchIC::AlbCre) expressing the intracellular domain of Notch receptor-1 (NICD) in the liver was able to generate cholangiocarcinomas (a similar approach was previously used by Sekiya and colleagues, ref. 105; and Fan and colleagues, ref. 106) derived from hepatic progenitors. This model describes a subtype characterized by the overexpression of the Notch pathway with a different genetic background from bile-duct derived cholangiocarcinomas (107). Interestingly, a more recent mouse model expressing the IDH2-mutant variants R140Q or R172K in adult mouse hepatocytes generated ICCs when combined with KRASG12D mice, suggesting the need for additional hits after the mutation of the IDH genes (76). IDH1/2 mutants reduced the expression of HNF4α, which is a master transcriptional regulator of hepatocyte differentiation. Notch1 activation as a transdifferentiating factor has also been observed in an animal model by Guest and colleagues (108).

A model of cholangiocarcinoma using zebrafish was generated by inducing the coexpression of viral hepatitis-B and hepatitis-C core proteins, the first animal model showing the involvement of these viral proteins in the pathogenesis of cholangiocarcinoma (109). Classic models in rodents have used a carcinogen-induced model, usually diethylnitrosamine (DEN) and thioacetamide (TAA) and infection with Opistorchis viverrini (110). Genetically engineered mouse models of cholangiocarcinoma were generated by targeting TP53, NF2, PTEN, SMAD4, and KRAS (110–112). In addition, in a transgenic mouse model, constitutive overexpression of ERBB2 in the basal layer of biliary tract epithelium led to the development of gallbladder adenocarcinoma (113).

Unfortunately, some of the key features of human disease, such as the genetic landscape, chronic inflammation, and cholestasis, are clearly underrepresented in these models. The latest published model, consisting of combining an activating mutation in KRAS and PTEN deletion, has not incorporated the new knowledge from NGS information yet (114). Little information about the involvement of the microenvironment has been generated in animal models. Furthermore, there is no model of non-gallbladder extrahepatic cholangiocarcinoma.

Targeting the Molecular Biology of BTC

A deeper understanding of the natural behavior and activated pathways involved in BTCs is required to guide the development of new drugs, aiming to improve patient outcomes. A summary of the main pathways and potential targeted therapies is shown in Fig. 5 and Table 1.

Figure 5.

Summary of the relevant pathways for BTCs. Activation links are described with black arrows. Negative links are described as red lines. Red asterisk identifies the mutated variant of the protein.

Figure 5.

Summary of the relevant pathways for BTCs. Activation links are described with black arrows. Negative links are described as red lines. Red asterisk identifies the mutated variant of the protein.

Close modal
Table 1.

Heat map summary of the status of evidence supporting known molecular biology involved in BTCs

Heat map summary of the status of evidence supporting known molecular biology involved in BTCs
Heat map summary of the status of evidence supporting known molecular biology involved in BTCs

NOTE: Available evidence is classified according to the type of research: basic/preclinical, translational, or clinical research. The table indicates if there is any evidence available (yes, gray; no, white); moreover, the evidence is ranked as follows: + (light gray, poor quantity/quality; retrospective data, absence of prospective/randomized studies), ++ (mid–dark gray color, medium quantity/quality; prospective clinical trials), and +++ (dark gray, high quantity/quality; randomized trials). Please refer to the main text for references applicable for each pathway.

IDH Metabolism

Results from the collective efforts of several groups to characterize IDH mutations have shown that (i) IDH1 mutation is more common than mutation of IDH2; (ii) the “hotspot” IDH1/2 mutations are point mutations located in the arginine 132 (R132) residue in IDH1 or the arginine 172 (R172) residue in IDH2 (47, 102, 115–118); (iii) these mutations are ubiquitously higher in ICC than in ECC (118); (iv) the prognostic significance of IDH mutations remains conflicting in cholangiocarcinoma (47, 115–117); and (v) the mutant IDH loses its normal enzymatic activity and gains a new ability to produce the oncometabolite 2-hydroxyglutarate (2-HG), which can be detected in the tumor and blood (102, 119).

Pharmacologic inhibitors highly specific to the individual IDH-mutant alleles (e.g., to IDH1R132 and IDH2R172) have been developed. These block the function of mutant IDH1 or IDH2 at nanomolar concentrations, leading to reduced 2-HG levels (Fig. 2). Rohle and colleagues found that a selective IDH1R132H inhibitor (AGI-5198) impeded the growth of IDH-mutant glioma cells (120). Similarly, Wang and colleagues demonstrated that AGI-6780 could selectively inhibit leukemic cells harboring mutant IDH2R140Q (121). In a phase I trial, AG-120 (IDH1 inhibitor; Agios) was well tolerated among patients with advanced solid tumors with IDH1 mutations (NCT02073994). There were no dose-limiting toxicities, and anemia was the most frequent grade 3 adverse event (5%). Of the 20 patients with advanced ICC, 1 patient (5%) achieved a partial response and 11 patients (55%) had stable disease. In all patients responding to AG-120, a reduction in circulating 2-HG levels ranging from 73% to 99% and a reduction in Ki67 staining ranging from 22% to 96% from baseline were observed. The expansion phase with 500 mg once daily is under way. Other IDH1 and IDH2 inhibitors have entered clinical trials recently (NCT02273739, NCT02381886, and NCT02481154) and are enrolling patients with ICC. Through a high-throughput drug screen of a large panel of cancer cell lines, including 17 BTCs, we recently found that ICC cells harboring IDH mutations exhibited a striking response to the multi-TKI dasatinib (122). In addition, dasatinib-treated IDH-mutant xenografts demonstrated pronounced apoptosis and tumor regression. A trial with dasatinib in patients with IDH-mutant advanced ICC is ongoing (NCT02428855).

FGFR

The recent discovery of recurrent FGFR2 fusions in 11% to 45% of patients with ICC, described previously, has opened a promising therapeutic avenue (52, 81–83, 123, 124). In genomic profiling studies, FGFR2 fusions are found concurrently with mutations in genes such as ARID1A, PBRM1, and TP53, among others (79). Histologically, FGFR2 fusions are associated with prominent intraductal growth and anastomosing tubular glands (123); prognostically, they appear to be associated with an indolent disease course and prolonged survival (83, 123).

The discovery of FGFR aberrations in multiple tumor types has stimulated pharmaceutical and scientific interest in the development of FGFR inhibitors. The earliest reported data of selective FGFR inhibition in cholangiocarcinoma are with the oral agent BGJ398 (infigratinib; Novartis), which has an IC50 for FGFR2 of 1.4 nmol/L. Preliminary results of 34 patients in the ongoing phase II trial of BGJ398 in advanced cholangiocarcinoma with FGFR aberrations after first-line chemotherapy (NCT02150967) included patients with FGFR2 fusions (n = 28), FGFR2 mutations (n = 2), FGFR2 amplification (n = 3), or FGFR3 amplification (n = 1); the median time on treatment was 188 days, and the objective response rate was 22% (all 8 patients with a partial response had an FGFR2 fusion; ref. 125). As seen with other oncogene-addicted tumors treated with tyrosine kinase inhibitors (TKI), acquired resistance limited the durability of response in some patients. Goyal and colleagues reported the first evidence of clinically acquired resistance to FGFR inhibition in an analysis of three patients with FGFR2 fusion–positive ICC who were treated with BGJ398 (126). Sequencing of cell-free DNA (cfDNA) and biopsy samples collected at baseline and postprogression revealed polyclonal secondary mutations in the FGFR2 kinase domain, including the gatekeeper mutation FGFR2V564F in all three patients. In vitro studies further identified structurally distinct FGFR inhibitors that may overcome the resistance, and these data may guide future treatment strategies in this scenario.

Other selective FGFR inhibitors, including INCB54828 (Incyte; NCT02924376), BAY1163877 (Bayer; NCT01976741), and the irreversible FGFR inhibitor TAS-120 (Taiho; NCT02052778), are currently being evaluated in early-phase trials in patients with advanced solid tumors, including ICC. Nonselective multi-TKIs that also target FGFR, including ponatinib and pazopanib, have demonstrated activity in individual patients with ICC who have developed resistance to chemotherapy (48). A third nonselective TKI, ARQ 087 (Arqule; NCT01752920), which inhibits RET, PDGFR, KIT, SRC, and FGFR1–3 (IC50 for FGFR2 = 0.68 nmol/L), is currently being evaluated in a phase II trial of previously treated patients with FGFR-aberrant tumors, including FGFR2 fusion–positive advanced ICC. Preliminary data from the phase I/II basket trial indicate that 3 of the 12 patients with FGFR2 fusion–positive advanced ICC treated with ARQ 087 had a partial response (with a disease control rate of 75%; ref. 127).

FGFR2 fusions appear to be driver alterations that predict sensitivity to FGFR inhibition in ICC, but the impact of the fusion partner and the sensitivity of FGFR mutations and amplifications to FGFR inhibition in ICC is yet unknown. Circulating levels of FGF ligands such as FGF19, FGF21, and FGF23 showed some correlation with response in the ARQ 087 trial (128), but further investigation into these biomarkers and others is warranted.

The safety profile of FGFR inhibitors is manageable, with hyperphosphatemia being one of the most common toxicities. This is a class effect due to on-target blockade of FGF23 in the bone and kidney. FGF23 is a phosphaturic hormone that regulates phosphorus excretion in the proximal tubule of the kidney, and inhibition of this hormone leads to phosphate reabsorption (129). FGFR inhibitors can also cause nail changes with onycholysis, mucosal dryness, ocular toxicity, nausea, anorexia, diarrhea, and constipation, and adequate management of the toxicities will be key to further development of this class of drugs.

Overall, the preliminary data for FGFR inhibitors in advanced ICC are encouraging.

Angiogenesis

Not only are the ligands regulating angiogenesis (particularly VEGFA) commonly present (40%–75%) in BTCs (130–132), but their expression is colocated with their receptors VEGFR1 and VEGFR2 in endothelial cells adjacent to the tumors (133). This appears to be most evident at the invasive edge of the tumor (134). VEGF expression is associated with a number of adverse prognostic features, including the presence of metastases in ICC (132) and increased microvascular density (MVD) in both cholangiocarcinoma (131) and gallbladder cancer (130). MVD is an independent prognostic factor for disease-free survival following resection of ECC (134) and for OS in lymph node–negative ICC (135) and gallbladder cancer (134). MVD is also an independent negative predictor of OS in ECC (136). Consequently, a number of clinical trials have evaluated VEGF inhibition.

In a phase II trial of bevacizumab combined with gemcitabine and oxaliplatin, our group demonstrated a significant decrease in standardized uptake values on FDG-PET scans after two treatment cycles, particularly in patients with a partial response or stable disease (137). However, the 6-month progression-free survival (PFS; 63%) was below the target rate of 70%. Combining bevacizumab with erlotinib (an anti-EGFR TKI) achieved partial responses in 12% of patients and stable disease in 51%, with a median OS of 9.9 months, notably in the absence of concurrent chemotherapy (138).

Cediranib is an oral VEGFR1, VEGFR2, and VEGFR3 TKI, with activity against PDGF receptors and c-KIT. In the randomized phase II, placebo-controlled ABC-03 study, we observed an improved response rate in patients receiving cisplatin/gemcitabine with cediranib (44%) versus placebo (19%; P = 0.0036) and improved 6-month PFS (70.5% vs. 61.3%; P > 0.05). However, the study did not meet its primary endpoint (improvement in median PFS), in part due to the relatively poor tolerability of cediranib (139).

Forays into VEGF inhibition with other TKIs have been disappointing. Single-arm, phase II studies of sorafenib as monotherapy (140, 141) or combined with erlotinib (142) or cisplatin/gemcitabine (143) have all failed to demonstrate sufficient activity in BTC. Most recently, sorafenib failed to improve PFS when added to gemcitabine in a randomized phase II, placebo-controlled study (144). A phase II clinical trial of sunitinib including 56 patients with BTC reported a median time to progression of only 1.7 months, an objective response rate of 8.9% and a disease control rate of 50% (145).

In addition, the VanGogh study failed to show an improvement in PFS in a 3-arm randomized phase II study exploring the role of vandetanib in 173 patients (146). Results of ongoing studies with pazopanib (NCT01855724), regorafenib (NCT02053376 and NCT02115542), and ramucirumab (NCT02711553) are awaited.

HER Family

EGFR amplifications and mutations have been described in around 6% and 13.6% to 15% of BTCs, respectively. However, the biological implication of these mutations is unclear (147–149).

Several phase II clinical trials have combined cetuximab, a monoclonal antibody targeting EGFR, with chemotherapy in the treatment of BTCs, most of them with gemcitabine and oxaliplatin (150–153). Initial promising results reporting high tumor response rates (63%) from a small study (n = 30; ref. 151) were not confirmed in the randomized phase II BINGO study, in keeping with results from other negative phase II studies combining cetuximab with chemotherapy (153).

KRAS wild-type patients with advanced BTC were treated with panitumumab combined with gemcitabine, capecitabine, and oxaliplatin (46 patients; ref. 154) and with gemcitabine and oxaliplatin (31 patients; ref. 155) in two separate phase II clinical trials. In each study, the primary endpoint was achieved [6-month PFS of 74% (95% CI, 58–84; ref. 154) and response rate of 45% (155), respectively]. A third phase II study (panitumumab with gemcitabine and irinotecan in nonselected patients) also supported further development of this compound in BTC with no difference in OS by KRAS status (7 of 31 patients harbored a KRAS mutation; ref. 156). Unfortunately, these signals have not been confirmed in other studies (157, 158), including the largest randomized phase II study combining panitumumab with gemcitabine and oxaliplatin (the Vecti-BIL study; ref. 158) which showed no differences in survival in 85 randomized patients.

Erlotinib, a TKI targeting EGFR, has also shown varying results (159–161). In the largest (phase III) study, 133 patients were randomized to receive gemcitabine and oxaliplatin chemotherapy with or without erlotinib (160); there were no differences in PFS or OS when all patients with BTC were analyzed together. However, the cholangiocarcinoma patient subgroup did appear to derive benefit from adding erlotinib to chemotherapy [median PFS 5.9 months (95% CI, 4.7–7.1) vs. 3.0 months (95% CI, 1.1–4.9); P = 0.049], and further clinical trials are ongoing (NCT00832637 and NCT00987766).

HER2 (v-ERBB2, erythroblastic leukaemia viral oncogene homolog-2) overexpression and gene amplification are also described in BTCs, with a higher incidence in gallbladder cancer (19%; ref. 162). The rate of HER2 overexpression was found to be higher in ECC (17.4%) than in ICC (4.8%) in a recent systematic review and meta-analysis published by Galdy and colleagues in 2016 (162). Good correlation between overexpression and gene amplification (75%) has been shown (148). Two phase II trials have yielded disappointing results of first-line/second-line lapatinib monotherapy in an unselected population of patients with advanced BTC (163, 164). Case reports using trastuzumab in patients with gallbladder carcinoma with HER2 overexpression have suggested activity (165, 166), and a phase II clinical trial is under way (NCT00478140). Afatinib has shown activity in one patient diagnosed with cholangiocarcinoma in a phase I clinical trial (167); a phase I study of afatinib combined with cisplatin and gemcitabine in patients with BTC is ongoing (NCT01679405).

Targeting WNT/β-catenin, Hedgehog, and HGF/c-MET

The WNT/β-catenin pathway is involved in the regulation of cell invasion and migration. High nuclear expression with low membranous expression of β-catenin expression has been described in ICC (15%; ref. 168); WNT signaling seems to be most relevant in hilar cholangiocarcinoma (169). WNT pathway activation was associated with chemoresistance and metastatic spread in a cholangiocarcinoma xenograft tumor model (170), and WNT inhibition reversed chemoresistance in cell lines (171). In contrast, its impact and the mutational status of this pathway's components are not completely understood in BTC (172, 173). Whole-exome sequencing of Opisthorchis viverrini–related cholangiocarcinoma identified mutations in the WNT pathway [RNF43 (9.3%; refs. 45, 174)]. β-catenin expression is associated with decreased apoptosis in gallbladder cancers (175), and Yadav and colleagues showed that most of the genetic variants of WNT signaling pathways that were evaluated influenced gallbladder cancer susceptibility (176). Although multiple WNT pathway inhibitors are currently under development (177), only a few clinical trials have been reported for BTC as of yet. Eads and colleagues explored the safety of DKK1, an inhibitor of the canonical WNT/β-catenin pathway, in combination with gemcitabine and cisplatin in a phase I clinical trial enrolling patients with BTCs (178). The combination was found to be safe, suggesting possible prolonged disease stabilization; further development is awaited.

The Hedgehog pathway may also be involved in the development of BTC (179–182). Among gallbladder cancer specimens, expression of Hedgehog pathway components by IHC has been described [SHH (81.7%), PTCH1 (75.3%), and GLI1 (70.0%)] with impact on stage and lymph node, venous, and hepatic infiltration; patients with activated Hedgehog pathway had more aggressive tumors and worse outcome (183). Similar findings have been described in cholangiocarcinoma [SHH (87.8%), PTCH1 (89.2%), GLI1 (85.4%); ref. 184]. Suppression of the Hedgehog pathway in gallbladder (185) and cholangiocarcinoma (186) cell lines implanted in mouse xenografts inhibited epithelial–mesenchymal transition and reduced tumor volume, suggesting this pathway as a potential new target.

c-MET tyrosine kinase plays an integral role in carcinogenesis by promoting angiogenesis, tumor invasion, and metastasis. Binding of this receptor by the ligand hepatocyte growth factor (HGF) activates multiple downstream signal transduction pathways, including the GRB2–RAS–MAPK cascade and the PI3K, EGFR, VEGF, and RAC1–CDC42 pathways (187). c-MET overexpression, associated with a poor prognosis in cholangiocarcinoma (188), is seen in 12% to 58% of ICCs (151, 188, 189) and 0% to 16% of ECCs (151), a wide variation likely accounted for by differences in the c-MET antibody used, definition of positivity, analysis of resection versus late-stage biopsy samples, and sample size per study. c-MET amplification is rare in cholangiocarcinoma, but has been reported at a frequency of 7% in one study of 26 cases of ICC analyzed by NGS (52). In addition to the above-mentioned effect in cholangiocarcinoma, the HGF–c-MET pathway promotes proteolytic activity and induces cellular motility, essential for the invasive progression of gallbladder carcinoma cell lines (190). In human tissue, c-MET expression is higher in cancer cells than in normal gallbladder tissue (191), up to 74% in some series (192).

Significant cross-talk has been demonstrated between the c-MET pathway and other pathways such as the EGFR and VEGF pathways in other tumor types. c-MET amplification has been shown to drive resistance to EGFR inhibitors via ERBB3-dependent activation of PI3K (193). Similarly, tumor hypoxia, which can be a consequence of VEGF pathway inhibition, has been shown to upregulate c-MET and enhance scatter and invasiveness (194). Thus, dual inhibition of c-MET with other pathways may be a strategy in cholangiocarcinoma. Cabozantinib, which has potent activity against both c-MET (IC50 = 1.3 nmol/L) and VEGFR2 (IC50 = 0.035 nmol/L), was tested in a phase II trial in patients with advanced cholangiocarcinoma; preliminary data showed minimal activity, with early trial discontinuation after 12 of 19 patients failed to be progression-free at 16 weeks (195). A randomized phase II study with merestinib in addition to cisplatin/gemcitabine as first-line therapy is ongoing (NCT02711553).

KRAS–BRAF–MEK–ERK Pathway

As in many other cancers, the RAS–RAF–MEK–ERK signal transduction pathway is frequently dysregulated in cholangiocarcinoma (196). The binding of ligands, including EGF and PDGF, to the receptors triggers a cascade of activation of downstream signaling molecules. Activated RAS triggers phosphorylation and activation of RAF kinase, leading to end phosphorylation of MEK1 and MEK2. Activated MEK phosphorylates ERK1 and ERK2, the only known substrates. Phosphorylated ERK (pERK) then dimerizes and translocates to the nucleus (197), where it regulates several important cellular functions. Gain-of-function KRAS mutation with a frequency of 9% to 40% has been reported in cholangiocarcinoma (52, 83). KRAS mutation has been associated with perineural invasion, advanced stage, and poor prognosis (198). KRAS mutations have also been found in up to 7.8% of gallbladder cancers (101, 199). BRAF mutations seem not to be significant in gallbladder cancer and appear to be restricted to ICC only (199, 200). However, other groups' results show mutation rates between 5.9% (101) and 33% (201).

Despite the recognized frequency of KRAS mutations, targeting this pathway remains challenging. BRAF is a proto-oncogene and is a key component of the RAS–RAF–MEK–ERK proliferation signaling pathway. The most common BRAF gene mutation found in human cancers is V600E, with varying frequency reported in cholangiocarcinoma (202). In a recent phase II basket study of vemurafenib in BRAFV600-mutated non-melanoma cancers, one patient with cholangiocarcinoma achieved a durable partial response of over a year (203).

MEK is also an attractive target, as ERK1 and ERK2 are the only known MEK substrates (204). Early evidence of the efficacy of a MEK inhibitor was reported in a single-arm study of selumetinib in advanced BTCs (205). Of 29 patients enrolled, 25 were evaluable for response: 3 patients (12%) had confirmed partial responses and 17 (68%) had stable disease. The median PFS was 3.7 months (95% CI, 3.5–4.9), and median OS was 9.8 months (95% CI, 5.97–not available). In this study, no BRAFV600E mutations were found, but the absence of pERK staining appeared to be associated with a lack of response to selumetinib. Recently, in the ABC-04 phase Ib study, we assessed the safety and tolerability of selumetinib in combination with cisplatin/gemcitabine in advanced BTC; 3 of 8 patients evaluable for response had partial responses. Selumetinib-related toxicities were manageable and included grade 1 to 2 edema and rash (206).

Given well-known redundancy and cross-talk in this pathway, novel combined strategies targeting different molecules within this pathway or different pathways represent attractive approaches in cholangiocarcinoma.

The PI3K–AKT–mTOR Pathway

This pathway is upregulated in cholangiocarcinoma cells; moreover, activation of this pathway is associated with adverse prognosis in some patients with BTC (207) and good prognosis in others. Some studies have shown that somatic PIK3CA mutations contribute to the frequent activation of the PI3K–AKT pathway in BTC (208).

In a study of 212 ECC cases, patients with high pAKT expression had shorter survival than those with low pAKT expression (P = 0.06). Cases with high p-mTOR expression also showed shorter survival (P = 0.06). Patients with low PTEN expression (median survival, 18 months) had a significantly worse survival time than those with high PTEN expression (median survival, 39 months; log-rank test P = 0.004; ref. 209). Conversely, a study on 101 ICCs showed the aberrant expression of pAKT1 and p-mTOR was associated with a favorable prognosis regardless of PTEN (210). PTEN overexpression was found as an independent favorable prognostic factor for patients with ICC. In addition, the overexpression of p-mTOR was more frequently observed in well-differentiated to moderately differentiated tumors and in tumors without metastasis. The comparison of these two studies underlined the difficulties in comparing different BTCs due to biological differences depending on their primary location and, ultimately, cell of origin. In addition, the redundancy and cross-talk involving this intracellular pathway makes targeting a single point/level unlikely to be a successful approach.

MK-2206 is an oral selective allosteric inhibitor of AKT that targets all three isoforms of human AKT (AKT1, AKT2, and AKT3) with IC50 values of 8, 12, and 65 nmol/L, respectively. An abbreviated phase II study using this compound in 8 patients with at least one prior systemic treatment was disappointing, with a median PFS of 1.7 months and median OS of 3.5 months (211).

A first-line phase II study with everolimus showed evidence of antitumor activity, with 14 out of 27 patients (56%, 95% CI, 35–76) achieving tumor control at 12 weeks and 2 of them achieving partial response. The median PFS was 6.0 months (95% CI, 2.1–11.2), and median OS was 9.5 months (95% CI, 5.5–16.6). Correlative studies suggest that KRAS mutational status and basal pAKT levels might be associated with resistance to everolimus treatment (212).

A phase II trial using a PI3K inhibitor, copanlisib (BAY 80-6946), as first line in combination with gemcitabine and cisplatin is ongoing (NCT02631590).

Currently, the most promising targets under development due to a more solid preclinical research background are IDH inhibitors for IDH-mutant BTC and molecules targeting FGFR2 gene fusions (Fig. 4 and Fig. 5). A window of opportunity is open, with new drugs in development targeting chromatin remodeling gene mutations (ARID1, BAP1, and PBRM1) such as bromodomain and extra-terminal (BET) inhibitors (213). Most of the remaining molecular targets that have been tested in clinical trials have been somewhat disappointing, with conflicting data and negative trials, underlining the need for new models and new approaches to unravel the complex molecular biology of BTC (Table 1).

Is Precision Medicine Regarding Targeted Therapies in BTC Ready for the Clinic?

As in other malignancies, the meaningful decrease in the cost of NGS technologies has opened the door for more sophisticated trials in which different molecular subtypes of a malignancy can be matched to targeted inhibitors. Obtaining tumor molecular profiling on patients who are fit to enroll in clinical trials beyond first-line systemic therapy may offer these patients additional promising treatment options. However, obtaining sufficient tissue for such analyses in BTC can be difficult, making this approach more challenging. For this scenario, the use of liquid biopsies [circulating tumor cells (CTC), cfDNA, exosomes, etc.], when validated, may lead the way to such approaches in these neoplasms.

Role of Immunotherapy

The relationship between chronic inflammation and the development of BTC has led investigators to harness the immune response through vaccination, adoptive immunotherapy, and checkpoint inhibition.

Immune cells (both innate and adoptive) are present in BTCs; this appears to be stage dependent (for macrophages), and the presence of dendritic cells, CD4+ helper T-lymphocytes, CD8+ cytotoxic T-lymphocytes, and B-lymphocytes/plasma cells is associated with improved survival (214).

Vaccination studies have yielded modest results in monotherapy; the commonest targets are Wilm's tumor-1 (WT1) and mucin protein 1 (MUC1). WT1, a transcription factor, is also a tumor suppressor through interaction with PDGFR, EGFR, c-MYC, and BCL2. A phase I study in combination with gemcitabine showed that patients demonstrating a T-cell response to WT1 vaccination had a longer OS than patients treated with gemcitabine only (215). MUC1, a glycoprotein forming a hydrophilic barrier to hydrophobic cytotoxic agents and immune surveillance, is highly overexpressed in gallbladder cancers (90%), and less so in cholangiocarcinoma (59%–77%), and is associated with advanced stage and impaired survival. An early study showed that MUC1 vaccination did not translate into clinical benefit despite achieving an IgG response (216). A dendritic cell–based vaccine targeting MUC1 in patients with resected pancreatic cancer and BTC (with adjuvant chemotherapy or radiotherapy as appropriate) saw 4 of 12 patients disease free at four years (217). Expanding vaccination to target two (218), three (219), or four (220) peptides, or even “personalizing” the vaccination (221), holds promise but remains investigational. Defining the optimal target among heterogeneous entities within BTC, vaccination against single versus multiple targets, and definition of optimal adjuvants are required.

Shimizu and colleagues vaccinated patients with resected ICC with autologous tumor lysate–pulsed dendritic cells plus ex vivo–activated T-cell transfer (adoptive immunotherapy). These patients had a near-double OS (31.9 vs. 17.4 months, P = 0.022) compared with surgery-alone patients, most marked in patients with prominent skin reactions (222).

Mutational load is known to be high in tumors in which immunotherapies have been shown to be effective, such as melanoma and lung cancer (223). Based on a similar rationale, efficacy of checkpoint inhibitors in tumors with mismatch-repair (MMR) deficiency was proven to be successful in a phase II study, achieving up to 40% objective responses (224). Mutational load has shown to be high in BTCs (88). In addition, MMR and microsatellite instability (MSI) have been explored in BTCs. MMR and MSI have been suggested to be infrequent in BTCs without hereditary nonpolyposis colorectal cancer (225). Results vary between series; high-level MSI has been shown in 5% of gallbladder carcinoma (226), 5%–13% of ECC (226, 227), and up to 10% of ICC (226). MMR status (hMLH1 and hMSH2 negativity) was shown in 51.3% and 59% of gallbladder carcinoma and 57.1% and 65.7% of ECC, respectively (228). In addition, O(6)-Methylguanine-DNA methyltransferase (MGMT) methylation was identified in 59% of gallbladder carcinoma and 60% of ECC (228). Both MGMT methylation and MMR status correlated with poor prognosis in gallbladder carcinoma and ECC (228).

A case report of tumor-infiltrating lymphocytes from a patient with metastatic cholangiocarcinoma containing CD4+ T-helper-1 cells recognizing a mutation in ERBB2-interacting protein induced an impressive and durable response; moreover, this effect was reproduced after subsequent disease progression (229). Adoptive immunotherapy studies in Thailand (NCT01868490) and the United States (NCT01174121) are ongoing.

Holcombe and colleagues explored a cohort of BTC samples (126 ECC, 434 ICC, 244 gallbladder cancer, and 11 not specified) and identified high PD-1 (40%) and PD-L1 in (15%) expression (50). In the BTC cohort of KEYNOTE-28 (NCT02054806), 37 of 89 patients (42%) were PD-L1–positive (defined as ≥1% staining of cells in tumor nests or PD-L1–positive bands in stroma by IHC). Four of 24 patients (17%) treated with pembrolizumab, a highly selective humanized monoclonal antibody targeting PD-1, had a partial response, with another 4 achieving stable disease; 5 patients entered long-term treatment, including all 4 responders (230). These encouraging results suggest that this strategy is worth pursuing (a phase I study in combination with FOLFOX chemotherapy, with an expanded phase II cohort in BTC, is under way; NCT02268825), along with validation of PD-L1 expression as a predictive biomarker, evaluation of the role of PD-L2 expression, and assessment of efficacy in the various BTC subgroups as well as in patients with MMR-deficient tumors (224).

The treatment paradigm for patients with advanced BTC is evolving; through international collaboration, BTCs are no longer considered “too rare” for adequately powered clinical studies. Emerging evidence suggests that BTC encompasses subgroups with discrete driver mutations, some of which are targetable with novel therapies. The role of conventional therapies (chemotherapy and radiotherapy) has yet to be fully defined, particularly in the adjuvant and second-line settings. In addition, investigation of a number of pathway-targeted therapies, as well as modulation of the immune environment, holds promise for patients with these diseases. Given the low prevalence of BTC, clinical development must go hand-in-hand with sound basic and translational research.

J.W. Valle has received honoraria from the speakers bureau of Celgene and is a consultant/advisory board member for AstraZeneca, Lilly, Merck, and Agios. A.X. Zhu is a consultant/advisory board member for Bayer, BMS, Eisai, Merck, Novartis, and Sanofi. No potential conflicts of interest were disclosed by the other authors.

The authors would like to thank Stephanie L. Reyes for her assistance with Figs. 2 and 4.

J.W. Valle received research funding from Cancer Research UK (CRUK) and The Alan Morement Memorial Fund (AMMF). A. Lamarca received funding from the Spanish Society of Medical Oncology (SEOM) Fellowship, the Cholangiocarcinoma Foundation Research Fellowship, and the Conquer Cancer Foundation Young Investigator Award. J. Barriuso is partly funded by the Spanish Society of Medical Oncology (SEOM) Fellowship. L. Goyal is supported by the MGH Executive Committee on Research Fund for Medical Discovery Award, the Cholangiocarcinoma Foundation Research Fellowship, the NIH Loan Repayment Program Grant, the Jonathan Kraft Translational Award, and NIH/NCI Gastrointestinal Cancer SPOREP50 CA127003. A. Zhu is supported by the TargetCancer Foundation, a Translational Research Grant Award from the V Foundation for Cancer Research, and the Jonathan Kraft Translational Award.

1.
Valle
JW
. 
Advances in the treatment of metastatic or unresectable biliary tract cancer
.
Ann Oncol
2010
;
21
;
Suppl 7
:
vii345
8
.
2.
Edge
S
,
Byrd
DR
,
Compton
CC
,
Fritz
AG
,
Greene
FL
,
Trotti
A
.
AJCC Cancer Staging Manual (Seventh Edition)
.
New York
:
Springer-Verlag
. 
2010
.
Full book
.
3.
Patel
T
. 
Increasing incidence and mortality of primary intrahepatic cholangiocarcinoma in the United States
.
Hepatology
2001
;
33
:
1353
7
.
4.
Bertuccio
P
,
Bosetti
C
,
Levi
F
,
Decarli
A
,
Negri
E
,
La Vecchia
C
. 
A comparison of trends in mortality from primary liver cancer and intrahepatic cholangiocarcinoma in Europe
.
Ann Oncol
2013
;
24
:
1667
74
.
5.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2016
.
CA Cancer J Clin
2016
;
66
:
7
30
.
6.
Wang
Y
,
Li
J
,
Xia
Y
,
Gong
R
,
Wang
K
,
Yan
Z
, et al
Prognostic nomogram for intrahepatic cholangiocarcinoma after partial hepatectomy
.
J Clin Oncol
2013
;
31
:
1188
95
.
7.
Wang
SJ
,
Lemieux
A
,
Kalpathy-Cramer
J
,
Ord
CB
,
Walker
GV
,
Fuller
CD
, et al
Nomogram for predicting the benefit of adjuvant chemoradiotherapy for resected gallbladder cancer
.
J Clin Oncol
2011
;
29
:
4627
32
.
8.
Anderson
C
,
Kim
R
. 
Adjuvant therapy for resected extrahepatic cholangiocarcinoma: a review of the literature and future directions
.
Cancer Treat Rev
2009
;
35
:
322
7
.
9.
DeOliveira
ML
,
Cunningham
SC
,
Cameron
JL
,
Kamangar
F
,
Winter
JM
,
Lillemoe
KD
, et al
Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution
.
Ann Surg
2007
;
245
:
755
62
.
10.
Bridgewater
J
,
Lopes
A
,
Wasan
H
,
Malka
D
,
Jensen
L
,
Okusaka
T
, et al
Prognostic factors for progression-free and overall survival in advanced biliary tract cancer
.
Ann Oncol
2016
;
27
:
134
40
.
11.
Tyson
GL
,
El-Serag
HB
. 
Risk factors for cholangiocarcinoma
.
Hepatology
2011
;
54
:
173
84
.
12.
Zatonski
WA
,
Lowenfels
AB
,
Boyle
P
,
Maisonneuve
P
,
Bueno de Mesquita
HB
,
Ghadirian
P
, et al
Epidemiologic aspects of gallbladder cancer: a case-control study of the SEARCH Program of the International Agency for Research on Cancer
.
J Natl Cancer Inst
1997
;
89
:
1132
8
.
13.
Chapman
RW
. 
Risk factors for biliary tract carcinogenesis
.
Ann Oncol
1999
;
10
Suppl 4
:
308
11
.
14.
Chaiyadet
S
,
Sotillo
J
,
Smout
M
,
Cantacessi
C
,
Jones
MK
,
Johnson
MS
, et al
Carcinogenic liver fluke secretes extracellular vesicles that promote cholangiocytes to adopt a tumorigenic phenotype
.
J Infect Dis
2015
;
212
:
1636
45
.
15.
Gourgiotis
S
,
Kocher
HM
,
Solaini
L
,
Yarollahi
A
,
Tsiambas
E
,
Salemis
NS
. 
Gallbladder cancer
.
Am J Surg
2008
;
196
:
252
64
.
16.
Gatto
M
,
Alvaro
D
. 
Cholangiocarcinoma: risk factors and clinical presentation
.
Eur Rev Med Pharmacol Sci
2010
;
14
:
363
7
.
17.
Heinrich
S
,
Clavien
PA
. 
Ampullary cancer
.
Curr Opin Gastroenterol
2010
;
26
:
280
5
.
18.
Valle
JW
,
Borbath
I
,
Khan
SA
,
Huguet
F
,
Gruenberger
T
,
Arnold
D
. 
Biliary cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
2016
;
27
:
v28
37
.
19.
Endo
I
,
Gonen
M
,
Yopp
AC
,
Dalal
KM
,
Zhou
Q
,
Klimstra
D
, et al
Intrahepatic cholangiocarcinoma: rising frequency, improved survival, and determinants of outcome after resection
.
Ann Surg
2008
;
248
:
84
96
.
20.
Zhu
AX
,
Knox
JJ
. 
Adjuvant therapy for intrahepatic cholangiocarcinoma: the debate continues
.
Oncologist
2012
;
17
:
1504
7
.
21.
Horgan
AM
,
Amir
E
,
Walter
T
,
Knox
JJ
. 
Adjuvant therapy in the treatment of biliary tract cancer: a systematic review and meta-analysis
.
J Clin Oncol
2012
;
30
:
1934
40
.
22.
Julien Edeline FBJMPea
. 
Gemox versus surveillance following surgery of localized biliary tract cancer: Results of the PRODIGE 12-ACCORD 18 (UNICANCER GI) phase III trial
.
ASCO 2017 Gastrointestinal Cancer Symposium
,
J Clin Oncol
2017
;
35
(suppl 4S; abstract 225).
23.
Primrose
JN
,
Fox
R
,
Palmer
D
,
Prasad
R
,
Mirza
D
,
Anthoney
DA
et al 
Adjuvant capecitabine for biliary tract cancer: The BILCAP randomized study
.
J Clin Oncol
2017
;
35
(
suppl; abstr 4006)
.
24.
Tao
R
,
Krishnan
S
,
Bhosale
PR
,
Javle
MM
,
Aloia
TA
,
Shroff
RT
, et al
Ablative radiotherapy doses lead to a substantial prolongation of survival in patients with inoperable intrahepatic cholangiocarcinoma: a retrospective dose response analysis
.
J Clin Oncol
2016
;
34
:
219
26
.
25.
Hong
TS
,
Wo
JY
,
Yeap
BY
,
Ben-Josef
E
,
McDonnell
EI
,
Blaszkowsky
LS
, et al
Multi-institutional phase II study of high-dose hypofractionated proton beam therapy in patients with localized, unresectable hepatocellular carcinoma and intrahepatic cholangiocarcinoma
.
J Clin Oncol
2016
;
34
:
460
8
.
26.
Al-Adra
DP
,
Gill
RS
,
Axford
SJ
,
Shi
X
,
Kneteman
N
,
Liau
SS
. 
Treatment of unresectable intrahepatic cholangiocarcinoma with yttrium-90 radioembolization: a systematic review and pooled analysis
.
Eur J Surg Oncol
2015
;
41
:
120
7
.
27.
Edeline
J
,
Du
FL
,
Rayar
M
,
Rolland
Y
,
Beuzit
L
,
Boudjema
K
, et al
Glass microspheres 90Y selective internal radiation therapy and chemotherapy as first-line treatment of intrahepatic cholangiocarcinoma
.
Clin Nucl Med
2015
;
40
:
851
5
.
28.
Hibi
T
,
Itano
O
,
Shinoda
M
,
Kitagawa
Y
. 
Liver transplantation for hepatobiliary malignancies: a new era of “Transplant Oncology” has begun
.
Surg Today
2017
;
47
:
403
15
.
29.
Rosen
CB
,
Heimbach
JK
,
Gores
GJ
. 
Surgery for cholangiocarcinoma: the role of liver transplantation
.
HPB (Oxford)
2008
;
10
:
186
9
.
30.
Eckel
F
,
Schmid
RM
. 
Chemotherapy in advanced biliary tract carcinoma: a pooled analysis of clinical trials
.
Br J Cancer
2007
;
96
:
896
902
.
31.
Tsavaris
N
,
Kosmas
C
,
Gouveris
P
,
Gennatas
K
,
Polyzos
A
,
Mouratidou
D
, et al
Weekly gemcitabine for the treatment of biliary tract and gallbladder cancer
.
Invest New Drugs
2004
;
22
:
193
8
.
32.
Raderer
M
,
Hejna
MH
,
Valencak
JB
,
Kornek
GV
,
Weinlander
GS
,
Bareck
E
, et al
Two consecutive phase II studies of 5-fluorouracil/leucovorin/mitomycin C and of gemcitabine in patients with advanced biliary cancer
.
Oncology
1999
;
56
:
177
80
.
33.
Valle
J
,
Wasan
H
,
Palmer
DH
,
Cunningham
D
,
Anthoney
A
,
Maraveyas
A
, et al
Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer
.
N Engl J Med
2010
;
362
:
1273
81
.
34.
Okusaka
T
,
Nakachi
K
,
Fukutomi
A
,
Mizuno
N
,
Ohkawa
S
,
Funakoshi
A
, et al
Gemcitabine alone or in combination with cisplatin in patients with biliary tract cancer: a comparative multicentre study in Japan
.
Br J Cancer
2010
;
103
:
469
74
.
35.
Lamarca
A
,
Benafif
S
,
Ross
P
,
Bridgewater
J
,
Valle
JW
. 
Cisplatin and gemcitabine in patients with advanced biliary tract cancer (ABC) and persistent jaundice despite optimal stenting: Effective intervention in patients with luminal disease
.
Eur J Cancer
2015
;
51
:
1694
703
.
36.
Lamarca
A
,
Hubner
RA
,
Ryder
WD
,
Valle
JW
. 
Second-line chemotherapy in advanced biliary cancer: a systematic review
.
Ann Oncol
2014
;
25
:
2328
38
.
37.
Walter
T
,
Horgan
AM
,
McNamara
M
,
McKeever
L
,
Min
T
,
Hedley
D
, et al
Feasibility and benefits of second-line chemotherapy in advanced biliary tract cancer: a large retrospective study
.
Eur J Cancer
2013
;
49
:
329
35
.
38.
Bridgewater
J
,
Palmer
D
,
Cunningham
D
,
Iveson
T
,
Gillmore
R
,
Waters
J
, et al
Outcome of second-line chemotherapy for biliary tract cancer
.
Eur J Cancer
2012
;
49
:
1511
.
39.
Oh
SY
,
Jeong
CY
,
Hong
SC
,
Kim
TH
,
Ha
CY
,
Kim
HJ
, et al
Phase II study of second line gemcitabine single chemotherapy for biliary tract cancer patients with 5-fluorouracil refractoriness
.
Invest New Drugs
2011
;
29
:
1066
72
.
40.
Lee
S
,
Oh
SY
,
Kim
BG
,
Kwon
HC
,
Kim
SH
,
Rho
MH
, et al
Second-line treatment with a combination of continuous 5-fluorouracil, doxorubicin, and mitomycin-C (conti-FAM) in gemcitabine-pretreated pancreatic and biliary tract cancer
.
Am J Clin Oncol
2009
;
32
:
348
52
.
41.
Sasaki
T
,
Isayama
H
,
Nakai
Y
,
Mizuno
S
,
Yamamoto
K
,
Yagioka
H
, et al
Multicenter phase II study of S-1 monotherapy as second-line chemotherapy for advanced biliary tract cancer refractory to gemcitabine
.
Invest New Drugs
2012
;
30
:
708
13
.
42.
Rao
S
,
Cunningham
D
,
Hawkins
RE
,
Hill
ME
,
Smith
D
,
Daniel
F
, et al
Phase III study of 5FU, etoposide and leucovorin (FELV) compared to epirubicin, cisplatin and 5FU (ECF) in previously untreated patients with advanced biliary cancer
.
Br J Cancer
2005
;
92
:
1650
4
.
43.
Lim
JY
,
Jeung
HC
,
Mun
HS
,
Lee
DK
,
Paik
YH
,
Lee
SJ
, et al
Phase II trial of oxaliplatin combined with leucovorin and fluorouracil for recurrent/metastatic biliary tract carcinoma
.
Anticancer Drugs
2008
;
19
:
631
5
.
44.
Lamarca
A
,
Palmer
D
,
Wasan
H
,
Ryder
WD
,
Davies
L
,
Flight
H
et al 
ABC-06: A randomised phase III, multi-centre, open-label study of active symptom control (ASC) alone or ASC with oxaliplatin/5-FU chemotherapy for patients with locally advanced/metastatic biliary tract cancer (ABC) previously treated with cisplatin/gemcitabine chemotherapy
.
Ann Oncol
2014
;
25
(
suppl_4
):
iv252
.
45.
Ong
CK
,
Subimerb
C
,
Pairojkul
C
,
Wongkham
S
,
Cutcutache
I
,
Yu
W
, et al
Exome sequencing of liver fluke-associated cholangiocarcinoma
.
Nat Genet
2012
;
44
:
690
3
.
46.
Chan-On
W
,
Nairismagi
ML
,
Ong
CK
,
Lim
WK
,
Dima
S
,
Pairojkul
C
, et al
Exome sequencing identifies distinct mutational patterns in liver fluke-related and non-infection-related bile duct cancers
.
Nat Genet
2013
;
45
:
1474
8
.
47.
Jiao
Y
,
Pawlik
TM
,
Anders
RA
,
Selaru
FM
,
Streppel
MM
,
Lucas
DJ
, et al
Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas
.
Nat Genet
2013
;
45
:
1470
3
.
48.
Borad
MJ
,
Champion
MD
,
Egan
JB
,
Liang
WS
,
Fonseca
R
,
Bryce
AH
, et al
Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma
.
PLoS Genet
2014
;
10
:
e1004135
.
49.
Gao
Q
,
Zhao
YJ
,
Wang
XY
,
Guo
WJ
,
Gao
S
,
Wei
L
, et al
Activating mutations in PTPN3 promote cholangiocarcinoma cell proliferation and migration and are associated with tumor recurrence in patients
.
Gastroenterology
2014
;
146
:
1397
407
.
50.
Holcombe
RF
,
Xiu
J
,
Pishvaian
MJ
,
Millis
SZ
,
Gatalica
Z
,
Reddy
SK
et al 
Tumor profiling of biliary tract carcinomas to reveal distinct molecular alterations and potential therapeutic targets
.
ASCO Annual Meeting 2015
,
J Clin Oncol
33
, 
2015
(
suppl 3; abstr 285
).
51.
Ross
JS
,
Wang
K
,
Catenacci
DVT
,
Chmielecki
J
,
Ali
SM
,
Elvin
JA
et al 
Comprehensive genomic profiling of biliary tract cancers to reveal tumor-specific differences and genomic alterations
.
ASCO Annual Meeting 2015
,
J Clin Oncol
33
, 
2015
(
suppl 3; abstr 231
).
52.
Ross
JS
,
Wang
K
,
Gay
L
,
Al-Rohil
R
,
Rand
JV
,
Jones
DM
, et al
New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing
.
Oncologist
2014
;
19
:
235
42
.
53.
Javle
M
,
Bekaii-Saab
T
,
Jain
A
,
Wang
Y
,
Kelley
RK
,
Wang
K
, et al
Biliary cancer: Utility of next-generation sequencing for clinical management
.
Cancer
2016
;
122
:
3838
47
.
54.
Sia
D
,
Hoshida
Y
,
Villanueva
A
,
Roayaie
S
,
Ferrer
J
,
Tabak
B
, et al
Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes
.
Gastroenterology
2013
;
144
:
829
40
.
55.
Furubo
S
,
Harada
K
,
Shimonishi
T
,
Katayanagi
K
,
Tsui
W
,
Nakanuma
Y
. 
Protein expression and genetic alterations of p53 and ras in intrahepatic cholangiocarcinoma
.
Histopathology
1999
;
35
:
230
40
.
56.
Momoi
H
,
Itoh
T
,
Nozaki
Y
,
Arima
Y
,
Okabe
H
,
Satoh
S
, et al
Microsatellite instability and alternative genetic pathway in intrahepatic cholangiocarcinoma
.
J Hepatol
2001
;
35
:
235
44
.
57.
Tannapfel
A
,
Weinans
L
,
Geissler
F
,
Schutz
A
,
Katalinic
A
,
Kockerling
F
, et al
Mutations of p53 tumor suppressor gene, apoptosis, and proliferation in intrahepatic cholangiocellular carcinoma of the liver
.
Dig Dis Sci
2000
;
45
:
317
24
.
58.
Tannapfel
A
,
Sommerer
F
,
Benicke
M
,
Weinans
L
,
Katalinic
A
,
Geissler
F
, et al
Genetic and epigenetic alterations of the INK4a-ARF pathway in cholangiocarcinoma
.
J Pathol
2002
;
197
:
624
31
.
59.
Sia
D
,
Tovar
V
,
Moeini
A
,
Llovet
JM
. 
Intrahepatic cholangiocarcinoma: pathogenesis and rationale for molecular therapies
.
Oncogene
2013
;
32
:
4861
70
.
60.
Lee
KT
,
Chang
WT
,
Wang
SN
,
Chuang
SC
,
Chai
CY
,
Hu
SW
. 
Expression of DPC4/Smad4 gene in stone-containing intrahepatic bile duct
.
J Surg Oncol
2006
;
94
:
338
43
.
61.
Yan
XQ
,
Zhang
W
,
Zhang
BX
,
Liang
HF
,
Zhang
WG
,
Chen
XP
. 
Inactivation of Smad4 is a prognostic factor in intrahepatic cholangiocarcinoma
.
Chin Med J (Engl)
2013
;
126
:
3039
43
.
62.
Zou
S
,
Li
J
,
Zhou
H
,
Frech
C
,
Jiang
X
,
Chu
JS
, et al
Mutational landscape of intrahepatic cholangiocarcinoma
.
Nat Commun
2014
;
5
:
5696
.
63.
Shinada
K
,
Tsukiyama
T
,
Sho
T
,
Okumura
F
,
Asaka
M
,
Hatakeyama
S
. 
RNF43 interacts with NEDL1 and regulates p53-mediated transcription
.
Biochem Biophys Res Commun
2011
;
404
:
143
7
.
64.
Loregger
A
,
Grandl
M
,
Mejias-Luque
R
,
Allgauer
M
,
Degenhart
K
,
Haselmann
V
, et al
The E3 ligase RNF43 inhibits Wnt signaling downstream of mutated beta-catenin by sequestering TCF4 to the nuclear membrane
.
Sci Signal
2015
;
8
:
ra90
.
65.
Koo
BK
,
van Es
JH
,
van den Born
M
,
Clevers
H
. 
Porcupine inhibitor suppresses paracrine Wnt-driven growth of Rnf43;Znrf3-mutant neoplasia
.
Proc Natl Acad Sci U S A
2015
;
112
:
7548
50
.
66.
Deng
Y
,
Wu
X
. 
Peg3/Pw1 promotes p53-mediated apoptosis by inducing Bax translocation from cytosol to mitochondria
.
Proc Natl Acad Sci U S A
2000
;
97
:
12050
5
.
67.
Chiba
T
,
Zheng
YW
,
Kita
K
,
Yokosuka
O
,
Saisho
H
,
Onodera
M
, et al
Enhanced self-renewal capability in hepatic stem/progenitor cells drives cancer initiation
.
Gastroenterology
2007
;
133
:
937
50
.
68.
Parsons
DW
,
Li
M
,
Zhang
X
,
Jones
S
,
Leary
RJ
,
Lin
JC
, et al
The genetic landscape of the childhood cancer medulloblastoma
.
Science
2011
;
331
:
435
9
.
69.
Jones
S
,
Zhang
X
,
Parsons
DW
,
Lin
JC
,
Leary
RJ
,
Angenendt
P
, et al
Core signaling pathways in human pancreatic cancers revealed by global genomic analyses
.
Science
2008
;
321
:
1801
6
.
70.
Morin
RD
,
Mendez-Lago
M
,
Mungall
AJ
,
Goya
R
,
Mungall
KL
,
Corbett
RD
, et al
Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma
.
Nature
2011
;
476
:
298
303
.
71.
Biankin
AV
,
Waddell
N
,
Kassahn
KS
,
Gingras
MC
,
Muthuswamy
LB
,
Johns
AL
, et al
Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes
.
Nature
2012
;
491
:
399
405
.
72.
Hsu
M
,
Sasaki
M
,
Igarashi
S
,
Sato
Y
,
Nakanuma
Y
. 
KRAS and GNAS mutations and p53 overexpression in biliary intraepithelial neoplasia and intrahepatic cholangiocarcinomas
.
Cancer
2013
;
119
:
1669
74
.
73.
Dal Molin
M
,
Matthaei
H
,
Wu
J
,
Blackford
A
,
Debeljak
M
,
Rezaee
N
, et al
Clinicopathological correlates of activating GNAS mutations in intraductal papillary mucinous neoplasm (IPMN) of the pancreas
.
Ann Surg Oncol
2013
;
20
:
3802
8
.
74.
Fecteau
RE
,
Lutterbaugh
J
,
Markowitz
SD
,
Willis
J
,
Guda
K
. 
GNAS mutations identify a set of right-sided, RAS mutant, villous colon cancers
.
PLoS One
2014
;
9
:
e87966
.
75.
Dang
L
,
Jin
S
,
Su
SM
. 
IDH mutations in glioma and acute myeloid leukemia
.
Trends Mol Med
2010
;
16
:
387
97
.
76.
Saha
SK
,
Parachoniak
CA
,
Ghanta
KS
,
Fitamant
J
,
Ross
KN
,
Najem
MS
, et al
Mutant IDH inhibits HNF-4alpha to block hepatocyte differentiation and promote biliary cancer
.
Nature
2014
;
513
:
110
4
.
77.
Borger
DR
,
Zhu
AX
. 
IDH mutations: new genetic signatures in cholangiocarcinoma and therapeutic implications
.
Expert Rev Anticancer Ther
2012
;
12
:
543
6
.
78.
Cairns
RA
,
Mak
TW
. 
Oncogenic isocitrate dehydrogenase mutations: mechanisms, models, and clinical opportunities
.
Cancer Discov
2013
;
3
:
730
41
.
79.
Wu
YM
,
Su
F
,
Kalyana-Sundaram
S
,
Khazanov
N
,
Ateeq
B
,
Cao
X
, et al
Identification of targetable FGFR gene fusions in diverse cancers
.
Cancer Discov
2013
;
3
:
636
47
.
80.
Maisonneuve
C
,
Guilleret
I
,
Vick
P
,
Weber
T
,
Andre
P
,
Beyer
T
, et al
Bicaudal C, a novel regulator of Dvl signaling abutting RNA-processing bodies, controls cilia orientation and leftward flow
.
Development
2009
;
136
:
3019
30
.
81.
Arai
Y
,
Totoki
Y
,
Hosoda
F
,
Shirota
T
,
Hama
N
,
Nakamura
H
, et al
Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma
.
Hepatology
2014
;
59
:
1427
34
.
82.
Sia
D
,
Losic
B
,
Moeini
A
,
Cabellos
L
,
Hao
K
,
Revill
K
, et al
Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma
.
Nat Commun
2015
;
6
:
6087
.
83.
Churi
CR
,
Shroff
R
,
Wang
Y
,
Rashid
A
,
Kang
HC
,
Weatherly
J
, et al
Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications
.
PLoS One
2014
;
9
:
e115383
.
84.
Ballaro
C
,
Ceccarelli
S
,
Tiveron
C
,
Tatangelo
L
,
Salvatore
AM
,
Segatto
O
, et al
Targeted expression of RALT in mouse skin inhibits epidermal growth factor receptor signalling and generates a Waved-like phenotype
.
EMBO Rep
2005
;
6
:
755
61
.
85.
Ferby
I
,
Reschke
M
,
Kudlacek
O
,
Knyazev
P
,
Pante
G
,
Amann
K
, et al
Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation
.
Nat Med
2006
;
12
:
568
73
.
86.
Anastasi
S
,
Fiorentino
L
,
Fiorini
M
,
Fraioli
R
,
Sala
G
,
Castellani
L
, et al
Feedback inhibition by RALT controls signal output by the ErbB network
.
Oncogene
2003
;
22
:
4221
34
.
87.
Zhang
L
,
Jiang
L
,
Sun
Q
,
Peng
T
,
Lou
K
,
Liu
N
, et al
Prostaglandin E2 enhances mitogen-activated protein kinase/Erk pathway in human cholangiocarcinoma cells: involvement of EP1 receptor, calcium and EGF receptors signaling
.
Mol Cell Biochem
2007
;
305
:
19
26
.
88.
Nakamura
H
,
Arai
Y
,
Totoki
Y
,
Shirota
T
,
Elzawahry
A
,
Kato
M
, et al
Genomic spectra of biliary tract cancer
.
Nat Genet
2015
;
47
:
1003
10
.
89.
Pena-Llopis
S
,
Vega-Rubin-de-Celis
S
,
Liao
A
,
Leng
N
,
Pavia-Jimenez
A
,
Wang
S
, et al
BAP1 loss defines a new class of renal cell carcinoma
.
Nat Genet
2012
;
44
:
751
9
.
90.
Harbour
JW
,
Onken
MD
,
Roberson
ED
,
Duan
S
,
Cao
L
,
Worley
LA
, et al
Frequent mutation of BAP1 in metastasizing uveal melanomas
.
Science
2010
;
330
:
1410
3
.
91.
Bott
M
,
Brevet
M
,
Taylor
BS
,
Shimizu
S
,
Ito
T
,
Wang
L
, et al
The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma
.
Nat Genet
2011
;
43
:
668
72
.
92.
Kadoch
C
,
Crabtree
GR
. 
Mammalian SWI/SNF chromatin remodeling complexes and cancer: Mechanistic insights gained from human genomics
.
Sci Adv
2015
;
1
:
e1500447
.
93.
Amatu
A
,
Sartore-Bianchi
A
,
Siena
S
. 
NTRK gene fusions as novel targets of cancer therapy across multiple tumour types
.
ESMO Open
2016
;
1
:
e000023
.
94.
Hong
DS
,
Brose
MS
,
Doebele
RC
,
Shaw
AT
,
Dowlati
A
,
Bauer
TM
, et al
Clinical safety and activity from a phase I study of LOXO-101, a selective TRKA/B/C inhibitor, in solid-tumor patients with NTRK gene fusions
.
Mol Cancer Ther
2015
(
14
) (
12 Supplement 2
;
PR13
).
95.
Drilon
A
,
Siena
S
,
Ou
SI
,
Patel
M
,
Ahn
MJ
,
Lee
J
, et al
Safety and antitumor activity of the multi-targeted Pan-TRK, ROS1, and ALK inhibitor entrectinib (RXDX-101): combined results from two phase 1 trials (ALKA-372-001 and STARTRK-1)
.
Cancer Discov
2017
;
7
:
400
9
.
96.
Lee
KH
,
Lee
KB
,
Kim
TY
,
Han
SW
,
Oh
DY
,
Im
SA
, et al
Clinical and pathological significance of ROS1 expression in intrahepatic cholangiocarcinoma
.
BMC Cancer
2015
;
15
:
721
.
97.
Gu
TL
,
Deng
X
,
Huang
F
,
Tucker
M
,
Crosby
K
,
Rimkunas
V
, et al
Survey of tyrosine kinase signaling reveals ROS kinase fusions in human cholangiocarcinoma
.
PLoS One
2011
;
6
:
e15640
.
98.
Ruzzenente
A
,
Fassan
M
,
Conci
S
,
Simbolo
M
,
Lawlor
RT
,
Pedrazzani
C
, et al
Cholangiocarcinoma heterogeneity revealed by multigene mutational profiling: clinical and prognostic relevance in surgically resected patients
.
Ann Surg Oncol
2016
;
23
:
1699
707
.
99.
Andersen
JB
,
Spee
B
,
Blechacz
BR
,
Avital
I
,
Komuta
M
,
Barbour
A
, et al
Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors
.
Gastroenterology
2012
;
142
:
1021
31
.
100.
Farshidfar
F
,
Zheng
S
,
Gingras
MC
,
Newton
Y
,
Shih
J
,
Robertson
AG
, et al
Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles
.
Cell Rep
2017
;
18
:
2780
94
.
101.
Li
M
,
Zhang
Z
,
Li
X
,
Ye
J
,
Wu
X
,
Tan
Z
, et al
Whole-exome and targeted gene sequencing of gallbladder carcinoma identifies recurrent mutations in the ErbB pathway
.
Nat Genet
2014
;
46
:
872
6
.
102.
Borger
DR
,
Tanabe
KK
,
Fan
KC
,
Lopez
HU
,
Fantin
VR
,
Straley
KS
, et al
Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping
.
Oncologist
2012
;
17
:
72
9
.
103.
Deshpande
V
,
Nduaguba
A
,
Zimmerman
SM
,
Kehoe
SM
,
Macconaill
LE
,
Lauwers
GY
, et al
Mutational profiling reveals PIK3CA mutations in gallbladder carcinoma
.
BMC Cancer
2011
;
11
:
60
.
104.
O'Dell
MR
,
Huang
JL
,
Whitney-Miller
CL
,
Deshpande
V
,
Rothberg
P
,
Grose
V
, et al
Kras(G12D) and p53 mutation cause primary intrahepatic cholangiocarcinoma
.
Cancer Res
2012
;
72
:
1557
67
.
105.
Sekiya
S
,
Suzuki
A
. 
Intrahepatic cholangiocarcinoma can arise from Notch-mediated conversion of hepatocytes
.
J Clin Invest
2012
;
122
:
3914
8
.
106.
Fan
B
,
Malato
Y
,
Calvisi
DF
,
Naqvi
S
,
Razumilava
N
,
Ribback
S
, et al
Cholangiocarcinomas can originate from hepatocytes in mice
.
J Clin Invest
2012
;
122
:
2911
5
.
107.
Zender
S
,
Nickeleit
I
,
Wuestefeld
T
,
Sorensen
I
,
Dauch
D
,
Bozko
P
, et al
A critical role for notch signaling in the formation of cholangiocellular carcinomas
.
Cancer Cell
2013
;
23
:
784
95
.
108.
Guest
RV
,
Boulter
L
,
Kendall
TJ
,
Minnis-Lyons
SE
,
Walker
R
,
Wigmore
SJ
, et al
Cell lineage tracing reveals a biliary origin of intrahepatic cholangiocarcinoma
.
Cancer Res
2014
;
74
:
1005
10
.
109.
Liu
W
,
Chen
JR
,
Hsu
CH
,
Li
YH
,
Chen
YM
,
Lin
CY
, et al
A zebrafish model of intrahepatic cholangiocarcinoma by dual expression of hepatitis B virus X and hepatitis C virus core protein in liver
.
Hepatology
2012
;
56
:
2268
76
.
110.
Ko
KS
,
Peng
J
,
Yang
H
. 
Animal models of cholangiocarcinoma
.
Curr Opin Gastroenterol
2013
;
29
:
312
8
.
111.
Kalamarides
M
,
Niwa-Kawakita
M
,
Leblois
H
,
Abramowski
V
,
Perricaudet
M
,
Janin
A
, et al
Nf2 gene inactivation in arachnoidal cells is rate-limiting for meningioma development in the mouse
.
Genes Dev
2002
;
16
:
1060
5
.
112.
Xu
X
,
Kobayashi
S
,
Qiao
W
,
Li
C
,
Xiao
C
,
Radaeva
S
, et al
Induction of intrahepatic cholangiocellular carcinoma by liver-specific disruption of Smad4 and Pten in mice
.
J Clin Invest
2006
;
116
:
1843
52
.
113.
Kiguchi
K
,
Carbajal
S
,
Chan
K
,
Beltran
L
,
Ruffino
L
,
Shen
J
, et al
Constitutive expression of ErbB-2 in gallbladder epithelium results in development of adenocarcinoma
.
Cancer Res
2001
;
61
:
6971
6
.
114.
Ikenoue
T
,
Terakado
Y
,
Nakagawa
H
,
Hikiba
Y
,
Fujii
T
,
Matsubara
D
, et al
A novel mouse model of intrahepatic cholangiocarcinoma induced by liver-specific Kras activation and Pten deletion
.
Sci Rep
2016
;
6
:
23899
.
115.
Wang
P
,
Dong
Q
,
Zhang
C
,
Kuan
PF
,
Liu
Y
,
Jeck
WR
, et al
Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas
.
Oncogene
2013
;
32
:
3091
100
.
116.
Zhu
AX
,
Borger
DR
,
Kim
Y
,
Cosgrove
D
,
Ejaz
A
,
Alexandrescu
S
, et al
Genomic profiling of intrahepatic cholangiocarcinoma: refining prognosis and identifying therapeutic targets
.
Ann Surg Oncol
2014
;
21
:
3827
34
.
117.
Goyal
L
,
Govindan
A
,
Sheth
RA
,
Nardi
V
,
Blaszkowsky
LS
,
Faris
JE
, et al
Prognosis and clinicopathologic features of patients with advanced stage isocitrate dehydrogenase (IDH) mutant and IDH wild-type intrahepatic cholangiocarcinoma
.
Oncologist
2015
;
20
:
1019
27
.
118.
Kipp
BR
,
Voss
JS
,
Kerr
SE
,
Barr Fritcher
EG
,
Graham
RP
,
Zhang
L
, et al
Isocitrate dehydrogenase 1 and 2 mutations in cholangiocarcinoma
.
Hum Pathol
2012
;
43
:
1552
8
.
119.
Borger
DR
,
Goyal
L
,
Yau
T
,
Poon
RT
,
Ancukiewicz
M
,
Deshpande
V
, et al
Circulating oncometabolite 2-hydroxyglutarate is a potential surrogate biomarker in patients with isocitrate dehydrogenase-mutant intrahepatic cholangiocarcinoma
.
Clin Cancer Res
2014
;
20
:
1884
90
.
120.
Rohle
D
,
Popovici-Muller
J
,
Palaskas
N
,
Turcan
S
,
Grommes
C
,
Campos
C
, et al
An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells
.
Science
2013
;
340
:
626
30
.
121.
Wang
F
,
Travins
J
,
DeLaBarre
B
,
Penard-Lacronique
V
,
Schalm
S
,
Hansen
E
, et al
Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation
.
Science
2013
;
340
:
622
6
.
122.
Saha
SK
,
Gordan
JD
,
Kleinstiver
BP
,
Vu
P
,
Najem
MS
,
Yeo
JC
, et al
Isocitrate dehydrogenase mutations confer dasatinib hypersensitivity and SRC-dependence in intrahepatic cholangiocarcinoma
.
Cancer Discov
2016
;
6
:
727
39
.
123.
Graham
RP
,
Barr Fritcher
EG
,
Pestova
E
,
Schulz
J
,
Sitailo
LA
,
Vasmatzis
G
, et al
Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma
.
Hum Pathol
2014
;
45
:
1630
8
.
124.
Turner
N
,
Grose
R
. 
Fibroblast growth factor signalling: from development to cancer
.
Nat Rev Cancer
2010
;
10
:
116
29
.
125.
Javle
MM
,
Shroff
RT
,
Zhu
A
,
Sadeghi
S
,
Choo
SP
,
Borad
MJ
et al 
A phase 2 study of BGJ398 in patients (pts) with advanced or metastatic FGFR-altered cholangiocarcinoma (CCA) who failed or are intolerant to platinum-based chemotherapy
.
2016 Gastrointestinal Cancers Symposium
,
J Clin Oncol
34
, 
2016
(
suppl 4S; abstr 335
).
126.
Goyal
L
,
Saha
SK
,
Liu
LY
,
Siravegna
G
,
Leshchiner
I
,
Ahronian
LG
, et al
Polyclonal secondary FGFR2 mutations drive acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive cholangiocarcinoma
.
Cancer Discov
2017
;
7
:
252
63
.
127.
Mazzaferro
V
,
Shaib
W
,
Rimassa
L
,
Harris
WP
,
Personeni
N
,
El-Rayes
B
, et al
ARQ 087, an oral pan-fibroblast growth factor receptor (FGFR) inhibitor, in patients with advanced and/or metastatic intrahepatic cholangiocarcinoma (iCCA)
.
18th ESMO World Congress on Gastrointestinal Cancer
.
29 June -2 July, 2016, Barcelona, Spain
. 
2016
.
Abstract PD19
.
128.
Hall
TG
,
Savage
RE
,
Schwartz
B
,
Yu
Y
,
Wang
Y
,
Kazakin
J
et al 
In FGFR2 driven tumors, preclinical pharmacokinetics (PK), pharmacodynamics (PD) and efficacy translate into clinical activity of ARQ 087
.
Mol Cancer Ther
2015
;
14
(
12 Suppl 2
):
Abstract nr B151
.
129.
Liu
S
,
Quarles
LD
. 
How fibroblast growth factor 23 works
.
J Am Soc Nephrol
2007
;
18
:
1637
47
.
130.
Giatromanolaki
A
,
Koukourakis
MI
,
Simopoulos
C
,
Polychronidis
A
,
Sivridis
E
. 
Vascular endothelial growth factor (VEGF) expression in operable gallbladder carcinomas
.
Eur J Surg Oncol
2003
;
29
:
879
83
.
131.
Tang
D
,
Nagano
H
,
Yamamoto
H
,
Wada
H
,
Nakamura
M
,
Kondo
M
, et al
Angiogenesis in cholangiocellular carcinoma: expression of vascular endothelial growth factor, angiopoietin-1/2, thrombospondin-1 and clinicopathological significance
.
Oncol Rep
2006
;
15
:
525
32
.
132.
Yoshikawa
D
,
Ojima
H
,
Iwasaki
M
,
Hiraoka
N
,
Kosuge
T
,
Kasai
S
, et al
Clinicopathological and prognostic significance of EGFR, VEGF, and HER2 expression in cholangiocarcinoma
.
Br J Cancer
2008
;
98
:
418
25
.
133.
Benckert
C
,
Jonas
S
,
Cramer
T
,
Von
MZ
,
Schafer
G
,
Peters
M
, et al
Transforming growth factor beta 1 stimulates vascular endothelial growth factor gene transcription in human cholangiocellular carcinoma cells
.
Cancer Res
2003
;
63
:
1083
92
.
134.
Mobius
C
,
Demuth
C
,
Aigner
T
,
Wiedmann
M
,
Wittekind
C
,
Mossner
J
, et al
Evaluation of VEGF A expression and microvascular density as prognostic factors in extrahepatic cholangiocarcinoma
.
Eur J Surg Oncol
2007
;
33
:
1025
9
.
135.
Shirabe
K
,
Shimada
M
,
Tsujita
E
,
Aishima
S
,
Maehara
S
,
Tanaka
S
, et al
Prognostic factors in node-negative intrahepatic cholangiocarcinoma with special reference to angiogenesis
.
Am J Surg
2004
;
187
:
538
42
.
136.
Hida
Y
,
Morita
T
,
Fujita
M
,
Miyasaka
Y
,
Horita
S
,
Fujioka
Y
, et al
Vascular endothelial growth factor expression is an independent negative predictor in extrahepatic biliary tract carcinomas
.
Anticancer Res
1999
;
19
:
2257
60
.
137.
Zhu
AX
,
Meyerhardt
JA
,
Blaszkowsky
LS
,
Kambadakone
AR
,
Muzikansky
A
,
Zheng
H
, et al
Efficacy and safety of gemcitabine, oxaliplatin, and bevacizumab in advanced biliary-tract cancers and correlation of changes in 18-fluorodeoxyglucose PET with clinical outcome: a phase 2 study
.
Lancet Oncol
2010
;
11
:
48
54
.
138.
Lubner
SJ
,
Mahoney
MR
,
Kolesar
JL
,
Loconte
NK
,
Kim
GP
,
Pitot
HC
, et al
Report of a multicenter phase II trial testing a combination of biweekly bevacizumab and daily erlotinib in patients with unresectable biliary cancer: a phase II consortium study
.
J Clin Oncol
2010
;
28
:
3491
7
.
139.
Valle
JW
,
Wasan
H
,
Lopes
A
,
Backen
AC
,
Palmer
DH
,
Morris
K
, et al
Cediranib or placebo in combination with cisplatin and gemcitabine chemotherapy for patients with advanced biliary tract cancer (ABC-03): a randomised phase 2 trial
.
Lancet Oncol
2015
;
16
:
967
78
.
140.
Bengala
C
,
Bertolini
F
,
Malavasi
N
,
Boni
C
,
Aitini
E
,
Dealis
C
, et al
Sorafenib in patients with advanced biliary tract carcinoma: a phase II trial
.
Br J Cancer
2010
;
102
:
68
72
.
141.
El-Khoueiry
AB
,
Rankin
C
,
Lenz
HJ
,
Philip
P
,
Rivkin
SE
,
Blanke
CD
. 
SWOG 0514: A phase II study of sorafenib (BAY 43-9006) as single agent in patients (pts) with unresectable or metastatic gallbladder cancer or cholangiocarcinomas
.
Journal of Clinical Oncology
,
25
;
18_suppl
(
June 2007
)
4639
9
.
142.
El-Khoueiry
AB
,
Rankin
C
,
Siegel
AB
,
Iqbal
S
,
Gong
IY
,
Micetich
KC
, et al
S0941: a phase 2 SWOG study of sorafenib and erlotinib in patients with advanced gallbladder carcinoma or cholangiocarcinoma
.
Br J Cancer
2014
;
110
:
882
7
.
143.
Lee
JK
,
Capanu
M
,
O'Reilly
EM
,
Ma
J
,
Chou
JF
,
Shia
J
, et al
A phase II study of gemcitabine and cisplatin plus sorafenib in patients with advanced biliary adenocarcinomas
.
Br J Cancer
2013
;
109
:
915
9
.
144.
Moehler
M
,
Maderer
A
,
Schimanski
C
,
Kanzler
S
,
Denzer
U
,
Kolligs
FT
, et al
Gemcitabine plus sorafenib versus gemcitabine alone in advanced biliary tract cancer: a double-blind placebo-controlled multicentre phase II AIO study with biomarker and serum programme
.
Eur J Cancer
2014
;
50
:
3125
35
.
145.
Yi
JH
,
Thongprasert
S
,
Lee
J
,
Doval
DC
,
Park
SH
,
Park
JO
, et al
A phase II study of sunitinib as a second-line treatment in advanced biliary tract carcinoma: a multicentre, multinational study
.
Eur J Cancer
2012
;
48
:
196
201
.
146.
Santoro
A
,
Gebbia
V
,
Pressiani
T
,
Testa
A
,
Personeni
N
,
Arrivas
BE
, et al
A randomized, multicenter, phase II study of vandetanib monotherapy versus vandetanib in combination with gemcitabine versus gemcitabine plus placebo in subjects with advanced biliary tract cancer: the VanGogh study
.
Ann Oncol
2015
;
26
:
542
7
.
147.
Gwak
GY
,
Yoon
JH
,
Shin
CM
,
Ahn
YJ
,
Chung
JK
,
Kim
YA
, 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
.
148.
Nakazawa
K
,
Dobashi
Y
,
Suzuki
S
,
Fujii
H
,
Takeda
Y
,
Ooi
A
. 
Amplification and overexpression of c-erbB-2, epidermal growth factor receptor, and c-met in biliary tract cancers
.
J Pathol
2005
;
206
:
356
65
.
149.
Leone
F
,
Cavalloni
G
,
Pignochino
Y
,
Sarotto
I
,
Ferraris
R
,
Piacibello
W
, et al
Somatic mutations of epidermal growth factor receptor in bile duct and gallbladder carcinoma
.
Clin Cancer Res
2006
;
12
:
1680
5
.
150.
Paule
B
,
Herelle
MO
,
Rage
E
,
Ducreux
M
,
Adam
R
,
Guettier
C
, et al
Cetuximab plus gemcitabine-oxaliplatin (GEMOX) in patients with refractory advanced intrahepatic cholangiocarcinomas
.
Oncology
2007
;
72
:
105
10
.
151.
Gruenberger
B
,
Schueller
J
,
Heubrandtner
U
,
Wrba
F
,
Tamandl
D
,
Kaczirek
K
, et al
Cetuximab, gemcitabine, and oxaliplatin in patients with unresectable advanced or metastatic biliary tract cancer: a phase 2 study
.
Lancet Oncol
2010
;
11
:
1142
8
.
152.
Borbath
I
,
Ceratti
A
,
Verslype
C
,
Demols
A
,
Delaunoit
T
,
Laurent
S
, et al
Combination of gemcitabine and cetuximab in patients with advanced cholangiocarcinoma: a phase II study of the Belgian Group of Digestive Oncology
.
Ann Oncol
2013
;
24
:
2824
9
.
153.
Malka
D
,
Cervera
P
,
Foulon
S
,
Trarbach
T
,
de la
FC
,
Boucher
E
, et al
Gemcitabine and oxaliplatin with or without cetuximab in advanced biliary-tract cancer (BINGO): a randomised, open-label, non-comparative phase 2 trial
.
Lancet Oncol
2014
;
15
:
819
28
.
154.
Jensen
LH
,
Lindebjerg
J
,
Ploen
J
,
Hansen
TF
,
Jakobsen
A
. 
Phase II marker-driven trial of panitumumab and chemotherapy in KRAS wild-type biliary tract cancer
.
Ann Oncol
2012
;
23
:
2341
6
.
155.
Hezel
AF
,
Noel
MS
,
Allen
JN
,
Abrams
TA
,
Yurgelun
M
,
Faris
JE
, et al
Phase II study of gemcitabine, oxaliplatin in combination with panitumumab in KRAS wild-type unresectable or metastatic biliary tract and gallbladder cancer
.
Br J Cancer
2014
;
111
:
430
6
.
156.
Sohal
DP
,
Mykulowycz
K
,
Uehara
T
,
Teitelbaum
UR
,
Damjanov
N
,
Giantonio
BJ
, et al
A phase II trial of gemcitabine, irinotecan and panitumumab in advanced cholangiocarcinoma
.
Ann Oncol
2013
;
24
:
3061
5
.
157.
Vogel
A
,
Kasper
S
,
Weichert
W
,
Bitzer
M
,
Block
A
,
Riess
H
et al 
Panitumumab in combination with gemcitabine/cisplatin (GemCis) for patients with advanced kRAS WT biliary tract cancer: a randomized phase II trial of the Arbeitsgemeinschaft Internistische Onkologie (AIO)
.
J Clin Oncol
33
, 
2015
(
suppl; abstr 4082
).
158.
Leone
F
,
Marino
D
,
Cereda
S
,
Filippi
R
,
Belli
C
,
Spadi
R
, et al
Panitumumab in combination with gemcitabine and oxaliplatin does not prolong survival in wild-type KRAS advanced biliary tract cancer: a randomized phase 2 trial (Vecti-BIL study)
.
Cancer
2016
;
122
:
574
81
.
159.
Philip
PA
,
Mahoney
MR
,
Allmer
C
,
Thomas
J
,
Pitot
HC
,
Kim
G
, et al
Phase II study of erlotinib in patients with advanced biliary cancer
.
J Clin Oncol
2006
;
24
:
3069
74
.
160.
Lee
J
,
Park
SH
,
Chang
HM
,
Kim
JS
,
Choi
HJ
,
Lee
MA
, et al
Gemcitabine and oxaliplatin with or without erlotinib in advanced biliary-tract cancer: a multicentre, open-label, randomised, phase 3 study
.
Lancet Oncol
2012
;
13
:
181
8
.
161.
Chiorean
EG
,
Ramasubbaiah
R
,
Yu
M
,
Picus
J
,
Bufill
JA
,
Tong
Y
, et al
Phase II trial of erlotinib and docetaxel in advanced and refractory hepatocellular and biliary cancers: Hoosier Oncology Group GI06-101
.
Oncologist
2012
;
17
:
13
.
162.
Galdy
S
,
Lamarca
A
,
McNamara
MG
,
Hubner
RA
,
Cella
CA
,
Fazio
N
, et al
HER2/HER3 pathway in biliary tract malignancies; systematic review and meta-analysis: a potential therapeutic target?
Cancer Metastasis Rev
2016
;
36
:
141
57
.
163.
Ramanathan
RK
,
Belani
CP
,
Singh
DA
,
Tanaka
M
,
Lenz
HJ
,
Yen
Y
, et al
A phase II study of lapatinib in patients with advanced biliary tree and hepatocellular cancer
.
Cancer Chemother Pharmacol
2009
;
64
:
777
83
.
164.
Peck
J
,
Wei
L
,
Zalupski
M
,
O'Neil
B
,
Villalona
CM
,
Bekaii-Saab
T
. 
HER2/neu may not be an interesting target in biliary cancers: results of an early phase II study with lapatinib
.
Oncology
2012
;
82
:
175
9
.
165.
Sorscher
S
. 
Marked radiographic response of a HER-2-overexpressing biliary cancer to trastuzumab
.
Cancer Manag Res
2013
;
9
:
1
3
.
166.
Roa
I
,
de Toro
G
,
Schalper
K
,
de
A X
,
Churi
C
,
Javle
M
. 
Overexpression of the HER2/neu gene: a new therapeutic possibility for patients with advanced gallbladder cancer
.
Gastrointest Cancer Res
2014
;
7
:
42
8
.
167.
Suder
A
,
Ang
JE
,
Kyle
F
,
Harris
D
,
Rudman
S
,
Kristeleit
R
, et al
A phase I study of daily afatinib, an irreversible ErbB family blocker, in combination with weekly paclitaxel in patients with advanced solid tumours
.
Eur J Cancer
2015
;
51
:
2275
84
.
168.
Settakorn
J
,
Kaewpila
N
,
Burns
GF
,
Leong
AS
. 
FAT, E-cadherin, beta catenin, HER 2/neu, Ki67 immuno-expression, and histological grade in intrahepatic cholangiocarcinoma
.
J Clin Pathol
2005
;
58
:
1249
54
.
169.
Chen
W
,
Liang
J
,
Huang
L
,
Cai
J
,
Lei
Y
,
Lai
J
, et al
Characterizing the activation of the Wnt signaling pathway in hilar cholangiocarcinoma using a tissue microarray approach
.
Eur J Histochem
2016
;
60
:
2536
.
170.
Wang
W
,
Zhong
W
,
Yuan
J
,
Yan
C
,
Hu
S
,
Tong
Y
, et al
Involvement of Wnt/beta-catenin signaling in the mesenchymal stem cells promote metastatic growth and chemoresistance of cholangiocarcinoma
.
Oncotarget
2015
;
6
:
42276
89
.
171.
Shen
DY
,
Zhang
W
,
Zeng
X
,
Liu
CQ
. 
Inhibition of Wnt/beta-catenin signaling downregulates P-glycoprotein and reverses multi-drug resistance of cholangiocarcinoma
.
Cancer Sci
2013
;
104
:
1303
8
.
172.
Sugimachi
K
,
Taguchi
K
,
Aishima
S
,
Tanaka
S
,
Shimada
M
,
Kajiyama
K
, et al
Altered expression of beta-catenin without genetic mutation in intrahepatic cholangiocarcinoma
.
Mod Pathol
2001
;
14
:
900
5
.
173.
Tokumoto
N
,
Ikeda
S
,
Ishizaki
Y
,
Kurihara
T
,
Ozaki
S
,
Iseki
M
, et al
Immunohistochemical and mutational analyses of Wnt signaling components and target genes in intrahepatic cholangiocarcinomas
.
Int J Oncol
2005
;
27
:
973
80
.
174.
Jusakul
A
,
Kongpetch
S
,
Teh
BT
. 
Genetics of Opisthorchis viverrini-related cholangiocarcinoma
.
Curr Opin Gastroenterol
2015
;
31
:
258
63
.
175.
Hirata
K
,
Ajiki
T
,
Okazaki
T
,
Horiuchi
H
,
Fujita
T
,
Kuroda
Y
. 
Frequent occurrence of abnormal E-cadherin/beta-catenin protein expression in advanced gallbladder cancers and its association with decreased apoptosis
.
Oncology
2006
;
71
:
102
10
.
176.
Yadav
A
,
Gupta
A
,
Yadav
S
,
Rastogi
N
,
Agrawal
S
,
Kumar
A
, et al
Association of Wnt signaling pathway genetic variants in gallbladder cancer susceptibility and survival
.
Tumour Biol
2016
;
37
:
8083
95
.
177.
Rosenbluh
J
,
Wang
X
,
Hahn
WC
. 
Genomic insights into WNT/beta-catenin signaling
.
Trends Pharmacol Sci
2014
;
35
:
103
9
.
178.
Eads
JR
Lipika
G
,
stein
S
,
El-Khoueiry
A
,
Manji
GA
,
Abrams
TA
et al 
Phase I study of DKN-01, an anti-DKK1 antibody, in combination with gemcitabine (G) and cisplatin (C) in patients (pts) with advanced biliary cancer
.
J Clin Oncol
2016
34
:
15
(
suppl,abstract e15603
).
179.
Fingas
CD
,
Mertens
JC
,
Razumilava
N
,
Sydor
S
,
Bronk
SF
,
Christensen
JD
, et al
Polo-like kinase 2 is a mediator of hedgehog survival signaling in cholangiocarcinoma
.
Hepatology
2013
;
58
:
1362
74
.
180.
Mertens
JC
,
Fingas
CD
,
Christensen
JD
,
Smoot
RL
,
Bronk
SF
,
Werneburg
NW
, et al
Therapeutic effects of deleting cancer-associated fibroblasts in cholangiocarcinoma
.
Cancer Res
2013
;
73
:
897
907
.
181.
Fingas
CD
,
Bronk
SF
,
Werneburg
NW
,
Mott
JL
,
Guicciardi
ME
,
Cazanave
SC
, et al
Myofibroblast-derived PDGF-BB promotes Hedgehog survival signaling in cholangiocarcinoma cells
.
Hepatology
2011
;
54
:
2076
88
.
182.
Xie
F
,
Xu
X
,
Xu
A
,
Liu
C
,
Liang
F
,
Xue
M
, et al
Aberrant activation of Sonic hedgehog signaling in chronic cholecystitis and gallbladder carcinoma
.
Hum Pathol
2014
;
45
:
513
21
.
183.
Li
J
,
Wu
T
,
Lu
J
,
Cao
Y
,
Song
N
,
Yang
T
, et al
Immunohistochemical evidence of the prognostic value of hedgehog pathway components in primary gallbladder carcinoma
.
Surg Today
2012
;
42
:
770
5
.
184.
Tang
L
,
Tan
YX
,
Jiang
BG
,
Pan
YF
,
Li
SX
,
Yang
GZ
, et al
The prognostic significance and therapeutic potential of hedgehog signaling in intrahepatic cholangiocellular carcinoma
.
Clin Cancer Res
2013
;
19
:
2014
24
.
185.
Matsushita
S
,
Onishi
H
,
Nakano
K
,
Nagamatsu
I
,
Imaizumi
A
,
Hattori
M
, et al
Hedgehog signaling pathway is a potential therapeutic target for gallbladder cancer
.
Cancer Sci
2014
;
105
:
272
80
.
186.
Riedlinger
D
,
Bahra
M
,
Boas-Knoop
S
,
Lippert
S
,
Bradtmoller
M
,
Guse
K
, et al
Hedgehog pathway as a potential treatment target in human cholangiocarcinoma
.
J Hepatobiliary Pancreat Sci
2014
;
21
:
607
15
.
187.
Gherardi
E
,
Birchmeier
W
,
Birchmeier
C
,
Vande
WG
. 
Targeting MET in cancer: rationale and progress
.
Nat Rev Cancer
2012
;
12
:
89
103
.
188.
Miyamoto
M
,
Ojima
H
,
Iwasaki
M
,
Shimizu
H
,
Kokubu
A
,
Hiraoka
N
, et al
Prognostic significance of overexpression of c-Met oncoprotein in cholangiocarcinoma
.
Br J Cancer
2011
;
105
:
131
8
.
189.
Terada
T
,
Nakanuma
Y
,
Sirica
AE
. 
Immunohistochemical demonstration of MET overexpression in human intrahepatic cholangiocarcinoma and in hepatolithiasis
.
Hum Pathol
1998
;
29
:
175
80
.
190.
Li
H
,
Shimura
H
,
Aoki
Y
,
Date
K
,
Matsumoto
K
,
Nakamura
T
, et al
Hepatocyte growth factor stimulates the invasion of gallbladder carcinoma cell lines in vitro
.
Clin Exp Metastasis
1998
;
16
:
74
82
.
191.
Yang
L
,
Guo
T
,
Jiang
S
,
Yang
Z
. 
Expression of ezrin, HGF and c-met and its clinicopathological significance in the benign and malignant lesions of the gallbladder
.
Hepatogastroenterology
2012
;
59
:
1769
75
.
192.
Moon
WS
,
Park
HS
,
Lee
H
,
Pai
R
,
Tarnawski
AS
,
Kim
KR
, et al
Co-expression of cox-2, C-met and beta-catenin in cells forming invasive front of gallbladder cancer
.
Cancer Res Treat
2005
;
37
:
171
6
.
193.
Engelman
JA
,
Zejnullahu
K
,
Mitsudomi
T
,
Song
Y
,
Hyland
C
,
Park
JO
, et al
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling
.
Science
2007
;
316
:
1039
43
.
194.
Pennacchietti
S
,
Michieli
P
,
Galluzzo
M
,
Mazzone
M
,
Giordano
S
,
Comoglio
PM
. 
Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene
.
Cancer Cell
2003
;
3
:
347
61
.
195.
Goyal
L
,
Yurgelun
MB
,
Abrams
TA
,
Kwak
EL
,
Cleary
JM
,
Knowles
M
, et al
A Phase II trial of Cabozantinib (XL-184) in Patients with Advanced Cholangiocarcinoma
.
J Clin Oncol
33
, 
2015
(
suppl 3; abstr 800
).
196.
O'Neill
E
,
Kolch
W
. 
Conferring specificity on the ubiquitous Raf/MEK signalling pathway
.
Br J Cancer
2004
;
90
:
283
8
.
197.
Khokhlatchev
AV
,
Canagarajah
B
,
Wilsbacher
J
,
Robinson
M
,
Atkinson
M
,
Goldsmith
E
, et al
Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation
.
Cell
1998
;
93
:
605
15
.
198.
Chen
TC
,
Jan
YY
,
Yeh
TS
. 
K-ras mutation is strongly associated with perineural invasion and represents an independent prognostic factor of intrahepatic cholangiocarcinoma after hepatectomy
.
Ann Surg Oncol
2012
;
19
Suppl 3
:
S675
81
.
199.
Pai
RK
,
Mojtahed
K
,
Pai
RK
. 
Mutations in the RAS/RAF/MAP kinase pathway commonly occur in gallbladder adenomas but are uncommon in gallbladder adenocarcinomas
.
Appl Immunohistochem Mol Morphol
2011
;
19
:
133
40
.
200.
Goeppert
B
,
Frauenschuh
L
,
Renner
M
,
Roessler
S
,
Stenzinger
A
,
Klauschen
F
, et al
BRAF V600E-specific immunohistochemistry reveals low mutation rates in biliary tract cancer and restriction to intrahepatic cholangiocarcinoma
.
Mod Pathol
2014
;
27
:
1028
34
.
201.
Saetta
AA
,
Papanastasiou
P
,
Michalopoulos
NV
,
Gigelou
F
,
Korkolopoulou
P
,
Bei
T
, et al
Mutational analysis of BRAF in gallbladder carcinomas in association with K-ras and p53 mutations and microsatellite instability
.
Virchows Arch
2004
;
445
:
179
82
.
202.
Tannapfel
A
,
Sommerer
F
,
Benicke
M
,
Katalinic
A
,
Uhlmann
D
,
Witzigmann
H
, et al
Mutations of the BRAF gene in cholangiocarcinoma but not in hepatocellular carcinoma
.
Gut
2003
;
52
:
706
12
.
203.
Hyman
DM
,
Puzanov
I
,
Subbiah
V
,
Faris
JE
,
Chau
I
,
Blay
JY
, et al
Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations
.
N Engl J Med
2015
;
373
:
726
36
.
204.
Voss
JS
,
Holtegaard
LM
,
Kerr
SE
,
Fritcher
EG
,
Roberts
LR
,
Gores
GJ
, et al
Molecular profiling of cholangiocarcinoma shows potential for targeted therapy treatment decisions
.
Hum Pathol
2013
;
44
:
1216
22
.
205.
Bekaii-Saab
T
,
Phelps
MA
,
Li
X
,
Saji
M
,
Goff
L
,
Kauh
JS
, et al
Multi-institutional phase II study of selumetinib in patients with metastatic biliary cancers
.
J Clin Oncol
2011
;
29
:
2357
63
.
206.
Bridgewater
J
,
Lopes
A
,
Beare
S
,
Duggan
M
,
Lee
D
,
Ricamara
M
, et al
A phase 1b study of selumetinib in combination with cisplatin and gemcitabine in advanced or metastatic biliary tract cancer: the ABC-04 study
.
BMC Cancer
2016
;
16
:
153
2174
.
207.
Herberger
B
,
Puhalla
H
,
Lehnert
M
,
Wrba
F
,
Novak
S
,
Brandstetter
A
, et al
Activated mammalian target of rapamycin is an adverse prognostic factor in patients with biliary tract adenocarcinoma
.
Clin Cancer Res
2007
;
13
:
4795
9
.
208.
Riener
MO
,
Bawohl
M
,
Clavien
PA
,
Jochum
W
. 
Rare PIK3CA hotspot mutations in carcinomas of the biliary tract
.
Genes Chromosomes Cancer
2008
;
47
:
363
7
.
209.
Chung
JY
,
Hong
SM
,
Choi
BY
,
Cho
H
,
Yu
E
,
Hewitt
SM
. 
The expression of phospho-AKT, phospho-mTOR, and PTEN in extrahepatic cholangiocarcinoma
.
Clin Cancer Res
2009
;
15
:
660
7
.
210.
Lee
D
,
Do
IG
,
Choi
K
,
Sung
CO
,
Jang
KT
,
Choi
D
, et al
The expression of phospho-AKT1 and phospho-MTOR is associated with a favorable prognosis independent of PTEN expression in intrahepatic cholangiocarcinomas
.
Mod Pathol
2012
;
25
:
131
9
.
211.
Ahn
DH
,
Li
J
,
Wei
L
,
Doyle
A
,
Marshall
JL
,
Schaaf
LJ
, et al
Results of an abbreviated phase-II study with the Akt inhibitor MK-2206 in patients with advanced biliary cancer
.
Sci Rep
2015
;
5
:
12122
.
212.
Yeung
YH
,
Chionh
FJM
,
Price
TJ
,
Scott
AM
,
Tran
H
,
Fang
G
et al 
Phase II study of everolimus monotherapy as first-line treatment in advanced biliary tract cancer: RADichol
.
J Clin Oncol
32
:
5s
, 
2014
(
suppl; abstr 4101
).
213.
Brand
M
,
Measures
AR
,
Wilson
BG
,
Cortopassi
WA
,
Alexander
R
,
Hoss
M
, et al
Small molecule inhibitors of bromodomain-acetyl-lysine interactions
.
ACS Chem Biol
2015
;
10
:
22
39
.
214.
Marks
EI
,
Yee
NS
. 
Immunotherapeutic approaches in biliary tract carcinoma: current status and emerging strategies
.
World J Gastrointest Oncol
2015
;
7
:
338
46
.
215.
Kaida
M
,
Morita-Hoshi
Y
,
Soeda
A
,
Wakeda
T
,
Yamaki
Y
,
Kojima
Y
, et al
Phase 1 trial of Wilms tumor 1 (WT1) peptide vaccine and gemcitabine combination therapy in patients with advanced pancreatic or biliary tract cancer
.
J Immunother
2011
;
34
:
92
9
.
216.
Yamamoto
K
,
Ueno
T
,
Kawaoka
T
,
Hazama
S
,
Fukui
M
,
Suehiro
Y
, et al
MUC1 peptide vaccination in patients with advanced pancreas or biliary tract cancer
.
Anticancer Res
2005
;
25
:
3575
9
.
217.
Lepisto
AJ
,
Moser
AJ
,
Zeh
H
,
Lee
K
,
Bartlett
D
,
McKolanis
JR
, et al
A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors
.
Cancer Ther
2008
;
6
:
955
64
.
218.
Kobayashi
M
,
Sakabe
T
,
Abe
H
,
Tanii
M
,
Takahashi
H
,
Chiba
A
, et al
Dendritic cell-based immunotherapy targeting synthesized peptides for advanced biliary tract cancer
.
J Gastrointest Surg
2013
;
17
:
1609
17
.
219.
Aruga
A
,
Takeshita
N
,
Kotera
Y
,
Okuyama
R
,
Matsushita
N
,
Ohta
T
, et al
Phase I clinical trial of multiple-peptide vaccination for patients with advanced biliary tract cancer
.
J Transl Med
2014
;
12
:
61
.
220.
Aruga
A
,
Takeshita
N
,
Kotera
Y
,
Okuyama
R
,
Matsushita
N
,
Ohta
T
, et al
Long-term vaccination with multiple peptides derived from cancer-testis antigens can maintain a specific T-cell response and achieve disease stability in advanced biliary tract cancer
.
Clin Cancer Res
2013
;
19
:
2224
31
.
221.
Yoshitomi
M
,
Yutani
S
,
Matsueda
S
,
Ioji
T
,
Komatsu
N
,
Shichijo
S
, et al
Personalized peptide vaccination for advanced biliary tract cancer: IL-6, nutritional status and pre-existing antigen-specific immunity as possible biomarkers for patient prognosis
.
Exp Ther Med
2012
;
3
:
463
9
.
222.
Shimizu
K
,
Kotera
Y
,
Aruga
A
,
Takeshita
N
,
Takasaki
K
,
Yamamoto
M
. 
Clinical utilization of postoperative dendritic cell vaccine plus activated T-cell transfer in patients with intrahepatic cholangiocarcinoma
.
J Hepatobiliary Pancreat Sci
2012
;
19
:
171
8
.
223.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Aparicio
SA
,
Behjati
S
,
Biankin
AV
, et al
Signatures of mutational processes in human cancer
.
Nature
2013
;
500
:
415
21
.
224.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
225.
Rashid
A
,
Ueki
T
,
Gao
YT
,
Houlihan
PS
,
Wallace
C
,
Wang
BS
, et al
K-ras mutation, p53 overexpression, and microsatellite instability in biliary tract cancers: a population-based study in China
.
Clin Cancer Res
2002
;
8
:
3156
63
.
226.
Silva
VW
,
Askan
G
,
Daniel
TD
,
Lowery
M
,
Klimstra
DS
,
Abou-Alfa
GK
, et al
Biliary carcinomas: pathology and the role of DNA mismatch repair deficiency
.
Chin Clin Oncol
2016
;
5
:
62
.
227.
Suto
T
,
Habano
W
,
Sugai
T
,
Uesugi
N
,
Kanno
S
,
Saito
K
, et al
Infrequent microsatellite instability in biliary tract cancer
.
J Surg Oncol
2001
;
76
:
121
6
.
228.
Kohya
N
,
Miyazaki
K
,
Matsukura
S
,
Yakushiji
H
,
Kitajima
Y
,
Kitahara
K
, et al
Deficient expression of O(6)-methylguanine-DNA methyltransferase combined with mismatch-repair proteins hMLH1 and hMSH2 is related to poor prognosis in human biliary tract carcinoma
.
Ann Surg Oncol
2002
;
9
:
371
9
.
229.
Tran
E
,
Turcotte
S
,
Gros
A
,
Robbins
PF
,
Lu
YC
,
Dudley
ME
, et al
Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer
.
Science
2014
;
344
:
641
5
.
230.
Bang
YJ
,
Doi
T
,
De Braud
F
,
Piha-Paul
S
,
Hollebecque
A
,
Abdul Razak
AR
, et al
Safety and efficacy of pembrolizumab (MK-3475) in patients (pts) with advanced biliary tract cancer: Interim results of KEYNOTE-028
.
Eur Cancer Congress
2015
,
Vienna, Abstract 525
.