Abstract
Cholangiocarcinomas (CCAs) are diverse epithelial tumors arising from the liver or large bile ducts with features of cholangiocyte differentiation, and are classified anatomically into intrahepatic (iCCA), perihilar (pCCA), and distal CCA (dCCA). Each subtype has distinct risk factors, molecular pathogenesis, therapeutic options, and prognosis. CCA is an aggressive malignancy with a poor overall prognosis and median survival of less than 2 years in patients with advanced disease. Potentially curative surgical treatment options are limited to the subset of patients with early stage disease. Presently, the available systemic medical therapies for advanced or metastatic CCA have limited therapeutic efficacy. Molecular alterations define the differences in biological behavior of each CCA subtype. Recent comprehensive genetic analysis have better characterized the genomic and transcriptomic landscape of each CCA subtype. Promising candidates for targeted, personalized therapy have emerged including potential driver FGFR gene fusions and somatic mutations in IDH 1/2 in iCCA, PRKACA or PRKACB gene fusions in pCCA, and ELF3 mutations in dCCA/ampullary carcinoma. A precision genomic medicine approach is dependent on an enhanced understanding of driver mutations in each subtype and stratification of patients according to their genetic drivers. We review the current genomic landscape of CCA, potentially actionable molecular aberrations in each CCA subtype, and role of immunotherapy in CCA.
Keywords: Distal cholangiocarcinoma, intrahepatic cholangiocarcinoma, immunotherapy, perihilar cholangiocarcinoma, targeted therapy
INTRODUCTION
Cholangiocarcinoma (CCA) is a heterogeneous hepatobiliary malignancy with a dismal prognosis. CCAs are epithelial tumors with markers of cholangiocyte differentiation (1). On the basis of their anatomic location, CCAs are classified into intrahepatic (iCCA), perihilar (pCCA) and distal (dCCA) subtypes (1). iCCAs arise above the second order bile ducts, whereas pCCAs are located between the insertion of the cystic duct and the second order bile ducts, and dCCAs are located below the insertion of the cystic duct (1). Each anatomic subtype has a distinct biologic behavior, therapeutic options, and prognosis (2). In addition to the anatomic definition of CCA, stratification by histopathologic and growth type patterns has also been proposed (3–5). CCA arising from large bile ducts, predominantly but not exclusively pCCA, is characterized by well/moderately differentiated mucin producing cylindrical cells and a periductal infiltrating growth pattern. In contrast, CCA developing from small ducts and or hepatocytes, largely iCCA, are characterized by cuboidal non-mucin producing cells with a mass forming growth pattern. The clinical implications of this classification have yet to be realized and therefore, we will use the anatomic definition.
CCA is a devastating malignancy with an abysmal overall 5-year survival rate of less than 10% (6). Surgical resection and liver transplantation are potentially curative treatment option for early stage disease in all three subtypes. However, the median 5-year survival after R0-resection is approximately 30% (7). Liver transplantation as a potentially curative treatment option is limited to iCCAs and pCCAs. Neoadjuvant chemoradiation followed by liver transplantation has been established as definitive therapy for a subset of carefully selected pCCA patients (1). Five-year survival rates following liver transplantation are 70% for patients with pCCA in the setting of primary sclerosing cholangitis and 55% for sporadic or de novo pCCA patients. Until recently, the presence of iCCA had been considered a contraindication for liver transplantation. According to an international multicenter study, patients with “very early” iCCA (tumor size < 2 cm) had a 5-year survival rate of 65% following liver transplantation compared to 45% in the “advanced” group (tumor size > 2 cm) (8). Despite these encouraging results, the preponderance of patients have advanced disease at diagnosis. For patients who are not candidates for surgical resection or liver transplantation, the practice standard is systemic chemotherapy with gemcitabine and cisplatin (9). However, this combination chemotherapeutic regimen confers a median overall survival of only 11.7 months (9). Currently, there are no potentially curative medical therapies for CCA. Furthermore, no targeted molecular therapies have been approved for use in CCA. Development of potentially curative medical treatment strategies for CCA has been limited by the molecular and genetic heterogeneity of these tumors. The advent of next-generation sequencing has made the discovery of possible targetable or actionable molecular alterations in CCA feasible. Precision therapy for CCA is dependent on an enhanced understanding of molecular and genetic aberrations, including driver mutations, for each CCA subtype. Herein, we review the evolving mutational landscape of CCA and summarize novel targeted therapies that will help build a precision approach for treatment of this devastating malignancy.
GENOMIC AND TRANSCRIPTOMIC LANDSCAPE OF CHOLANGIOCARCINOMA DIFFER BY ETIOLOGY
It is well-known that genomic alterations in CCA vary by anatomic subtype. However, different etiologic exposures can also significantly influence the pattern of somatic mutations leading to a distinct mutational landscape. This varying impact of carcinogenic etiologies was initially demonstrated in a whole-exome sequencing analysis of eight liver fluke-associated CCAs (10). Opisthorchis viverrini (O. viverrini)-related tumors had 206 somatic mutations which included mutations in known cancer-related genes, such as tumor protein 53 (TP53) which was mutated in 44.4% of cases, Kristen ras sarcoma viral oncogene homolog (KRAS) and SMAD4, both mutated in 16.7% of cases (10). KRAS mutations were associated with a poor overall survival (10). Somatic mutations were also identified in several newly implicated genes with distinct biological functions. Genes involved in genomic stability and G-protein signaling which had somatic mutations included GNAS, ROBO2, RNF43, PEG3, XIRP2, RADIL, NDC80, PCDHA13 and LAMA2 (10). Inactivating mutations in MLL3, which has a role in histone modification, were also identified (10). Whole-exome sequencing of 108 cases of O. viverrini related CCA and 101 cases of non- O. viverrini highlighted the impact of different causative etiologies (11). Somatic mutations in BAP1, IDH1, and IDH2 were a more frequent occurrence in non-O. viverrini CCAs, whereas TP53 mutations were observed more frequently in O. viverrini CCAs. The detection of recurrent somatic mutations in BAP1 and ARID1A was a novel finding in this study. Moreover, a role of chromatin remodeling in CCA carcinogenesis was ascertained by functional assays demonstrating a tumor suppressive function for BAP1 and ARID1A (11). A subsequent whole-exome sequencing of 32 intrahepatic CCAs also identified inactivating mutations in multiple chromatin-remodeling genes including BAP1, ARID1A and PBRM1 (12). Together, these studies underscored the importance of dysregulated chromatin remodeling in non-O. viverrini CCA carcinogenesis.
MOLECULAR ABERRATIONS AND TARGETED THERAPIES IN CHOLANGIOCARCINOMA
Molecular Aberrations and Targeted Therapies in Intrahepatic Cholangiocarcinoma (Figure 1, Table 1)
Table 1.
Target | Agent | Trial Description | NCT Number |
---|---|---|---|
FGFR | NVP-BGJ398 | Phase II trial in patients with advanced CCA harboring FGFR gene fusions/aberrations | NCT02150967 |
FGFR | JNJ-42756493 | Phase I trial in patients with advanced solid organ malignancy or lymphoma | NCT01703481 |
FGFR | ARQ 087 | Phase 1/II trial in patients with solid organ malignancy and FGFR genetic alterations (including iCCA patients with FGFR2 gene fusions) | NCT01752920 |
FGFR | TAS-120 | Phase I trial in patients with advanced solid malignancy or multiple myeloma with or without FGF/FGFR-related abnormalities | NCT02052778 |
FGFR | CH5183284/Debio 1347 | Phase I trial in patients with solid organ malignancy and genetic aberration of FGFR1, 2, or 3 | NCT01948297 |
FGFR2 | Ponatinib | Phase II trial in patients with advanced cholangiocarcinoma harboring FGFR2 gene fusions | NCT02265341 |
FGFR2 | FPA144 | Phase I trial in patients with advanced solid tumors with FGFR2b overexpression/amplification | NCT02318329 |
IDH1 | AG-120 | Phase I trial in patients with advanced solid tumors harboring an IDH1 mutation | NCT02073994 |
IDH1 | AG-120 | Phase III, multi-center, double-blind, placebo-controlled trial in patients with advanced cholangiocarcinoma harboring IDH1 mutations | NCT02989857 |
MEK | Trametinib | Phase II trial in patients with advanced CCA or gallbladder cancer | NCT02042443 |
MEK, VEGFR/PDGFR/Raf | GSK1120212 plus Pazopanib | Phase I trial in patients with advanced solid tumors | NCT01438554 |
PD-1 | Pembrolizumab | Phase II trial in patients with advanced solid tumors | NCT02628067 |
PD-1 | MK-3475 (Pembrolizumab) | Phase I/II trial in patients with advanced gastrointestinal tumors | NCT02268825 |
FGFR Gene Fusions and Inhibitors
FGF signaling regulates a multitude of biological processes including cell proliferation, differentiation, survival, wound repair, angiogenesis, and migration (13). The integral role of the FGF-FGFR axis in essential cellular processes fosters the oncogenic potential of aberrant FGF signaling. Deregulation of FGF signaling with consequent carcinogenesis has been implicated in various malignancies including CCA. FGFR2 gene fusions have been detected in iCCA in several recent studies (14–17). In three different CCA cohorts, FGFR2 gene fusions were identified in 11–14% of iCCAs with report of several different FGFR2 gene fusions including FGFR2-BICC1, FGFR2-AHCYL1, FGFR2-TACC3, and FGFR2-KIAA 1598 (15, 16, 18). A higher frequency of FGFR gene fusions was noted by Sia et al. in a cohort of 107 iCCA patients (45%, 17/107) (19). FGFR2 gene fusions are rare in pCCA/dCCA. Mechanistic studies indicate that the FGFR binding partners mediate oligomerization and resultant activation of the respective FGFR kinase in tumors harboring FGFR translocations (17). Moreover, FGFR2 fusion proteins activate FGFR signaling with mitogen-activated protein kinase (MAPK) activation and confer anchorage-independent growth (18). Uncovering of these gene fusions is significant as these are often driver mutations and potentially targetable. For instance, targeting of the BCR-ABL gene fusion with Imatinib in chronic myelogenous leukemia has been one of the earliest instances of effective precision medicine (20).
Intrahepatic cholangiocarcinomas harboring FGFR2 gene fusions appear to have distinct clinical and pathologic features (15). Morphologically, cases harboring FGFR2 gene fusions appear to have a prominent intraductal growth pattern, anastomosing tubular structures, and desmoplasia (15). Immunohistochemically, iCCAs with FGFR2 gene fusions have strong cytokeratin (CK)-7 positivity but only weak and patchy expression of CK-19 (15). In addition to describing the pathologic features of tumors with FGFR2 gene fusions, Graham et al. also reported a survival advantage (median cancer specific survival 123 months versus 37 months) indicating that the presence of FGFR2 gene fusions may have prognostic significance (15). However, in an Asian cohort the presence of FGFR2 gene fusions in iCCA did not appear to have an impact on overall survival, clinical stage or tumor differentiation (18).
FGFR-Selective Small molecule kinase inhibitors
Preclinical studies have demonstrated antitumor efficacy of FGFR inhibition selectively in cells harboring FGFR2 gene fusions (17, 18). Moreover, BGJ398, a pan-FGFR inhibitor, significantly reduced tumor burden in a genetic murine model of CCA as well as a patient-derived xenograft model of iCCA (21). These promising data support the notion that patients who harbor FGFR2 gene fusions may benefit from FGFR-directed therapy in a precision medicine approach. On this premise, several small molecule kinase inhibitors (SMKIs) of FGFR are currently in early phase clinical trials. Evaluation of BGJ398 in a phase I trial single-agent dose escalation and dose-expansion study in 132 patients with advanced solid organ malignancies harboring FGFR gene aberrations demonstrated antitumor activity in several tumor types as well as a manageable safety profile (22). Interim analysis from a phase II, multi-center study of BGJ398 in patients with advanced CCA with FGFR2 gene fusions or other FGFR genetic alterations who have failed platinum-based chemotherapy (NCT02150967) demonstrated an impressive disease control rate of 82% (23). JNJ-42756493 or erdafitinib is another oral pan-FGFR selective SMKI being evaluated in clinical trials (24). Results of a phase I dose-escalation study of JNJ-42756493 in patients with advanced solid tumors and FGFR pathway aberrations indicated 4 confirmed responses, one unconfirmed partial response and disease stability in 16 patients (24). Notably, the 36 patients in this study without known FGFR alterations did not demonstrate any significant response (24). ARQ 087 is an ATP-competitive SMKI with anti-proliferative activity in cell lines driven by FGFR aberrations including amplifications, fusions, and mutations (25). ARQ 087 is currently being studied in a phase 1/2 trial in patients with advanced solid tumors with genetic alterations including iCCA patients with FGFR2 gene fusions (NCT01752920). Other FGFR SMKIs which have demonstrated efficacy against FGFR-driven malignancies in preclinical studies and are currently being assessed in early phase clinical trials include TAS-120, AZD4547, and CH5183284/Debio 1347 (NCT02052778, NCT01948297).
Non-Selective Small molecule kinase inhibitors
Borad et al. demonstrated early evidence of efficacy of the non-selective FGFR inhibitors ponatinib and pazopanib in advanced iCCA patients harboring FGFR2 gene fusions (14). In a metastatic iCCA patient with the FGFR2-MGEA5 fusion, ponatinib monotherapy as salvage treatment resulted in tumor necrosis, reduction in levels of the tumor marker carbohydrate antigen 19-9, and shrinkage in size of metastatic lymph nodes (14). In a patient with metastatic iCCA and FGFR2-TACC3 fusion, partial response to pazopanib was noted. Ponatinib is currently being assessed in an active phase II clinical trial in patients with advanced biliary tract cancer harboring FGFR2 gene fusions detected by next generation sequencing or FISH break-apart assay (NCT02265341). Combination chemotherapies are an attractive option in malignancies with marked molecular heterogeneity such as CCA. Deregulation of the Raf/MEK/ERK pathway is a well-established occurrence in CCA and the MEK inhibitor Selumetinib has been shown to have some benefit in advanced biliary tract cancer (26). A phase I trial to assess the combination of pazopanib and the MEK inhibitor trametinib in advanced solid organ malignancies including CCA is currently underway (NCT01438554).
A significant shortcoming of first-generation FGFR inhibitors including BGJ398 and AZD457 is the eventual emergence of drug-resistant tumors (27). Goyal et al. identified different FGFR2 mutations in individual resistant clones in patients with acquired resistance to BGJ398 (28). Preclinical studies have indicated that mutations in the FGFR gatekeeper residue can confer resistance to FGFR inhibitors. Tan et al. described novel, next-generation covalent FGFR inhibitors, FGFR irreversible inhibitors 2 and 3, which can overcome the gatekeeper residue mutation and potently inhibit tumor cells dependent upon FGFR1 and FGFR2 gatekeeper mutants (27).
Monoclonal antibodies
FGFRs have a variety of isoforms through tissue-specific alternative splicing of their mRNAs (29). Borad et al. detected the FGFR-IIIb isoform in all of the identified FGFR2 gene fusions in their study (14). The FGFR2-IIIb isoform has binding specificity for FGF7 and FGF10. Monoclonal antibodies directed against specific isoforms would theoretically be an attractive therapeutic option in advanced iCCA cases harboring FGFR gene aberrations as they would avoid the off-target effects of SMKIs. FPA144, a humanized monoclonal antibody directed against the FGFR2b isoform with demonstrated efficacy in preclinical tumor models of gastric cancer (29), is being evaluated in a phase I clinical trial in patients with advanced solid tumors with FGFR2b overexpression or amplification (NCT02318329).
MET-HGF
MET tyrosine kinase is a plasma membrane protein which is activated when its extracellular domain binds to hepatocyte growth factor (HGF) or scatter factor (30). HGF-MET signaling has a critical role in essential cellular behaviors including proliferation, resistance to apoptosis, increased cell motility, and angiogenesis (30). Tumors can harness these processes to promote tumor growth and invasion. MET overexpression occurs in iCCA and correlates with the degree of tumor differentiation (31). An integrated molecular analysis identified two distinct biological classes in iCCA, an inflammation class (38% of iCCAs) and a proliferation class (62% of iCCAs) (32). The proliferation class featured activation of oncogenic signaling pathways such as MET, epidermal growth factor receptor (EGFR), and MAPK (32). Cross-talk between MET-HGF and ERRB family of receptors occurs and may account for resistance to MET and ERBB2 inhibitors (30). MET amplification has been detected in iCCA, albeit the incidence of these mutations is relatively low (16, 33).
A number of MET kinase inhibitors are being evaluated in clinical trials for various malignancies (34). Phase I results of a study of tivantinib, an oral MET inhibitor, in combination with gemcitabine in patients with solid tumors including CCA demonstrated partial response in 46% of patients and stable disease in 27% of patients (35). A phase II study of cabozantinib in patients with advanced CCA after progression on first or second line chemotherapy is currently ongoing (NCT01954745).
Mcl-1 and JAK/STAT Pathway
Myeloid cell leukemia sequence 1 (Mcl-1) is a potent antiapoptotic Bcl-2 protein. MCL1 amplification occurs in iCCA (16–21%) (16, 33). Mcl-1 mediates tumor necrosis factor-related apoptosis ligand (TRAIL) resistance in CCA by blocking the mitochondrial pathway of cell death (36). Interleukin (IL)-6, an inflammatory cytokine implicated in CCA biology, upregulates Mcl-1 via an AKT-dependent mechanism (37). IL-6 inhibition overcomes apoptosis resistance via downregulation of AKT and Mcl-1 (37). IL-6 upregulation of Mcl-1 is also mediated via Janus kinase (JAK) and signal transducer and activator of transcription (STAT) signaling cascade (38). STAT3 directly regulates Mcl-1 transcription suggesting that STAT3 inhibition may have therapeutic utility in CCA (38). Epigenetic silencing of suppressor of cytokine signaling 3 (SOCS-3) mediates sustained IL-6/STAT-3 signaling and enhanced Mcl-1 expression in CCA (39). Furthermore, biliary transduction of constitutively-activated AKT and yes-associated protein and systemic IL-33 administration induces tumor formation in mice via an IL-6 sensitive mechanism, underscoring the role of inflammatory cytokines in CCA oncogenesis (40). Mcl-1 amplification is a critical event in many cancers as cancer cells with Mcl-1 amplification are dependent on this potent anti-apoptotic protein for survival (41). Detection of Mcl-1 amplification utilizing FISH analysis is an essential component of a FISH probe set with high specificity for the detection of cholangiocarcinoma (42). Collectively, these findings provide several modes of therapeutic targeting in CCA. Novel, selective Mcl-1 inhibitors have been developed and preliminary preclinical studies have demonstrated evidence of their efficacy in pancreatic cancer (43, 44). Mechanistic studies have also demonstrated that S63845, a small molecule inhibitor of Mc-1 which binds with high affinity to the BH3-binding groove of Mcl-1, potently induces cell death in various solid cancer-derived cell lines (45). Preclinical studies have also demonstrated efficacy of the JAK2 inhibitor, AZD1480, in suppression of STAT-3 mediated oncogenesis in solid tumor cell lines (46).
IDH/IDH2 Mutations and Inhibitors
Isocitrate dehydrogenase 1 and 2 are enzymes that catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate. IDH mutations are heterozygous point mutations in catalytic amino acid residues and these alteration occur in Arginine 132 of IDH1 and Arginine 140 or Arginine 172 of IDH2 (47). IDH1/IDH2 mutations results in elevated levels of 2-hydroxyglutarate, an oncometabolite which induces widespread epigenetic changes. Consequently, IDH mutations have pleiotropic effect on differentiation, growth factor dependence, collagen processing and hypoxia signaling (47).
IDH1/IDH2 mutations, albeit a relatively common occurrence in gliomas, were previously thought to be rare in other solid organ tumors (48). However, several recent mutational profiling studies have demonstrated that IDH mutations are a relatively frequent genetic aberration in CCA (16, 33, 49). IDH mutations are more frequently observed in intrahepatic cholangiocarcinomas than pCCAs and dCCAs (23–28% versus 0–7%) (48, 50). Histopathologic analysis of 94 surgically resected primary cholangiocarcinomas demonstrated that IDH mutations are associated with clear cell change and poorly differentiated histology (48). In this study, patients with IDH1/2 gene mutations also appeared to have a better overall survival a year after surgical resection compared to patients without the presence of IDH mutations (48). A positive prognostic association with IDH mutations was also observed in a study of 326 iCCAs which demonstrated that IDH mutations were associated with longer overall survival and longer time to tumor recurrence after iCCA resection (51). However, subsequent studies have demonstrated that the prognostic significance of IDH mutations remains unclear. In a smaller cohort of 34 iCCAs, Jiao et al. demonstrated that patients with IDH mutations had a 3-year survival of 33% compared to 81% for those with wild-type IDH genes, although patients with IDH mutations were somewhat older and had a higher tumor stage (12). Additionally, the sample size of this study (n = 32) was much smaller compared to the prior study. A subsequent mutational profiling of 200 resected iCCA specimens demonstrated that although IDH-mutant tumors were more frequently multicentric in the liver, there was no impact on long-term prognosis (49). Although these 4 studies had conflicting results regarding prognosis, they focused mainly on early-stage or resectable iCCA, whereas targeted therapy with IDH inhibitors would be considered in patients with unresectable or advanced iCCA (52). Accordingly, in a subsequent study Goyal et al. assessed the correlation of IDH mutations with prognosis in 104 patients with unresectable or advanced iCCA (52). The presence of IDH mutations did not have a significant impact on the median overall survival (52).
The finding that IDH mutations are a relatively frequent occurrence in several human malignancies logically lead to speculation whether inhibition of mutant IDH activity may have therapeutic benefits. Small molecule inhibitors of mutant IDH were assessed in two proof of concept preclinical studies (53, 54). AGI-5198, a selective IDH1 inhibitor, blocked the ability of this enzyme to produce the oncometabolite 2-HG with resultant impaired growth of IDH1-mutant cells (53). Likewise, AGI-6780, a selective mutant IDH2 inhibitor, induced differentiation in hematopoietic cell lines (54). AG-120 is a first-in-class, orally bioavailable inhibitor of mutant IDH1. Phase 1 results of AG-120 in solid organ malignancies (NCT02073994) including iCCA presented at the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics in 2015 demonstrated that AG-120: i) is well-tolerated in the solid tumor patient population, ii) had encouraging evidence of clinical activity, iii) reduced intra-tumoral 2-HG levels. On the basis of these preliminary results, an advanced phase multi-center, randomized, double-blind, placebo-controlled study of AG-120 in previously treated subjects with unresectable or metastatic cholangiocarcinoma with an IDH1 mutation is planned (NCT02989857). AG-221, an orally available, selective inhibitor of mutant IDH2, has received fast-track designation from the Food and Drug Administration (FDA) and is currently being evaluated in multiple clinical trials including a Phase 1/2, multi-center study in subjects with advanced solid tumors including iCCA who harbor an IDH2 mutation (NCT02273739). Saha et al. recently demonstrated that iCCA cells with mutant IDH have a unique dependency on SRC kinase (55). Consequently, the SRC inhibitor, dasatinib, had a significant anti-tumor effect in mutant IDH xenografts, signifying a potential new therapeutic strategy against mutant IDH iCCA (55)
Protein tyrosine phosphatases (PTPs)
Members of the protein tyrosine phosphatase (PTP) are enzymes which remove phosphate from tyrosine residues in proteins (56). Genetic aberrations of PTPs can result in disequilibrium of protein tyrosine-kinase phosphatase activity with consequent oncogenic potential (56). A prevalence screen of 124 pairs of iCCAs and nontumor samples demonstrated that 41.4% of iCCAs had gain of function mutations in PTPN3 (57). Presence of these mutations conferred markedly enhanced pro-tumor activity and significantly increased postoperative tumor recurrence (57). This study has important therapeutic implications as it is well-known that gain of function mutations are potential therapeutic targets in various malignancies. Consequently, the high frequency of these mutations and their significant functional relevance makes these mutations an important potential therapeutic target.
Chromatin-remodeling Genes and HDAC Inhibitors
The role of dysregulated chromatin remodeling in iCCA was highlighted through exome sequencing of 32 iCCAs which detected genetic aberration of at least one chromatin-remodeling gene in 47% of cases (15 of 32) (12). Inactivating mutations were frequently identified in BAP1, ARID1A and PBRM1. BAP1 encodes a nuclear deubiquitinase involved in chromatin remodeling, whereas ARID1A and PBRM1 encode subunits of the SWI/SNF chromatin remodeling complexes (12). Subjects with mutations in any one of these genes tended to have shorter survival times and worse survival compared to subjects with wild-type genes, albeit these observations were not statistically significant (12). Frequent alterations of ARID1A (36% of iCCAs) were noted in next-generation sequencing of 28 iCCAs (16). ARID1A mutations appear to be more frequent in iCCA than pCCA/dCCA as indicated by a recent mutational profiling which identified ARID1A aberrations in 20% of iCCAs compared to 5% of pCCA/dCCAs (33). BAP1 and PBRM1 alterations were identified in 9% and 11% of iCCAs, respectively (33). BAP1 mutations were also detected in 10% of pCCA/dCCA cases, and in these patients the presence of BAP1 mutation was significantly associated with reduced progression-free survival (median 3 months vs. 8.8 months) and reduced overall survival (median 8.9 vs. 19.9 months) (33). Overall, in all cases harboring BAP1 mutations in this study, progression after first-line chemotherapy was observed within 4 months (33).
Several small molecule inhibitors targeting chromatin remodeling have been approved by the FDA (58). These include the histone deacetylase (HDAC) inhibitors vorinostat and romidepsin and DNA methyltransferase (DNMT) inhibitors azacitidine and decitabine (58). As the role of chromatin remodeling in CCA carcinogenesis is being illuminated, these inhibitors may have increased therapeutic utility in CCA.
Molecular Aberrations and Targeted Therapies in Perihilar Cholangiocarcinoma (Figure 2)
EGFR/HER2
Aberrant expression and signaling of the epidermal growth factor receptor (ERBB) family of receptor tyrosine kinases has been reported in cholangiocarcinoma. Activation of this pathway leads to downstream oncogenic pathway activation including the MAPK pathway. The ERBB family has four distinct receptors including ERBB1 (EGFR), ERBB2 (Her-2/neu), ERBB3 and ERBB4 (59). In a cohort of surgically resected CCA cases, HER2 amplification was identified using FISH analysis in a single iCCA case and two pCCA/dCCA cases (15). A recent mutation profiling of 75 CCA specimens identified ERBB2 genetic aberrations in 20% of pCCAs/dCCAs and in only 1.8% of iCCAs (33). The pCCA/dCCA ERBB2 alterations included one amplification and four mutations, the latter being a novel finding, as mutations had previously not been reported in CCA. These mutations included one in the kinase domain (V777L) and three in the extracellular domain (S310F) (33). The V777L mutation is an activating mutation with some demonstrated resistance to lapatinib, a reversible, dual inhibitor of EGFR and HER2 (60). However, the V777L mutation was sensitive to nertatinib, an irreversible, dual inhibitor of EGFR and HER2 (60). The presence of kinase domain mutations may explain the limited success of erlotinib, a reversible EGFR inhibitor, in human CCA clinical trials (61, 62). Irreversible EGFR inhibitors such as neratinib, afatinib, and dacomatinib may have greater utility in this setting (33).
Protein kinase cyclic AMP (cAMP)-activated catalytic subunit alpha PRKACA) and beta (PRKACB)
Protein kinase A (PKA) is a cAMP-dependent protein kinase. Both PRKACA and PRKACB are catalytic submits of PKA and are members of the serine/threonine protein kinase family (63). Novel gene fusions of the PKA signaling components and mitochondrial ATP synthase, α subunit (ATP1B) were recently detected in pCCA/dCCA (63). Nakumura et al. detected ATP1B-PRKACA and ATP1B-PRKACB fusions in pCCA/dCCA cases (63). These fusions stimulated significantly enhanced expression of the PRKACA and PRKACB genes with consequent downstream MAPK signaling activation (63). Interestingly, fusion of the same exon of the PRKACA gene to DNAJB1 has been demonstrated in fibrolamellar hepatocellular carcinoma (64). Genetic aberrations in the PKA regulatory subunits, PRKAR1A (nonsense mutation) and PRKAR1B (PRKAR1B- C7orf50 gene fusion), were also detected in two different cases of pCCA/dCCA. PRKAR1A expression is enhanced in CCA, and PRKAR1A knockdown induces growth inhibition and apoptosis of CCA cells with resultant decrease in MAPK, phosphatidylinositol 3-kinase (PI3K)/Akt, JAT/STAT, and Wnt/β-catenin pathway signaling (65). Isoquinoline H89, a small molecule inhibitor of PKA, significantly inhibited CCA cell proliferation, albeit this may be an off-target effect as H89 is known to have a number of PKA-independent effects (65).
Molecular Aberrations and Targeted Therapies in dCCA/ampullary carcinoma (Figure 3)
Distal CCA as an entity is not well-described. Ampullary carcinomas are rare neoplasms arising from the ampulla of Vater at the convergence of three different epithelia – pancreatic, biliary and duodenal (66). Intestinal and pancreatobiliary histological subtypes of ampullary carcinoma have been proposed (66). The pancreatobiliary subtype can be considered a subset of dCCA, and thus the genetic landscape of ampullary carcinoma can be extrapolated to dCCA given their significant overlap. Moreover, preponderance of recent genomic analyses of CCA have grouped pCCA and dCCA as extrahepatic CCA. Therefore, there is substantial overlap in the molecular aberrations identified in these two subtypes and the potential targeted therapies.
ELF3
ELF3 encodes an E26 transformation-specific transcription factor and hence is implicated in the regulation of several genes with essential roles in multiple cellular processes (67). By interacting with their promoter regions ELF3 increases transcription of TGFBR2 and EGF1 activation, two known tumor suppressor gene (68). TGFBR2 also has an integral role in TGF-β signaling. Two recent genomic analyses detected inactivating mutations of ELF3 in ampullary carcinoma/dCCA. In a cohort of 98 periampullary carcinomas and 44 dCCAs, Gingras et al. identified inactivating frameshift or nonsense mutations in 10.6% of periampullary tumors (68). Similarly, ELF3 mutations had been reported in 9.5% of extrahepatic CCAs (pCCA/dCCA) in a molecular characterization of biliary tract cancers (63). In a cohort of 172 ampullary carcinomas, driver mutations in ELF3 were also identified in 13.3% of cases (69). ELF3 mutations were present at high allele frequencies, indicating that ELF3 mutation may represent a founder or driver mutation in ampullary carcinoma (69). Functional studies were subsequently carried out and demonstrated that ELF3 knockdown promotes motility and invasion in epithelial cells (69).
HER2 (ERBB2)/ERBB3
Molecular alterations in the ERBB family are well-described in pCCA (please see above) and have recently been reported in dCCA/ampullary carcinoma. ERBB3 mutations were identified in 14% of pancreatobiliary-type ampullary carcinomas and ERBB2 mutations were identified in 11.6% of ampullary carcinomas (69). ERBB2 mutations overlapped with ELF3 patients.
Molecular Aberrations and Targeted Therapies Common to all Cholangiocarcinoma Subtypes (Table 1)
Although each of the anatomic subtypes of CCA has a distinct mutational landscape, there are some molecular alterations that are observed with similar frequency in all of the subtypes. KRAS mutations and the activation of the PI3K-AKT-mTOR pathway are amongst the most prevalent aberrations noted in a number of malignancies and have a significant role in carcinogenesis.
KRAS
Activating mutations of the proto-oncogene KRAS are one of the most frequently encountered genetic mutations in CCA. The rate of KRAS mutations appear to be higher in pCCA/dCCA (40%) compared to iCCA (9%–24%) (16, 33, 49). The presence of these mutations has prognostic utility as patients harboring KRAS mutations have worse progression-free and overall survival (33, 70). Moreover, the KRAS mutant tumors are more likely to have adjacent organ involvement and R1 margin status (49). KRAS activation leads to upregulation of downstream effector pathways including PI3k-AKT-mammalian target of rapamycin (mTOR) and Raf/MEK/ERK. Presently, there are no effective direct inhibitors of KRAS and therefore the therapeutic approach for KRAS mutant tumors is inhibition of the downstream pathways. Selumetinib, a selective MEK 1/2 inhibitor, had demonstrated some efficacy in a phase II trial of advanced biliary tract cancers (26). Subsequently, phase I results of a study with selumetinib in combination with gemcitabine and cisplatin in advanced biliary tract cancer demonstrated a median progression-free survival of 6.4 months and acceptable adverse event profile (71). Other early phase trials of MEK inhibitors with conventional chemotherapy or tyrosine kinase inhibitors in biliary tract cancer are ongoing (NCT02042443, NCT01438554).
PI3K-AKT-mTOR Pathway
The PI3K pathway is activated by stimulation of several different receptor tyrosine kinases (e.g. c-MET, FGFR) with consequent activation of AKT (72). This in turn phosphorylates the mTOR complex which is integral in regulation of several cellular processes including proliferation, survival, tumor invasion, metabolism and angiogenesis (72, 73). Consequently, the activation of the PI3K-AKT-mTOR pathway mediates oncogenesis in a multitude of malignancies including CCA (74–76). Exome sequencing has identified mutations in multiple family members of the PI3K pathway including inactivating mutations of PIK3R1 and activating mutations of PIK3CA, PIK3C2A, PIK3C2G (12, 63). Activating mutations of PIK3CA have been reported in 4–8% of iCCAs and 4% of pCCA/dCCA cases (16, 49, 63).
Presently, there is a multitude of clinical trials investigation various inhibitors of the PI3K-AKT-mTOR pathway in different solid organ malignancies (77). Selective inhibition of mTOR can lead to feedback activation of AKT (78). However, preclinical data suggests that AKT inhibition can potentiate the anti-tumor efficacy of mTOR inhibition (78). The presence of functional redundancy within this pathway and extensive cross-talk with other pathways such as the RAF-MEK-ERK pathway makes combination chemotherapeutic approaches an attractive option. Preclinical data has demonstrated that dual targeting of the PI3K/AKT/mTOR and RAF-MEK-ERK pathways has synergistic effects in CCA (79). However, phase I trial of the PI3K inhibitor BKM120 in combination with mFOLFOX6 (a common chemotherapeutic regimen in gastrointestinal malignancies) revealed increased toxicity with the combination compared to both as single agents (80). Conversely, a phase I trial of the mTOR inhibitor everolimus in combination with gemcitabine and cisplatin, the current practice standard for metastatic CCA, in patients with biliary tract cancer demonstrated stable disease in 6/10 patients and an acceptable safety profile (81).
S100A4
S100A4, a cytoskeleton-associated calcium binding protein, has garnered recent attention for its role in metastasis of various malignancies including cholangiocarcinoma (82, 83). Depending on its subcellular localization, S100A4 contributes to the regulation of a multitude of cell biological processes including proliferation, survival, differentiation, and cytoskeletal rearrangement (82). S100A4 expression has prognostic implications as well, as it is has been identified in the subset of patients with worse outcomes in several malignancies including cholangiocarcinoma (83, 84). Fabris et al. identified enhanced nuclear expression of S100A4 in the CCA patients with a worse overall survival following surgical resection (83). Furthermore, nuclear expression of S100A4 was linked with increased CCA cell invasiveness and metastasization, suggesting that it may be a potential therapeutic target in CCA (83). A subsequent study demonstrated that low-dose paclitaxel down-regulated nuclear S100A4 expression. Consequently, a mouse xenograft model treated with low-dose paclitaxel had reduced lung metastasis, albeit no significant effect on local tumor growth (85). Overall, these findings support the notion that therapeutic targeting of S100A4 may have utility in hindering metastatic progression of cholangiocarcinoma.
Hedgehog Signaling Pathway
The Hedgehog (HH) signaling pathway is an evolutionary conserved, developmental pathway which is known to be deregulated in a number of malignancies including cholangiocarcinoma (86, 87). Preclinical studies have demonstrated that hedgehog signaling inhibition with cyclopamine hinders CCA cell proliferation, migration, and invasion and promotes tumor suppression in vivo (86, 88). Hh signaling-induced apoptosis resistance in CCA is mediated by polo-like kinase 2 (PLK2), a cell division regulating kinase (89). Profiling of human iCCA as well as pCCA/dCCA has demonstrated the presence of PLK immunoreactive cells (89). Accordingly, Hh signaling inhibition by cyclopamine reduces PLK expression (89). The role of the non-canonical Hh pathway in CCA has been examined as CCA cells often do not express cilia, a prerequisite for canonical Hh signaling (90). Indeed, the non-canonical Hh pathway promotes tumor progression in CCA. Moreover, a small molecule antagonist of the Hh plasma membrane protein Smoothened, vismodegib, resulted in tumor suppression and inhibition of metastasis in a preclinical CCA model (90). Another small molecule Hh antagonist, BMS-833923, had a more profound tumor suppressive effect in CCA xenografts when combined with gemcitabine, compared to either treatment alone (91).
Mesothelin
Mesothelin, a cell surface protein expressed in normal mesothelial cells, has aberrant tissue expression in a several malignancies including pancreatic adenocarcinoma and CCA (92, 93). Moreover, mesothelin expression appears to have prognostic implications in CCA. In a cohort of 61 surgically resected pCCA and dCCA specimens, mesothelin expression was associated with intrahepatic metastasis, peritoneal metastasis, and worse overall patient survival (94). A series of surgically resected iCCAs also demonstrated an association of mesothelin expression with poor overall survival following surgical resection (95). On the basis of these findings, mesothelin has become a potential target for antibody-based cancer therapy (96). Consequently, several therapeutic agents are under investigation in preclinical and clinical studies. Amatuximab, a chimeric monoclonal antimesothelin antibody, combined with cisplatin was well-tolerated and had a stable disease rate of 90% in patients with advanced pleural mesothelioma (97). However, a phase I study of amatuximab in 17 patients with advanced solid tumors including pancreatic adenocarcinomas demonstrated that only 3 patients had stable disease (98). Phase I results of SS1P, a recombinant anti-mesothelin immunotoxin, in 33 patients with mesothelin expressing malignancies including pancreatic adenocarcinoma, 19 had stable disease and 4 had minor responses (99). A phase II trial of SS1P and pentostatin plus cyclophosphamide in mesothelin expressing malignancies including pancreatic adenocarcinoma is currently ongoing (NCT01362790).
IMMUNOTHERAPY IN CHOLANGIOCARCINOMA (TABLE 1)
Immune checkpoints are essential for the maintenance of self-tolerance and prevention of normal tissue damage during an immune response. However, these checkpoints are dysregulated in cancer and tumors can harness them as an important immune resistance mechanism (100). Accordingly, activating therapeutic antitumor immunity via blockade of immune checkpoints holds significant potential in cancer therapy.
The immune system has several coinhibitory pathways for maintenance of T-cell tolerance (101). The finding of the critical role of cytotoxic t-lymphocyte associated protein 4 (CTLA-4) in T-cell downregulation was paradigm shifting and spurred the notion that coinhibitory signals block anti-tumor T-cell responses and removal of these signals would have an anti-tumor effect (101). This revolutionary concept ultimately resulted in the use of anti-CTLA-4 monoclonal antibody as effective cancer therapy in patients (102, 103). The same premise lead to therapeutic targeting of the programmed death 1(PD-1)/programmed death ligand-1 (PD-L1) pathway with significant response in human clinical trials for a number of malignancies (104).
A recent molecular characterization of 260 biliary tract cancers demonstrated that 45.2% of the cases had increased expression of immune checkpoint molecules (63). Interestingly, the cluster of patients with the worst outcomes had enrichment of genes involved in the immune system (63). Ye et al. reported significantly enhanced expression of PD-1 and PD-L1 in tumor tissue from surgically resected iCCA specimens compared to adjacent tissue (105). Cases with higher tumor tissue PD-L1 expression were characterized by poor histological differentiation and had more advanced pTNM staging (105). Moreover, the presence of higher PD-L1 expression was inversely correlated with CD8-positive tumor infiltrating lymphocytes (105). In another series of iCCA cases, a favorable clinical course was noted in cases with positive HLA class 1 antigen expression and negative PD-L1 expression, indicating that HLA expression defects in combination with PD-L1 expression may provide an immune escape mechanism for iCCAs (106).
Clinical experience with immune checkpoint inhibitors is very preliminary at this point. Early data from a phase I trial of pembrolizumab, a humanized monoclonal antibody against PD-1, in patients with PD-L1 positive advanced biliary tract cancer demonstrated encouraging evidence of efficacy (107). Overall, 4 patients (17%) had partial response, 4 patients (17%) had stable disease, and 12 patients (52%) had progressive disease (107). A phase II trial of pembrolizumab in advanced solid tumors is currently ongoing (NCT02628067). In another phase II trial, the combination of pembrolizumab and mFOLFOX6 is being investigated in patients with advanced gastrointestinal malignancies including CCA (NCT02268825).
FUTURE PERSPECTIVES
CCA, albeit an orphan disease, is beginning to attract attention from investigators and industry at an accelerated pace, given its lethality (108). Sophisticated clinical trials will require improved methods to stratify CCA patients and the time has come to abandon the inclusion of all patients with biliary tract cancers into a single trial. Future trials must stratify patients according to their anatomic subtype, genetic drivers, and stage of disease. Biomarker-guided information will also be imperative in the development of effective medical therapies for CCA. Towards this end, a well-developed clinical staging system for pCCA has been proposed (109); but even this staging system requires incorporation of genetic and biomarker information. We await the results of the FGFR and IDH directed therapies described above; however, it is unlikely their effects will be durable even if efficacious in a subset of patients. Cancers are extremely heterogeneous even within a single mass and, unfortunately, efficacious single agent targeted therapy usually results in emergence of resistant clones over time. CCA contain a rich tumor microenvironment including cancer associated fibroblasts (CAF), tumor associated macrophages, and lymphocytes.(110–112). The tumor microenvironment can be exploited to treat CCA. For example, cancer associated fibroblasts can be therapeutically targeted in CCA using BH3 mimetics in preclinical models and such strategies should be applied to the human cancer (112). Immunotherapy to overcome T-cell exhaustion using so called check point inhibitors is also likely to be useful in a subset of CCA patients as described above. However, more attention must also be given to inducing modes of immunogenic cell death to aid immunotherapy strategies (113). In the future, one can envision a portfolio of therapies to treat this disease guided by genetic biomarkers (e.g., FGFR2 directed therapy for a CCA with FGFR2 fusion aberrations) coupled to a checkpoint inhibitor (if the CCA also expresses checkpoint modulators such as PD-L1), or a CAF-directed therapy (if the CCA is highly desmoplastic). We await with eagerness (and impatience) for such combination therapies targeting both CCA cell specific genetic abberations and the tumor microenvironment – our patients have little time.
Acknowledgments
Financial Support: This work was supported by National Institutes of Health grants DK59427 (G.J.G.) DK84567 (Mayo Center for Cell Signaling in Gastroenterology) and the Mayo Foundation. S. Rizvi also received support from the Cholangiocarcinoma Foundation.
We thank Ms. Courtney Hoover for excellent secretarial support.
Abbreviations
- 2-HG
2-hydroxyglutarate
- BAP1
BRCA1-associated protein 1
- CCA
cholangiocarcinoma
- dCCA
distal cholangiocarcinoma
- EGFR
epidermal growth factor receptor
- FDA
Food and Drug Administration
- FISH
fluorescence in situ hybridization
- FGFR
fibroblast growth factor receptor
- HGF
hepatocyte growth factor
- HDAC
histone deacetylase
- iCCA
intrahepatic CCA
- IDH
isocitrate dehydrogenase
- IL
interleukin
- MAPK
mitogen-activated protein kinase
- Mcl-1
myeloid cell leukemia sequence 1
- mTOR
mammalian target of rapamycin
- O. viverrini
opisthorchis viverrini
- pCCA
perihilar cholangiocarcinoma
- PI3K
phosphatidylinositol 3-kinase
- PD-1
Programmed death 1
- PD-L1
Programmed death ligand-1
- PTEN
phosphatase and tensin homolog
- SMKI
small molecule kinase inhibitor
- SOCS-3
suppressor of cytokine signaling 3
- STAT
signal transducer and activator of transcription
- TP53
tumor protein 53
- TRAIL
tumor necrosis factor-related apoptosis-inducing ligand
Footnotes
Conflict of interest: The authors have no commercial relationships or interests to disclose relevant to this manuscript.
Authors Contributions: Dr. Sumera Rizvi contributed to the outline and drafting of the manuscript, critical revision of the manuscript, and important intellectual content; Dr. Gregory J. Gores contributed to the outline of the manuscript, critical revision of the manuscript for important intellectual content, and provided manuscript writing supervision.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Rizvi S, Gores GJ. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology. 2013;145:1215–1229. doi: 10.1053/j.gastro.2013.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Rizvi S, Borad MJ, Patel T, Gores GJ. Cholangiocarcinoma: molecular pathways and therapeutic opportunities. Seminars in liver disease. 2014;34:456–464. doi: 10.1055/s-0034-1394144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, et al. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA) Nat Rev Gastroenterol Hepatol. 2016;13:261–280. doi: 10.1038/nrgastro.2016.51. [DOI] [PubMed] [Google Scholar]
- 4.Cardinale V, Bragazzi MC, Carpino G, Torrice A, Fraveto A, Gentile R, Pasqualino V, et al. Cholangiocarcinoma: increasing burden of classifications. Hepatobiliary Surg Nutr. 2013;2:272–280. doi: 10.3978/j.issn.2304-3881.2013.10.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Komuta M, Govaere O, Vandecaveye V, Akiba J, Van Steenbergen W, Verslype C, Laleman W, et al. Histological diversity in cholangiocellular carcinoma reflects the different cholangiocyte phenotypes. Hepatology. 2012;55:1876–1888. doi: 10.1002/hep.25595. [DOI] [PubMed] [Google Scholar]
- 6.Everhart JE, Ruhl CE. Burden of digestive diseases in the United States Part III: Liver, biliary tract, and pancreas. Gastroenterology. 2009;136:1134–1144. doi: 10.1053/j.gastro.2009.02.038. [DOI] [PubMed] [Google Scholar]
- 7.DeOliveira ML, Cunningham SC, Cameron JL, Kamangar F, Winter JM, Lillemoe KD, Choti MA, et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Annals of surgery. 2007;245:755–762. doi: 10.1097/01.sla.0000251366.62632.d3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sapisochin G, Facciuto M, Rubbia-Brandt L, Marti J, Mehta N, Yao FY, Vibert E, et al. Liver transplantation for “very early” intrahepatic cholangiocarcinoma: International retrospective study supporting a prospective assessment. Hepatology. 2016;64:1178–1188. doi: 10.1002/hep.28744. [DOI] [PubMed] [Google Scholar]
- 9.Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, Madhusudan S, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. The New England journal of medicine. 2010;362:1273–1281. doi: 10.1056/NEJMoa0908721. [DOI] [PubMed] [Google Scholar]
- 10.Ong CK, Subimerb C, Pairojkul C, Wongkham S, Cutcutache I, Yu W, McPherson JR, et al. Exome sequencing of liver fluke-associated cholangiocarcinoma. Nature genetics. 2012;44:690–693. doi: 10.1038/ng.2273. [DOI] [PubMed] [Google Scholar]
- 11.Chan-On W, Nairismagi ML, Ong CK, Lim WK, Dima S, Pairojkul C, Lim KH, et al. Exome sequencing identifies distinct mutational patterns in liver fluke-related and non-infection-related bile duct cancers. Nature genetics. 2013;45:1474–1478. doi: 10.1038/ng.2806. [DOI] [PubMed] [Google Scholar]
- 12.Jiao Y, Pawlik TM, Anders RA, Selaru FM, Streppel MM, Lucas DJ, Niknafs N, et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nature genetics. 2013;45:1470–1473. doi: 10.1038/ng.2813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Turner N, Grose R. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer. 2010;10:116–129. doi: 10.1038/nrc2780. [DOI] [PubMed] [Google Scholar]
- 14.Borad MJ, Champion MD, Egan JB, Liang WS, Fonseca R, Bryce AH, McCullough AE, 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. doi: 10.1371/journal.pgen.1004135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, Murphy SJ, et al. Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Hum Pathol. 2014;45:1630–1638. doi: 10.1016/j.humpath.2014.03.014. [DOI] [PubMed] [Google Scholar]
- 16.Ross JS, Wang K, Gay L, Al-Rohil R, Rand JV, Jones DM, Lee HJ, et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. The oncologist. 2014;19:235–242. doi: 10.1634/theoncologist.2013-0352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wu YM, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, Lonigro RJ, et al. Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov. 2013;3:636–647. doi: 10.1158/2159-8290.CD-13-0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Arai Y, Totoki Y, Hosoda F, Shirota T, Hama N, Nakamura H, Ojima H, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology. 2014;59:1427–1434. doi: 10.1002/hep.26890. [DOI] [PubMed] [Google Scholar]
- 19.Sia D, Losic B, Moeini A, Cabellos L, Hao K, Revill K, Bonal D, et al. Massive parallel sequencing uncovers actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nat Commun. 2015;6:6087. doi: 10.1038/ncomms7087. [DOI] [PubMed] [Google Scholar]
- 20.Druker BJ. Translation of the Philadelphia chromosome into therapy for CML. Blood. 2008;112:4808–4817. doi: 10.1182/blood-2008-07-077958. [DOI] [PubMed] [Google Scholar]
- 21.Rizvi S, Yamada D, Hirsova P, Bronk SF, Werneburg NW, Krishnan A, Salim W, et al. A Hippo and Fibroblast Growth Factor Receptor Autocrine Pathway in Cholangiocarcinoma. J Biol Chem. 2016;291:8031–8047. doi: 10.1074/jbc.M115.698472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nogova L, Sequist LV, Perez Garcia JM, Andre F, Delord JP, Hidalgo M, Schellens JH, et al. Evaluation of BGJ398, a Fibroblast Growth Factor Receptor 1–3 Kinase Inhibitor, in Patients With Advanced Solid Tumors Harboring Genetic Alterations in Fibroblast Growth Factor Receptors: Results of a Global Phase I, Dose-Escalation and Dose-Expansion Study. J Clin Oncol. 2016 doi: 10.1200/JCO.2016.67.2048. JCO2016672048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Javle M. 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. J Clin Oncol. 2016;34(suppl 4S) abstr 335. [Google Scholar]
- 24.Tabernero J, Bahleda R, Dienstmann R, Infante JR, Mita A, Italiano A, Calvo E, et al. Phase I Dose-Escalation Study of JNJ-42756493, an Oral Pan-Fibroblast Growth Factor Receptor Inhibitor, in Patients With Advanced Solid Tumors. J Clin Oncol. 2015;33:3401–3408. doi: 10.1200/JCO.2014.60.7341. [DOI] [PubMed] [Google Scholar]
- 25.Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre JM, Schwartz B, et al. Preclinical Activity of ARQ 087, a Novel Inhibitor Targeting FGFR Dysregulation. PLoS One. 2016;11:e0162594. doi: 10.1371/journal.pone.0162594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bekaii-Saab T, Phelps MA, Li X, Saji M, Goff L, Kauh JS, O’Neil BH, et al. Multi-institutional phase II study of selumetinib in patients with metastatic biliary cancers. J Clin Oncol. 2011;29:2357–2363. doi: 10.1200/JCO.2010.33.9473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tan L, Wang J, Tanizaki J, Huang Z, Aref AR, Rusan M, Zhu SJ, et al. Development of covalent inhibitors that can overcome resistance to first-generation FGFR kinase inhibitors. Proc Natl Acad Sci U S A. 2014;111:E4869–4877. doi: 10.1073/pnas.1403438111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, Lennerz JK, et al. Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma. Cancer Discov. 2016 doi: 10.1158/2159-8290.CD-16-1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhao WM, Wang L, Park H, Chhim S, Tanphanich M, Yashiro M, Kim KJ. Monoclonal antibodies to fibroblast growth factor receptor 2 effectively inhibit growth of gastric tumor xenografts. Clin Cancer Res. 2010;16:5750–5758. doi: 10.1158/1078-0432.CCR-10-0531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Appleman LJ. MET signaling pathway: a rational target for cancer therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011;29:4837–4838. doi: 10.1200/JCO.2011.37.7929. [DOI] [PubMed] [Google Scholar]
- 31.Terada T, Nakanuma Y, Sirica AE. Immunohistochemical demonstration of MET overexpression in human intrahepatic cholangiocarcinoma and in hepatolithiasis. Human pathology. 1998;29:175–180. doi: 10.1016/s0046-8177(98)90229-5. [DOI] [PubMed] [Google Scholar]
- 32.Sia D, Hoshida Y, Villanueva A, Roayaie S, Ferrer J, Tabak B, Peix J, et al. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes. Gastroenterology. 2013;144:829–840. doi: 10.1053/j.gastro.2013.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, et al. Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PloS one. 2014;9:e115383. doi: 10.1371/journal.pone.0115383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gherardi E, Birchmeier W, Birchmeier C, Vande Woude G. Targeting MET in cancer: rationale and progress. Nature reviews Cancer. 2012;12:89–103. doi: 10.1038/nrc3205. [DOI] [PubMed] [Google Scholar]
- 35.Pant S, Saleh M, Bendell J, Infante JR, Jones S, Kurkjian CD, Moore KM, et al. A phase I dose escalation study of oral c-MET inhibitor tivantinib (ARQ 197) in combination with gemcitabine in patients with solid tumors. Annals of oncology : official journal of the European Society for Medical Oncology. 2014;25:1416–1421. doi: 10.1093/annonc/mdu157. [DOI] [PubMed] [Google Scholar]
- 36.Taniai M, Grambihler A, Higuchi H, Werneburg N, Bronk SF, Farrugia DJ, Kaufmann SH, et al. Mcl-1 mediates tumor necrosis factor-related apoptosis-inducing ligand resistance in human cholangiocarcinoma cells. Cancer research. 2004;64:3517–3524. doi: 10.1158/0008-5472.CAN-03-2770. [DOI] [PubMed] [Google Scholar]
- 37.Kobayashi S, Werneburg NW, Bronk SF, Kaufmann SH, Gores GJ. Interleukin-6 contributes to Mcl-1 up-regulation and TRAIL resistance via an Akt-signaling pathway in cholangiocarcinoma cells. Gastroenterology. 2005;128:2054–2065. doi: 10.1053/j.gastro.2005.03.010. [DOI] [PubMed] [Google Scholar]
- 38.Isomoto H, Kobayashi S, Werneburg NW, Bronk SF, Guicciardi ME, Frank DA, Gores GJ. Interleukin 6 upregulates myeloid cell leukemia-1 expression through a STAT3 pathway in cholangiocarcinoma cells. Hepatology. 2005;42:1329–1338. doi: 10.1002/hep.20966. [DOI] [PubMed] [Google Scholar]
- 39.Isomoto H, Mott JL, Kobayashi S, Werneburg NW, Bronk SF, Haan S, Gores GJ. Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing. Gastroenterology. 2007;132:384–396. doi: 10.1053/j.gastro.2006.10.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Yamada D, Rizvi S, Razumilava N, Bronk SF, Davila JI, Champion MD, Borad MJ, et al. IL-33 facilitates oncogene-induced cholangiocarcinoma in mice by an interleukin-6-sensitive mechanism. Hepatology. 2015;61:1627–1642. doi: 10.1002/hep.27687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. doi: 10.1038/nature08822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Barr Fritcher EG, Voss JS, Brankley SM, Campion MB, Jenkins SM, Keeney ME, Henry MR, et al. An Optimized Set of Fluorescence In Situ Hybridization Probes for Detection of Pancreatobiliary Tract Cancer in Cytology Brush Samples. Gastroenterology. 2015;149:1813–1824. e1811. doi: 10.1053/j.gastro.2015.08.046. [DOI] [PubMed] [Google Scholar]
- 43.Abulwerdi F, Liao C, Liu M, Azmi AS, Aboukameel A, Mady AS, Gulappa T, et al. A novel small-molecule inhibitor of mcl-1 blocks pancreatic cancer growth in vitro and in vivo. Molecular cancer therapeutics. 2014;13:565–575. doi: 10.1158/1535-7163.MCT-12-0767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Abulwerdi FA, Liao C, Mady AS, Gavin J, Shen C, Cierpicki T, Stuckey JA, et al. 3-Substituted-N-(4-hydroxynaphthalen-1-yl)arylsulfonamides as a novel class of selective Mcl-1 inhibitors: structure-based design, synthesis, SAR, and biological evaluation. Journal of medicinal chemistry. 2014;57:4111–4133. doi: 10.1021/jm500010b. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kotschy A, Szlavik Z, Murray J, Davidson J, Maragno AL, Le Toumelin-Braizat G, Chanrion M, et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature. 2016;538:477–482. doi: 10.1038/nature19830. [DOI] [PubMed] [Google Scholar]
- 46.Hedvat M, Huszar D, Herrmann A, Gozgit JM, Schroeder A, Sheehy A, Buettner R, et al. The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer cell. 2009;16:487–497. doi: 10.1016/j.ccr.2009.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Grassian AR, Pagliarini R, Chiang DY. Mutations of isocitrate dehydrogenase 1 and 2 in intrahepatic cholangiocarcinoma. Current opinion in gastroenterology. 2014;30:295–302. doi: 10.1097/MOG.0000000000000050. [DOI] [PubMed] [Google Scholar]
- 48.Kipp BR, Voss JS, Kerr SE, Barr Fritcher EG, Graham RP, Zhang L, Highsmith WE, et al. Isocitrate dehydrogenase 1 and 2 mutations in cholangiocarcinoma. Human pathology. 2012;43:1552–1558. doi: 10.1016/j.humpath.2011.12.007. [DOI] [PubMed] [Google Scholar]
- 49.Zhu AX, Borger DR, Kim Y, Cosgrove D, Ejaz A, Alexandrescu S, Groeschl RT, et al. Genomic profiling of intrahepatic cholangiocarcinoma: refining prognosis and identifying therapeutic targets. Annals of surgical oncology. 2014;21:3827–3834. doi: 10.1245/s10434-014-3828-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Borger DR, Tanabe KK, Fan KC, Lopez HU, Fantin VR, Straley KS, Schenkein DP, et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist. 2012;17:72–79. doi: 10.1634/theoncologist.2011-0386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang P, Dong Q, Zhang C, Kuan PF, Liu Y, Jeck WR, Andersen JB, et al. Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas. Oncogene. 2013;32:3091–3100. doi: 10.1038/onc.2012.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Goyal L, Govindan A, Sheth RA, Nardi V, Blaszkowsky LS, Faris JE, Clark JW, et al. Prognosis and Clinicopathologic Features of Patients With Advanced Stage Isocitrate Dehydrogenase (IDH) Mutant and IDH Wild-Type Intrahepatic Cholangiocarcinoma. Oncologist. 2015;20:1019–1027. doi: 10.1634/theoncologist.2015-0210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rohle D, Popovici-Muller J, Palaskas N, Turcan S, Grommes C, Campos C, Tsoi J, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science. 2013;340:626–630. doi: 10.1126/science.1236062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Wang F, Travins J, DeLaBarre B, Penard-Lacronique V, Schalm S, Hansen E, Straley K, et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 2013;340:622–626. doi: 10.1126/science.1234769. [DOI] [PubMed] [Google Scholar]
- 55.Saha SK, Gordan JD, Kleinstiver BP, Vu P, Najem MS, Yeo JC, Shi L, et al. Isocitrate Dehydrogenase Mutations Confer Dasatinib Hypersensitivity and SRC Dependence in Intrahepatic Cholangiocarcinoma. Cancer Discov. 2016;6:727–739. doi: 10.1158/2159-8290.CD-15-1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Julien SG, Dube N, Hardy S, Tremblay ML. Inside the human cancer tyrosine phosphatome. Nature reviews Cancer. 2011;11:35–49. doi: 10.1038/nrc2980. [DOI] [PubMed] [Google Scholar]
- 57.Gao Q, Zhao YJ, Wang XY, Guo WJ, Gao S, Wei L, Shi JY, et al. Activating mutations in PTPN3 promote cholangiocarcinoma cell proliferation and migration and are associated with tumor recurrence in patients. Gastroenterology. 2014;146:1397–1407. doi: 10.1053/j.gastro.2014.01.062. [DOI] [PubMed] [Google Scholar]
- 58.Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27. doi: 10.1016/j.cell.2012.06.013. [DOI] [PubMed] [Google Scholar]
- 59.Sirica AE. Role of ErbB family receptor tyrosine kinases in intrahepatic cholangiocarcinoma. World journal of gastroenterology. 2008;14:7033–7058. doi: 10.3748/wjg.14.7033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, Monsey J, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–237. doi: 10.1158/2159-8290.CD-12-0349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lubner SJ, Mahoney MR, Kolesar JL, Loconte NK, Kim GP, Pitot HC, Philip PA, 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;28:3491–3497. doi: 10.1200/JCO.2010.28.4075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Philip PA, Mahoney MR, Allmer C, Thomas J, Pitot HC, Kim G, Donehower RC, et al. Phase II study of erlotinib in patients with advanced biliary cancer. J Clin Oncol. 2006;24:3069–3074. doi: 10.1200/JCO.2005.05.3579. [DOI] [PubMed] [Google Scholar]
- 63.Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, Hama N, et al. Genomic spectra of biliary tract cancer. Nature genetics. 2015;47:1003–1010. doi: 10.1038/ng.3375. [DOI] [PubMed] [Google Scholar]
- 64.Honeyman JN, Simon EP, Robine N, Chiaroni-Clarke R, Darcy DG, Lim II, Gleason CE, et al. Detection of a recurrent DNAJB1-PRKACA chimeric transcript in fibrolamellar hepatocellular carcinoma. Science. 2014;343:1010–1014. doi: 10.1126/science.1249484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Loilome W, Juntana S, Namwat N, Bhudhisawasdi V, Puapairoj A, Sripa B, Miwa M, et al. PRKAR1A is overexpressed and represents a possible therapeutic target in human cholangiocarcinoma. International journal of cancer. 2011;129:34–44. doi: 10.1002/ijc.25646. [DOI] [PubMed] [Google Scholar]
- 66.Tan J, Tan P, Teh BT. Defining the Molecular Alterations of Ampullary Carcinoma. Cancer Cell. 2016;29:135–136. doi: 10.1016/j.ccell.2016.01.011. [DOI] [PubMed] [Google Scholar]
- 67.Oliver JR, Kushwah R, Hu J. Multiple roles of the epithelium-specific ETS transcription factor, ESE-1, in development and disease. Lab Invest. 2012;92:320–330. doi: 10.1038/labinvest.2011.186. [DOI] [PubMed] [Google Scholar]
- 68.Gingras MC, Covington KR, Chang DK, Donehower LA, Gill AJ, Ittmann MM, Creighton CJ, et al. Ampullary Cancers Harbor ELF3 Tumor Suppressor Gene Mutations and Exhibit Frequent WNT Dysregulation. Cell Rep. 2016;14:907–919. doi: 10.1016/j.celrep.2015.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Yachida S, Wood LD, Suzuki M, Takai E, Totoki Y, Kato M, Luchini C, et al. Genomic Sequencing Identifies ELF3 as a Driver of Ampullary Carcinoma. Cancer Cell. 2016;29:229–240. doi: 10.1016/j.ccell.2015.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Andersen JB, Spee B, Blechacz BR, Avital I, Komuta M, Barbour A, Conner EA, et al. Genomic and Genetic Characterization of Cholangiocarcinoma Identifies Therapeutic Targets for Tyrosine Kinase Inhibitors. Gastroenterology. 2012;142:1021–U1552. doi: 10.1053/j.gastro.2011.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bridgewater J, Lopes A, Beare S, Duggan M, Lee D, Ricamara M, McEntee D, 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. doi: 10.1186/s12885-016-2174-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Whittaker S, Marais R, Zhu AX. The role of signaling pathways in the development and treatment of hepatocellular carcinoma. Oncogene. 2010;29:4989–5005. doi: 10.1038/onc.2010.236. [DOI] [PubMed] [Google Scholar]
- 73.Yothaisong S, Dokduang H, Techasen A, Namwat N, Yongvanit P, Bhudhisawasdi V, Puapairoj A, et al. Increased activation of PI3K/AKT signaling pathway is associated with cholangiocarcinoma metastasis and PI3K/mTOR inhibition presents a possible therapeutic strategy. Tumour Biol. 2013;34:3637–3648. doi: 10.1007/s13277-013-0945-2. [DOI] [PubMed] [Google Scholar]
- 74.Leelawat K, Narong S, Udomchaiprasertkul W, Leelawat S, Tungpradubkul S. Inhibition of PI3K increases oxaliplatin sensitivity in cholangiocarcinoma cells. Cancer Cell Int. 2009;9:3. doi: 10.1186/1475-2867-9-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Morton SD, Cadamuro M, Brivio S, Vismara M, Stecca T, Massani M, Bassi N, et al. Leukemia inhibitory factor protects cholangiocarcinoma cells from drug-induced apoptosis via a PI3K/AKT-dependent Mcl-1 activation. Oncotarget. 2015;6:26052–26064. doi: 10.18632/oncotarget.4482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Voss JS, Holtegaard LM, Kerr SE, Fritcher EG, Roberts LR, Gores GJ, Zhang J, et al. Molecular profiling of cholangiocarcinoma shows potential for targeted therapy treatment decisions. Hum Pathol. 2013;44:1216–1222. doi: 10.1016/j.humpath.2012.11.006. [DOI] [PubMed] [Google Scholar]
- 77.Sheppard K, Kinross KM, Solomon B, Pearson RB, Phillips WA. Targeting PI3 kinase/AKT/mTOR signaling in cancer. Critical reviews in oncogenesis. 2012;17:69–95. doi: 10.1615/critrevoncog.v17.i1.60. [DOI] [PubMed] [Google Scholar]
- 78.Ewald F, Grabinski N, Grottke A, Windhorst S, Norz D, Carstensen L, Staufer K, et al. Combined targeting of AKT and mTOR using MK-2206 and RAD001 is synergistic in the treatment of cholangiocarcinoma. International journal of cancer. 2013;133:2065–2076. doi: 10.1002/ijc.28214. [DOI] [PubMed] [Google Scholar]
- 79.Ewald F, Norz D, Grottke A, Hofmann BT, Nashan B, Jucker M. Dual Inhibition of PI3K-AKT-mTOR- and RAF-MEK-ERK-signaling is synergistic in cholangiocarcinoma and reverses acquired resistance to MEK-inhibitors. Investigational new drugs. 2014;32:1144–1154. doi: 10.1007/s10637-014-0149-7. [DOI] [PubMed] [Google Scholar]
- 80.McRee AJ, Sanoff HK, Carlson C, Ivanova A, O’Neil BH. A phase I trial of mFOLFOX6 combined with the oral PI3K inhibitor BKM120 in patients with advanced refractory solid tumors. Investigational new drugs. 2015;33:1225–1231. doi: 10.1007/s10637-015-0298-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Costello BA, Borad MJ, Qi Y, Kim GP, Northfelt DW, Erlichman C, Alberts SR. Phase I trial of everolimus, gemcitabine and cisplatin in patients with solid tumors. Investigational new drugs. 2014;32:710–716. doi: 10.1007/s10637-014-0096-3. [DOI] [PubMed] [Google Scholar]
- 82.Boye K, Maelandsmo GM. S100A4 and metastasis: a small actor playing many roles. Am J Pathol. 2010;176:528–535. doi: 10.2353/ajpath.2010.090526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fabris L, Cadamuro M, Moserle L, Dziura J, Cong X, Sambado L, Nardo G, et al. Nuclear expression of S100A4 calcium-binding protein increases cholangiocarcinoma invasiveness and metastasization. Hepatology. 2011;54:890–899. doi: 10.1002/hep.24466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Gongoll S, Peters G, Mengel M, Piso P, Klempnauer J, Kreipe H, von Wasielewski R. Prognostic significance of calcium-binding protein S100A4 in colorectal cancer. Gastroenterology. 2002;123:1478–1484. doi: 10.1053/gast.2002.36606. [DOI] [PubMed] [Google Scholar]
- 85.Cadamuro M, Spagnuolo G, Sambado L, Indraccolo S, Nardo G, Rosato A, Brivio S, et al. Low-Dose Paclitaxel Reduces S100A4 Nuclear Import to Inhibit Invasion and Hematogenous Metastasis of Cholangiocarcinoma. Cancer Res. 2016;76:4775–4784. doi: 10.1158/0008-5472.CAN-16-0188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Fingas CD, Bronk SF, Werneburg NW, Mott JL, Guicciardi ME, Cazanave SC, Mertens JC, et al. Myofibroblast-derived PDGF-BB promotes Hedgehog survival signaling in cholangiocarcinoma cells. Hepatology. 2011;54:2076–2088. doi: 10.1002/hep.24588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Jinawath A, Akiyama Y, Sripa B, Yuasa Y. Dual blockade of the Hedgehog and ERK1/2 pathways coordinately decreases proliferation and survival of cholangiocarcinoma cells. J Cancer Res Clin Oncol. 2007;133:271–278. doi: 10.1007/s00432-006-0166-9. [DOI] [PubMed] [Google Scholar]
- 88.El Khatib M, Kalnytska A, Palagani V, Kossatz U, Manns MP, Malek NP, Wilkens L, et al. Inhibition of hedgehog signaling attenuates carcinogenesis in vitro and increases necrosis of cholangiocellular carcinoma. Hepatology. 2013;57:1035–1045. doi: 10.1002/hep.26147. [DOI] [PubMed] [Google Scholar]
- 89.Fingas CD, Mertens JC, Razumilava N, Sydor S, Bronk SF, Christensen JD, Rizvi SH, et al. Polo-like kinase 2 is a mediator of hedgehog survival signaling in cholangiocarcinoma. Hepatology. 2013;58:1362–1374. doi: 10.1002/hep.26484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Razumilava N, Gradilone SA, Smoot RL, Mertens JC, Bronk SF, Sirica AE, Gores GJ. Non-canonical Hedgehog signaling contributes to chemotaxis in cholangiocarcinoma. J Hepatol. 2014;60:599–605. doi: 10.1016/j.jhep.2013.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Riedlinger D, Bahra M, Boas-Knoop S, Lippert S, Bradtmoller M, Guse K, Seehofer D, et al. Hedgehog pathway as a potential treatment target in human cholangiocarcinoma. J Hepatobiliary Pancreat Sci. 2014;21:607–615. doi: 10.1002/jhbp.107. [DOI] [PubMed] [Google Scholar]
- 92.Argani P, Iacobuzio-Donahue C, Ryu B, Rosty C, Goggins M, Wilentz RE, Murugesan SR, et al. Mesothelin is overexpressed in the vast majority of ductal adenocarcinomas of the pancreas: identification of a new pancreatic cancer marker by serial analysis of gene expression (SAGE) Clin Cancer Res. 2001;7:3862–3868. [PubMed] [Google Scholar]
- 93.Yu L, Feng M, Kim H, Phung Y, Kleiner DE, Gores GJ, Qian M, et al. Mesothelin as a potential therapeutic target in human cholangiocarcinoma. J Cancer. 2010;1:141–149. doi: 10.7150/jca.1.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kawamata F, Kamachi H, Einama T, Homma S, Tahara M, Miyazaki M, Tanaka S, et al. Intracellular localization of mesothelin predicts patient prognosis of extrahepatic bile duct cancer. Int J Oncol. 2012;41:2109–2118. doi: 10.3892/ijo.2012.1662. [DOI] [PubMed] [Google Scholar]
- 95.Nomura R, Fujii H, Abe M, Sugo H, Ishizaki Y, Kawasaki S, Hino O. Mesothelin expression is a prognostic factor in cholangiocellular carcinoma. Int Surg. 2013;98:164–169. doi: 10.9738/INTSURG-D-13-00001.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Tang Z, Qian M, Ho M. The role of mesothelin in tumor progression and targeted therapy. Anticancer Agents Med Chem. 2013;13:276–280. doi: 10.2174/1871520611313020014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hassan R, Kindler HL, Jahan T, Bazhenova L, Reck M, Thomas A, Pastan I, et al. Phase II clinical trial of amatuximab, a chimeric antimesothelin antibody with pemetrexed and cisplatin in advanced unresectable pleural mesothelioma. Clin Cancer Res. 2014;20:5927–5936. doi: 10.1158/1078-0432.CCR-14-0804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Fujisaka Y, Kurata T, Tanaka K, Kudo T, Okamoto K, Tsurutani J, Kaneda H, et al. Phase I study of amatuximab, a novel monoclonal antibody to mesothelin, in Japanese patients with advanced solid tumors. Invest New Drugs. 2015;33:380–388. doi: 10.1007/s10637-014-0196-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hassan R, Bullock S, Premkumar A, Kreitman RJ, Kindler H, Willingham MC, Pastan I. Phase I study of SS1P, a recombinant anti-mesothelin immunotoxin given as a bolus I.V. infusion to patients with mesothelin-expressing mesothelioma, ovarian, and pancreatic cancers. Clin Cancer Res. 2007;13:5144–5149. doi: 10.1158/1078-0432.CCR-07-0869. [DOI] [PubMed] [Google Scholar]
- 100.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature reviews Cancer. 2012;12:252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Boussiotis VA. Molecular and Biochemical Aspects of the PD-1 Checkpoint Pathway. The New England journal of medicine. 2016;375:1767–1778. doi: 10.1056/NEJMra1514296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, et al. Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine. 2010;363:711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lipson EJ, Drake CG. Ipilimumab: an anti-CTLA-4 antibody for metastatic melanoma. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17:6958–6962. doi: 10.1158/1078-0432.CCR-11-1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. The New England journal of medicine. 2012;366:2443–2454. doi: 10.1056/NEJMoa1200690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Ye Y, Zhou L, Xie X, Jiang G, Xie H, Zheng S. Interaction of B7-H1 on intrahepatic cholangiocarcinoma cells with PD-1 on tumor-infiltrating T cells as a mechanism of immune evasion. J Surg Oncol. 2009;100:500–504. doi: 10.1002/jso.21376. [DOI] [PubMed] [Google Scholar]
- 106.Sabbatino F, Villani V, Yearley JH, Deshpande V, Cai L, Konstantinidis IT, Moon C, et al. PD-L1 and HLA Class I Antigen Expression and Clinical Course of the Disease in Intrahepatic Cholangiocarcinoma. Clin Cancer Res. 2016;22:470–478. doi: 10.1158/1078-0432.CCR-15-0715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.YJB Safety and efficacy of pembrolizumab (MK-3475) in patients (pts) with advanced biliary tract cancer: interim results of KEYNOTE-028. European Cancer Congress 2015 (ECCO and ESMO), Poster Spotlight Session: Immunotherapy in Cancer; 2015 September 26, 2015; Vienna, Austria. 2015. [Google Scholar]
- 108.Abou-Alfa GK, Andersen JB, Chapman W, Choti M, Forbes SJ, Gores GJ, Hong TS, et al. Advances in cholangiocarcinoma research: report from the third Cholangiocarcinoma Foundation Annual Conference. J Gastrointest Oncol. 2016;7:819–827. doi: 10.21037/jgo.2016.11.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Chaiteerakij R, Harmsen WS, Marrero CR, Aboelsoud MM, Ndzengue A, Kaiya J, Therneau TM, et al. A new clinically based staging system for perihilar cholangiocarcinoma. Am J Gastroenterol. 2014;109:1881–1890. doi: 10.1038/ajg.2014.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Affo S, Yu LX, Schwabe RF. The Role of Cancer-Associated Fibroblasts and Fibrosisin Liver Cancer. Annu Rev Pathol. 2017;12:153–186. doi: 10.1146/annurev-pathol-052016-100322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Duffy AG, Makarova-Rusher OV, Greten TF. The case for immune-based approaches in biliary tract carcinoma. Hepatology. 2016;64:1785–1791. doi: 10.1002/hep.28635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Rizvi S, Mertens JC, Bronk SF, Hirsova P, Dai H, Roberts LR, Kaufmann SH, et al. Platelet-derived growth factor primes cancer-associated fibroblasts for apoptosis. J Biol Chem. 2014;289:22835–22849. doi: 10.1074/jbc.M114.563064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Galluzzi L, Buque A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. 2017;17:97–111. doi: 10.1038/nri.2016.107. [DOI] [PubMed] [Google Scholar]