Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Gastrointestinal Cancer

Molecular Targets in Biliary Carcinogenesis and Implications for Therapy TOLUTOPE OYASIJI,a JIANLIANG ZHANG,a BORIS KUVSHINOFF,a RENUKA IYER,b STEVEN N. HOCHWALDa Departments of aSurgical Oncology and bMedicine, Roswell Park Cancer Institute, Buffalo, New York, USA Disclosures of potential conflicts of interest may be found at the end of this article.

Key Words. Biliary x Carcinogenesis x Molecular x Targets x Therapy

Biliary tract cancers (BTCs) encompass a group of invasive carcinomas, including cholangiocarcinoma (intrahepatic, perihilar, or extrahepatic), and gallbladder carcinoma. Approximately 90% of patients present with advanced, unresectable disease and have a poor prognosis. The latest recommendation is to treat advanced or metastatic disease with gemcitabine and cisplatin, although chemotherapy has recorded modest survival benefits.

Comprehension of the molecular basis of biliary carcinogenesis has resulted in experimental trials of targeted therapies in BTCs, with promising results. This review addresses the emerging role of targeted therapy in the treatment of BTCs. Findings from preclinical studies were reviewed and correlated with the outcomes of clinical trials that were undertaken to translate the laboratory discoveries. The Oncologist 2015;20:1–10

Implications for Practice: Biliary tract cancers are rare. Approximately 90% of patients present with advanced, unresectable disease and have a poor prognosis. Median overall and progression-free survival are 12 and 8 months, respectively. Because chemotherapy has recorded modest survival benefits, targeted therapies are being explored for personalized treatment of these cancers. A comprehensive review of targeted therapies in biliary tract cancers was undertaken to present emerging evidence from laboratory and/or molecular studies as they translate to clinical trials and outcomes.The latest evidence on this topic is presented to clinicians and practitioners to guide decisions on treatment of this disease.

INTRODUCTION Biliary tract cancers (BTCs) encompass a group of invasive carcinomas, including cholangiocarcinoma (CC), which refers to cancers arising in the intrahepatic, perihilar, or extrahepatic biliary tree; and gallbladder carcinoma (GBC) [1]. They originate from the epithelium of the gallbladder and the bile ducts [2]. More than 90% of BTCs are well-differentiated, mucin-producing adenocarcinomas, while squamous cell carcinoma and small-cell carcinoma occur less frequently [3]. The rate of incidence for BTCs in the Western world is 1–2 cases per 100,000. Conversely, these neoplasms occur more frequently in Asia and South America, with an incidence rate of 96 cases per 100,000 [4]. BTCs affect up to 12,000 people annually in the U.S., with a documented increase in the incidence of intrahepatic cholangiocarcinoma [5]. CC occurs more frequently in the seventh decade of life, with greater predilection for men. Women are more commonly affected by GBC; the median age at onset is 65 years.The difference in genderdistribution correlates with the pattern of distribution of predisposing factors; for example, cholelithiasis is more common in women [3]. BTCs frequently stem from conditions that cause chronic inflammation, injury, and reparative biliary epithelial cell

proliferation,suchasprimarysclerosingcholangitis,clonorchiasis, hepatolithiasis, or complicated fibropolycystic diseases. A close interplay among chronic inflammation, cellular injury within bile ducts, and partial biliary tract obstruction appears to be fundamental in the pathogenesis of cholangiocarcinoma [6–8]. Regional nodal and distant metastases tend to occur early in BTCs. Only 10% of patients present with early-stage, surgically resectable, and curable disease. The prognosis is poor for most patients with locally advanced or metastatic BTCs, with a median survival of ,1 year [1]. A common denominator for this group of diseases is their anatomic origin in the biliary tract. Significant differences, however, are noticeable in disease course, molecular profiling, and response to treatment. GBC is known to be associated with worse survival compared with CC. Ironically, it exhibits greater response rates to chemotherapy. GBC tends to develop distant metastasis after surgical treatment, whereas CC is more likely to recur locally [9]. Before results of the ABC02 randomized, controlled, phase III clinical trial were reported, scientific evidence for chemotherapy in advanced biliary tract cancers was from nonrandomized phase

Correspondence: Steven N. Hochwald, M.D., Department of Surgical Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, New York 14263, USA. Telephone: 716-845-3493; E-Mail: [email protected] Received November 10, 2014; accepted for publication March 27, 2015. ©AlphaMed Press 1083-7159/2015/$20.00/0 http://dx.doi.org/10.1634/theoncologist.2014-0442

The Oncologist 2015;20:1–10 www.TheOncologist.com

©AlphaMed Press 2015

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

ABSTRACT

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Molecular Targets in Biliary Carcinogenesis

2

The infrequent incidence of BTCs and lack of randomized trials do not allow for definite conclusions regarding the role of radiation therapy. Molecular targetbased cancer therapy holds the prospect of improved therapeutic efficacy, particularly in cases that are refractory to conventional chemotherapy.

GENETIC AND EPIGENETIC CHANGES IN BILIARY CARCINOGENESIS Complex changes in cholangiocyte genes that characterize transformation and progression to cholangiocarcinoma have been identified at the molecular level. Because these changes do not disrupt the DNA sequence of affected genes, they are not mutations. Rather, they oversee the mechanism of specific gene expression [13–16]. As such,theyarebetterdescribed as genetic/ epigenetic changes. For the most part, these changes lead to silencing of genes involved in tumor suppression, cell cycle entry/ progression, apoptosis, and cell death [13, 15, 16]. Epigenetic changes have also been shown to impact regulation of genes involved in inflammation, DNA repair, cell adhesion, and invasion. Two key processes of genetic/epigenetic changes in biliary carcinogenesis areDNAmethylation andregulationviamicroRNA.

DNA Methylation The presence of abnormal DNA methylomes is a well-recognized event in carcinogenesis [17]. Sriraksa et al. were able to demonstrate that DNA methylation was a prevalent molecular abnormality in cholangiocarcinoma. They showed that the pattern of DNA methylation in primary CC cells was significantly different from those of surrounding normal tissues [18]. While hypermethylation was documented in the homeobox genes HOXA9 and HOXD9 (PCR2 targets) for CC cells, hypomethylation was observed at these sites in surrounding normal tissues. Thus, HOXA9 and HOXD9 hypermethylation has potential for use as a biomarker for early diagnosis of CC. A related observation is the overexpression of EZH2 (the catalytic subunit of PCR2 genes) in CC. This was reported by Sasaki et al. [19]. Hypermethylation

of the promoter sequence of p16INK4a results in its inactivation, which allows CDK4 to bind to cyclin D1 and subsequent unbridled entry into the S phase of the cell cycle [20]. Sasaki and coworkers also demonstrated the correlation between EZH2 levels and progression through the spectrum of low-grade dysplasia to invasive disease in cholangiocarcinogenesis [19]. Other cell cycle inhibitors implicated in cholangiocarcinogenesis that are suppressed via hypermethylation include p14ARF [21], p16INK4b [22],and14-3-3s [23].Tumorsuppressorgenesthathave been shown to be inactivated via hypermethylation in cholangiocarcinomas include Semaphorin3B [24], RassF1A and p73 [22]. Studies havealso shown an arrayof DNA repairgenesthatare suppressed in cholangiocarcinoma, through hypermethylation. These include hMLH1, O6-methylguanine-DNA methyltransferase, and glutathione S-transferase P1 (GSTP1) genes [22, 23]. Epigenetic silencing of SOCS-3 has also been documented in cholangiocarcinoma [25]. The result is overexpression of interleukin-6 (IL-6) [26–28], which translates to modulation of growth regulatory pathways like epidermal growth factor receptor (EGFR) in favor of tumor progression [29]. E-cadherin inhibits tumor cell invasion and metastasis [30–32]. Almost half of CC samples studied show methylation of the E-cadherin promoter, which indicates epigenetic suppression of the gene and consequent progression of the disease [22–24, 33].

MicroRNA MicroRNAs are noncoding RNAs comprising approximately 22 nucleotides. They have regulatory roles in cellular processes like proliferation, death, apoptosis, fat metabolism, and differentiation [34–37]. Upregulation of miR-141, miR-200b, and miR-21 have been documented in CC. While miR-141 targets the CLOCK gene, which can act as a tumor suppressor, miR-200b dysregulates PTPN 12, thereby enhancing tumor cell survival and oncogenesis. miR-21 epigenetically silences PTEN, a tumor suppressor gene [38]. On the other hand, miR-29b and miR-370 were shown to be downregulated in CC. miR-29b downregulation may be related to overexpression of Mcl-1, an antiapoptotic protein, resulting in increased survival of CC cells [39, 40]. Upregulation of IL-6 in CC cells is associated with decreased expression of let-7a microRNA.This leads to downregulation of the NF2 gene, which is a known negative regulator of Stat-3. Activation of Stat-3 is reputed to play a role in many cancers [41]. Figure 1 presents a flowchart of DNA methylation and microRNA regulation of gene expression in BTC. Currently, there are no known DNA repair or cell adhesion genes involved in biliary carcinogenesis that are regulated by microRNAs.

ALTERED GENE EXPRESSION IN BTC Aberrant levels of many transcripts and proteins in BTC have been observed using proteomic analysis, gene profiling, and immunodetection. The correlations between altered protein levels and BTC can be used as biomarkers to detect transformation, progress, and metastasis. The altered effectors are members of several signal transduction pathways that modulate inflammation, proliferation, anchorage dependency, and metabolism. Brief descriptions of some BTC-related proteins are shown in Table 1.

OTncologist he

©AlphaMed Press 2015

®

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

II trials, which were underpowered. The progression-free survival advantage demonstrated in the ABC01 trial triggered the ABC02 trial, which showed significant survival advantage with a combination of gemcitabine and cisplatin versus gemcitabine alone (median overall survival [OS] of 11.7 vs 8.1 months and progression-free survival [PFS] of 8.0 vs 5.0 months) [10]. A similar randomized study in the Japanese population posted a similar median OS advantage for the gemcitabine/cisplatin combination compared with gemcitabine alone (11.2 vs 7.7 months) [11]. While the ABC02 trial reported a tumor control rate of 81.4% for the combination regimen, the Japanese study documented a rate of 68.3% [10, 11].These two landmark trials established the gemcitabine/cisplatin combination as the standard of care for advanced biliary tract cancers. The infrequent incidence of BTCs and lack of randomized trials do not allow for definite conclusions regarding the role of radiation therapy. Molecular target-based cancer therapy holds the prospect of improved therapeutic efficacy, particularly in cases that are refractory to conventional chemotherapy [12].

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Oyasiji, Zhang, Kuvshinoff et al.

3

Table 1. Altered gene expression in biliary tract cancer Name

Up/Down

Impacted pathway

Study

Actinin-1/4 Protein DJ-1 Cathepsin B a-Enolase IL-6 IDH1/2

Up Up Up Up

Cytoskeleton Glycolysis EGFR TCA cycle

EphB2 FBXW7 Glut-1/2 N/P-cadherin

Up Down Up Up

FAK F-box protein family Glucose transport Integrin-FAK

Kawase et al. [42] Yonglitthipagon et al. [43] Wehbe et al. [29] Borger et al. [44] Kipp et al. [45] Khansaard et al. [46] Enkhbold et al. [47] Borger et al. [44] Yi et al. [48]

Abbreviation: TCA, tricarboxylic acid.

Inflammation and Abnormal Growth Persistent inflammation of the biliary tract is associated with cholangiocarcinoma. For example, IL-6 overexpression decreases the methylation of the EGFR promoter, leading to increased levels of EGFR mRNA and protein [29]. This suggests that IL-6-induced epigenetic regulation of EGFR can contribute to uncontrolled BTC growth.

Attachment and Survival Transformation of the biliary tract epithelium leads to anchorageindependent growth. Decreased E-cadherin and increased N-cadherin levels are associated with epithelial to mesenchymal transition in prostate cancer [49]. Immunohistochemical analysis of tissues derived from 126 patients with gallbladder cancer

www.TheOncologist.com

has revealed a correlation between positive N- and P-cadherin expression with poor prognosis [48]. This indicates the involvement of cell adhesion modulation, including integrin-FAK signaling, in tumorigenesis of BTC.

Immunohistochemical analysis of tissues derived from 126 patients with gallbladder cancer has revealed a correlation between positive N- and P-cadherin expression with poor prognosis.This indicatesthe involvement of cell adhesion modulation, including integrin-FAK signaling, in tumorigenesis of BTC.

©AlphaMed Press 2015

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

Figure 1. DNA methylation and microRNA regulation of gene expression in biliary tract carcinoma. Abnormal activation of major signal transduction pathways and molecular targeted inhibitors under clinic trials are summarized. MicroRNAs (miR-141, miR-200b, miR-21, miR-29b, and miR-370) regulate PI3K-associated effectors (Clock, PTEN, and Mcl-1), activating Akt/mTOR-mediated apoptosis resistance, survival, and angiogenesis [37–39]. Alterations of DNA methylation in the promoter regions of p16INKa/b, SOCS-3, Semaphorin-3B, and RassF1A, and changes of Let-7a miR levels upregulate CDK4/cyclin D, EGFR/HER2, and RAS/RAF, leading to MEK1/2 and ERK1/2-triggered cell cycle progression and tumor growth [19–28]. DNA methylation and modification of the E-cadherin HOXA9/D9 promoters and FAK hyperactivation contribute to aberranttransformation and differentiation[17, 18, 29–31]. Small molecular inhibitors andmonoclonalantibodies have been developedto target the abnormal activity of the receptor or downstream kinases, preventing tumor proliferation and growth.

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Molecular Targets in Biliary Carcinogenesis

4

Oncogenic Metabolism Comparison studies between normal and BTC tissues or cells, using different approaches, have revealed increased levels of several proteins important in glycolysis and the tricarboxylic acid cycle. The levels of Glut-1, Glut-2, enolase, IDH-1, and IDH-2 are correlated with BTC [43, 44, 50]. This suggests glucose oncometabolism in BTC and potential sites for targeted therapy.

SIGNAL TRANSDUCTION PATHWAYS IN BTC

HER2/EGFR Signaling The erbB2 (HER2) gene was constitutively expressed in the epidermis of transgenic mice to elucidate the role of HER/EGFR signaling in malignancy of the gallbladder epithelium [55]. All transgenic mice developed gallbladder adenocarcinoma and cholangiocarcinoma in the biliary tree by 3 months of age [55]. Hyperphosphorylation of HER2 and EGFR indicates their heterodimerization and activation in erbB2 transgenic mice. HER2 pathways can trigger activation of the mitogen-activated protein kinase (MAPK) ERK1/2, PI3K/Akt/mTOR, and STAT. Indeed, erbB2 overexpression leads to increased phosphorylation of MAPK [55].

K-ras/RAF Signaling Tissue-specific activation of K-ras by a gain-of-function mutation G12D led to the development of intrahepatic cholangiocarcinoma in mice [50]. Combination of K-ras activation and p53 deletion shortens the mean survival time and causes widespread metastasis [51]. As an on-off switch, K-ras recruits and activates RAF and PI3K in cellular responses to growth factors such as VEGF and IGF-1. This is in agreement with the observations that mutations of K-ras, p53, and PIK3CA are common in specimens derived from patients with cholangiocarcinoma [55].

PATTERN OF MOLECULAR DRIVERS OF BTCS Mutations in the TP53 gene are well documented for gallbladder cancer. Incidence rates for this mutationrangefrom 44% to47% in human gallbladder cancer series [57–59]. The incidence rate for this mutation in intrahepatic cholangiocarcinoma varies between 8.6% and 36% [59, 60], while for extrahepatic cholangiocarcinoma, it is as high as 17.5% [59]. KRAS was significantly mutated in a series of 51 gallbladder cancer cells matched with normal tissue. The mutation rate was 7.8% [58].Two series that examined all BTCs showed much higher mutation rates for this gene in extrahepatic cholangiocarcinoma (23%–47%) compared with intrahepatic cholangiocarcinoma (5%–16%) and gallbladder cancer (4%–19%) [59].

MOLECULAR TARGETS EGFR EGFR belongs to the Erb B family ofclass I tyrosine kinases. It plays a key role in the proliferation of cancers and is overexpressed in several solid tumors. Receptor-specific ligands that belong to the EGF family of growth factors bind onto EGFR (Erb1), Erb3, and Erb4. The EGFR inhibitor erlotinib was studied as monotherapy in a single-arm, phase II trial for patients with advanced or metastatic BTCs [64]. The overall response rate was 7%, and 81% of the assessable tumors demonstrated EGFR expression (Table 3). However, EGFR mutational status was not assessed. Subsequently, a randomized phase III trial evaluated the combination of gemcitabine and oxaliplatin with or without continuous dosing of erlotinib for unresectable BTCs [65]. While the overall response rate was significantly higher in the chemotherapy plus erlotinib group (30% vs 16%; p 5 .005), PFS and OS did not differ (Table 3). Due to the mechanism of erlotinib and potential cell cycle sequence-specific synergy of erlotinib with gemcitabine, a phase Ib study evaluated the combination of gemcitabine and oxaliplatin with intermittent dosing of erlotinib for advanced BTCs [66]. Preliminary results demonstrated a 24% overall response rate and 6-month PFS rate of 75% (Table 3). This study highlighted the potential importance of mechanistic-driven dosing of targeted therapies when combined with cytotoxic chemotherapies. Monoclonal antibodies targeting EGFR showed promising results in BTCs, particularly in combination with traditional cytotoxic drugs. Two phase II trials evaluated the efficacy of cetuximab with gemcitabine and oxaliplatin. Gruenberger et al. reported an objective response rate of 63% in a trial of 30 patients with BTC; 30% of patients underwent potentially curative

OTncologist he

©AlphaMed Press 2015

®

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

Gene overexpression and deletion have been reported to define causal effectors that contribute to the development of BTC. Epigenetic, mutational, and transcriptional modulation promotes activation of several signaling pathways through effectors such as EGFR, human epidermal growth factor receptor 2 (HER2),VEGFR, IGF-1R, mitogen-activated protein kinase kinase (MEK), extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K), Akt, and mammalian target of rapamycin (mTOR). Defining the cause-and-effect relationship of these signaling molecules and BTC can establish the molecular foundation of targeted therapy. Gene manipulation studies suggest several key players that contribute to transformation and/or progression of BTC; these are summarized in Table 2.

Somatic mutations in IDH1 and IDH2 were documented for intrahepatic cholangiocarcinoma in different series of BTCs. Mutation rates are reported to be between 20% and 36%. More mutations were recorded in IDH1, with rates between 16% and 35%, compared with 3%–4% for IDH2 [57, 59, 60]. Several studies have reported on the role of FGFR fusions in intrahepatic cholangiocarcinoma. Among the fusions described are FGFR2-AHCYL1, FGFR2-BICC1, FGFR2-MGEA5, and FGFR2TACC3. These fusions occur exclusively in intrahepatic cholangiocarcinoma at a rate of 13.6%. FGFR2 fusions result in cellular morphologic change that causes abnormal cell proliferation. In vitro and in vivo evaluations have also shown sensitivity of cholangiocarcinomacells withFGFR2mutationstoFGFRinhibitors [61–63]. BAP1, ARID1A, and PBRM1 are chromatin-remodeling genes that have demonstrated mutations in BTCs. Mutation involving ARID1A have been identified in intrahepatic cholangiocarcinoma (11%–36%), extrahepatic cholangiocarcinoma (12%), and gallbladder cancer (6%–11.5%). Studies have demonstrated BAP1 mutation rates for gallbladder cancer (4%–13%) and intrahepatic cholangiocarcinoma (14%–20%). PBRM1 mutations were also identified in gallbladder cancer (7.7%–25%), extrahepatic cholangiocarcinoma (3.5%), and intrahepatic cholangiocarcinoma (13%–14.3%). The implication for practice is that BTCs that have these mutations may be sensitive to therapeutic agents targeting chromatin-remodeling genes [57, 59, 60].

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Oyasiji, Zhang, Kuvshinoff et al.

5

Table 2. Major signal transduction pathways associated with BTC tumorigenesis Name

Manipulation

Impacted pathway

Relationship

Study

K-ras (G12D)

Activation

MEK/ERK

Causes BTC

p53

Deletion

Suppressor

Enhances BTC

PIK3CA ErbB-2/ EGFR Notch

Mutation Transgenic Mice

Akt/mTOR MEK Notch

Associated with BTC Causes BTC Causes BTC

O’Dell et al. [51] Chen et al. [52] Borger et al. [44] Isa et al. [53] O’Dell et al. [51] Liu et al. [54] Borger et al. [44] Kiguchi et al. [55] Sekiya et al. [56]

Abbreviation: BTC, biliary tract cancer.

Type of cancers studied

Author, year

Regimen

Phase

Patients. no.

Philip et al., 2006 [64]

Erlotinib

II

42

Lee et al., 2012 [65]

GEMOX vs. GEMOX 1 erlotinib

III

268

Ciombor et al., 2012 [66]

GEMOX 1 erlotinib

Ib

17

Pancreas: 8, ABTC: 9

Gruenberger et al., 2010 [67]

GEMOX 1 cetuximab

II

30

GBC: 3; IHCC: 18; EHCC: 9

Malka et al., 2012 [68]

GEMOX 1 cetuximab vs. GEMOX GEMOX 1 panitumumab

II

150

ABC

II

46

IHCC: 10; EHCC: 21; Perihilar: 7; unclassified: 8

Jensen et al., 2011 [69]

GBC: 16; IHCC: 16; EHCC: 9; unclassified: 2 CC: 180; GBC: 82; ampulla of Vater: 6

Results RR: 7% PFS (6 months): 17% Median PFS: 4.2 months vs. 5.8 months Median OS: 9.5 months vs. 9.5 months RR: 16% vs. 30% (p 5 .05) RR: 24% PFS at 6 months: 75% RR: 63%; CR: 10%; PR: 53% Median PFS: 8.3 months; median OS: 12.7 months PFS at 4 months: 61% vs. 44% PFS (6 months): 71.6%; RR: 33% Median PFS: 8.3 months; median OS: 9.8 months

Abbreviations: ABC, advanced biliary cancer; ABTC, advanced biliary tract cancer; CR, complete response; EHCC, extrahepatic cholangiocarcinoma; GBC, gall bladder cancer; GEMOX, gemcitabine 1 oxaliplatin; IHCC, intrahepatic cholangiocarcinoma; OS, overall survival; PFS, progression-free survival; PR, partial response; RR, response rate.

resection after treatment (Table 3) [67]. Final analysis of the randomized phase II BINGO (Gemcitabine and Oxaliplatin With or Without Cetuximab in Advanced Biliary Tract Cancer) trial showed that the primary endpoint of 4-month PFS $60% was exceeded in the gemcitabine/oxaliplatin plus cetuximab arm, but median PFS and OS were similar in both arms (Table 1) [68]. Enrollment was not limited according to KRAS status in either of these trials, and given the proven importance of this biomarker in colorectal cancer, perhaps the efficacy of anti-EGFR antibodies in BTCs could be further improved by biomarker-driven patient selection. In contrast to the cetuximab trials, a phase II trial evaluating gemcitabine, oxaliplatin, capecitabine, and panitumumab enrolled patients with KRAS wild-type cholangiocarcinoma only, with a 71.6% 6-month PFS, response rate of 33%, and median OS of 9.8 months (Table 3) [69]. A phase III randomized trial compared the combination of gemcitabine and oxaliplatin (GEMOX) alone (arm A) with

www.TheOncologist.com

GEMOX plus erlotinib (arm B) in 268 Korean patients with BTCs (ampullary carcinomas were included). There was no difference in OS and PFS for both arms. Subgroup analysis, however, showed a PFS advantage for arm B in CC patients (5.9 vs. 3.0 months; p 5 .049) [70].

VEGF VEGF plays a prominent role in tumor-associated angiogenesis [71–73]. VEGFR-1 and VEGFR-2 are seldom observed in nondividing endothelial cells but are significantly expressed in association with tumor neovascularization [74–78]. VEGF expression in BTCs is associated with poor survival, metastasis, and disease recurrence. A phase II study of gemcitabine, oxaliplatin, and bevacizumab in advanced BTC reported a response rate of 40%, median PFS of 7 months, and OS of 12.7 months (Table 4) [79]. A single-arm phase II trial of erlotinib and bevacizumab without traditional cytotoxic ©AlphaMed Press 2015

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

Table 3. Clinical studies of EGFR inhibition in biliary tract carcinoma

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Molecular Targets in Biliary Carcinogenesis

6

Table 4. Clinical studies of VEGF inhibition in biliary tract carcinoma Regimen

Phase

Patients, no.

Type of cancers studied

Results

Zhu et al., 2010 [79]

Bevacizumab 1 GEMOX

II

35

IHCC: 22; EHCC: 3; GBC: 10

Lubner et al., 2010 [80]

Bevacizumab 1 erlotinib

II

53

IHCC: 35; EHCC: 8; GBC: 10

Bengala et al., 2010 [81]

Sorafenib

II

46

IHCC: 27; EHCC: 5; GBC: 14

Moehler et al., 2011 [82]

Sorafenib 1 GEM vs. placebo 1 GEM

II

62

IHCC: 40; EHCC: 13; GBC: 9

Yi et al., 2011 [83]

Sunitinib

II

56

IHCC: 35; EHCC: 6; GBC: 15

RR: 40%; median PFS = 7 months ; PFS at 6 months: 63% Median OS: 12.7 months PR: 12%; SD: 51% Median OS: 9.9 months Median PFS: 2.3 months Median OS: 4.4 months PR: 7%; SD: 63% Median PFS: 2.9 months Median OS: 9.4 months Median PFS: 1.7 months PR: 8.9%; SD: 50%

Abbreviations: EHCC, extrahepatic cholangiocarcinoma; GBC, gall bladder cancer; GEM, gemcitabine; GEMOX, gemcitabine 1 oxaliplatin; IHCC, intrahepatic cholangiocarcinoma; SD, stable disease.

chemotherapy in patients with advanced CC and gallbladder cancers demonstrated an 18.4% response rate, time to progression of 4.4 months, and OS of 9.9 months, with potential predictive signal for response seen from EGFR and KRAS status (Table 4) [80]. Another phase II trial combining bevacizumab with gemcitabine and capecitabine (NCT01007552) in patients with advanced BTCs is under way. Other antiangiogenic agents such as sorafenib and sunitinib have failed to show efficacy in this disease, either as single agents or in combination with gemcitabine, with response rates ,10% and survival times less than that seen with other regimens [81–84]. Sorafenib is a multikinase inhibitor of VEGFR-2/-3, PDGFR-b, B-Raf, and C-Raf; it has shown some level of activity in preclinical models of cholangiocarcinoma. Based on this, in a single-arm, open-label, nonrandomized phase II clinical trial, Bengala et al. treated a total of 46 patients with advanced BTCs [81]. Twenty-six patients (56%) were pretreated with chemotherapy. A minimum of 45 days of treatment with sorafenib was completed by 36 patients. The objective response rate was 2% and the rate of stable disease at 12 weeks was 32.6%. For this study, PFS was documentedas 2.3 months (range: 0–12 months) and the median OS was 4.4 months (range: 0–22 months). A significant association was noted between performance status and PFS, with median PFS values for Eastern Cooperative Oncology Group (ECOG) 0 and 1 being 5.7 and 2.1 months, respectively (p 5 .0002). Treatment toxicities included skin rash (35%) and fatigue (33%), which necessitated dose reduction in 22% of patients (Table 4) [81].

HER2 HER2 is also known as ErbB2, c-erbB2, or HER2/neu. It has an intracellular tyrosine kinase domain and an extracellular ligand binding domain. The major signaling pathways mediated by HER2 involve MAPK pathways and PI3K pathways. The California Consortium conducted a phase II study using lapatinib (a dual tyrosine kinase inhibitor of HER2/neu and EGFR pathways) in patients with advanced BTC or hepatobiliary cancer [83]. In all 17 patients recruited into the study, no response was observed. However, HER2 status was not reported (Table 5). A phase I study by Siegel-Lakhai et al. using lapatinib in

combination with oxaliplatin/fluorouracil/leucovorin documented partial response in 2 out of 34 patients (Table 5) [86]. This study also failed to document HER2 status. Another phase II clinical trial reported no response to lapatinib in nine patients with advanced, unresectable BTCs.There were no mutations in HER2/neu or HER2 overexpression in these patients [87].

MEK/MAPK MAP kinase signaling pathways feature in cell proliferation, differentiation, and migration, through the activation of protooncogenes such as JUN, FOS, MYC, and ELK1. There are four identified MAP kinase signaling pathways: the ERK1 pathway, the c-jun N-terminal-regulated kinase (JNK) pathway, the p38 pathway, and the ERK5 pathway [88]. JNK and p38 pathways are mainly stress activated by proinflammatory cytokines. ERK1 and ERK5 are induced by EGFR activation [89]. The inhibition of MEK or ERK carries significant potential as a therapeutic target for BTCs and other solid tumors, but is still being investigated. A phase II trial of selumetinib (MEK1/2 inhibitor) for patients with unresectable BTC recorded an objective response rate (ORR) of 12% and median OS of 9.8 months [90]. Although the ORR was low, 68% of patients had stable disease. Of these, 44% had stable disease for a minimum of 16 weeks; while 12% had stable disease for .1 year (Table 6). The treatment was well tolerated by the patients. Immunohistochemistry testing for KRAS/BRAF genotyping and phosphorylated ERK (pERK) and AKT (pAKT) was performed on all tumor tissues. Patients with short-lived stable disease had KRAS mutations, and absence of pERK staining was associated with no response. In another study, MEK 162 (a selective, ATP-uncompetitive inhibitor of MEK1/2) was used to treat 28 patients with advanced BTC. One complete response, 1 partial response, and 11 cases of stable disease were observed in 26 evaluable patients. The two responders had wild-type extrahepatic CC [91]. There is an ongoing randomized clinical trial (ClinicalTrials.gov identifier NCT02042443) studying trametinib (an MEK inhibitor) compared with chemotherapy with 5-fluorouracil or capecitabine in advanced BTCs.

OTncologist he

©AlphaMed Press 2015

®

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

Author, year

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Oyasiji, Zhang, Kuvshinoff et al.

7

Table 5. Clinical studies of HER2 inhibition in biliary tract carcinoma Author, year

Regimen

Phase

Patients, no.

Type of cancers studied

Results

Ramanathan et al., 2006 [85]

Lapatinib

II

49

BTC: 19; HCC: 30

Siegel-Lakhai et al., 2007 [86]

Lapatinib 1 FOLFOX 4

I

34

Peck et al., 2012 [87]

Lapatinib

II

9

Rectum: 5; pancreatic: 5; CC: 5; duodenum: 2; gastric: 2; cervix: 2; esophagus: 2; ovary: 1; GBC: 1; ACUP: 2 BC: 9

PFS: 1.8 months for BTC and HCC RR: 0; SD: 5 patients (BTC) PR: 2 patients; SD: 8 patients (HCC) PR: 5.9%; SD: 41%

Median PFS: 2.6 months Median OS: 5.1 months

Abbreviations: ACUP, adenocarcinoma of undetermined primary; BC, biliary cancer; BTC, biliary tract cancer; CC, cholangiocarcinoma; GBC, gall bladder cancer; HCC, hepatocellular cancer; OS, overall survival; PFS, progression-free survival; RR, response rate; SD, stable disease.

Author, year

Regimen

Phase

Patients, no.

Type of cancers studied

Results

MEK/MAPK inhibition Bekaii-Saab et al., 2011 [90]

Selumetinib

II

28

GBC: 7; IHCC: 17; EHCC: 4

Finn et al., 2012 [91]

MEK 162

I

28

GBC: 7; IHCC: 14; EHCC: 7

RR: 12% SD: 68% (SD $16 weeks in 56%) Median PFS: 3.7 months Median OS: 9.8 months CR: 1 patient PR: 1 patient SD ($ 6 weeks): 11 patients

PI3K/mTOR/AKT inhibition Rizell et al., 2008 [113]

Sirolimus

30

HCC: 21; CC: 9

SD: 30% (CC) Median OS for CC patients: 7 months

Abbreviations:CC, cholangiocarcinoma; CR, completeresponse;EHCC, extrahepatic cholangiocarcinoma; GBC,gallbladdercancer; HCC, hepatocellularcancer; IHCC, intrahepatic cholangiocarcinoma; PFS, progression-free survival; OS, overall survival; PR, partial response; RR, response rate; SD, stable disease.

IGF/IGFR Autophosphorylation of the IGF-I receptor occurs when IGF-I and IGF-II bind toit.This interaction activates signaling pathways like ERK and the phosphatidylinositide 3-kinase/Akt-1 axis [92]. Other factors that play a role in the activation of IGF-I receptor are IGF-binding proteins and IGF-2 receptors [93–95]. Dysregulation of the IGF system contributes to the proliferation of several neoplasms [96, 97]. While mutations and chromosomal amplifications of IGF-IR are rare, the regulation of its expression is closely associated with the function of several oncogenes and tumor suppressor genes [97]. Elevated serum IGF-I level also increasesthe riskofdeveloping several cancers [94]. Since IGF-IR signaling is crucial to processes like cell migration, angiogenesis, invasion, and metastasis, it plays a significant role in tumor dissemination [98]. The IGF pathway (more specifically, IGF-IR) is active in many malignancies [99], including colorectal, breast, pancreatic, lung, head and neck, prostate, renal, ovarian, and endometrial cancer, as well as sarcomas [100–109]. As such, novel therapeutics are evolving with monoclonal antibodies targeted at the IGF-IR. Inhibition of IGF-IR tyrosine kinase with small molecules has also been explored. However, therapeutic development has been approached very cautiously because of significant concerns for potential toxicity of inhibiting the IGF pathway.The IGF pathway is present in nearly all healthy and malignantcells alike.The IGF-IR

www.TheOncologist.com

also shares significant homology with the insulin receptor. Therefore, the potential for metabolic dysfunction like hyperglycemia is considerable and has been observed in human trials. Successful in vivo and in vitro studies led to several phase I studies that subsequently evolved into phase II or III trials. Results regarding safety and efficacy have been satisfactory; however, long-term effects are still unknown. Details about dosing and scheduling are still being determined. Notably, when anti-IGF pathway treatment is combined with other targeted therapies or cytotoxic chemotherapeutics, toxicity increases. To the best of our knowledge, we are not aware of any clinical trial specifically targeting IGF/IGFR in BTCs. However, we know of phase I/II trials directed at breast cancer, adrenocortical cancer, soft tissue sarcomas, Ewing sarcoma, lung cancer, and hepatocellular cancer [110–112].

PI3K/mTOR/AKT PI3K signaling plays a critical role in cholangiocarcinogenesis, anticancer drug resistance, and autophagy (type II programmed cell-death regulation). Studies have shown disruption of the phosphatidylinositol 3-kinase/AKT/mTOR pathways to be a common occurrence in CC. A prospective, single-arm protocol was used to assess tumor response to sirolimus (an mTOR inhibitor) in 21 patients with advanced HCC and 9 patients with CC. For the HCC patient ©AlphaMed Press 2015

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

Table 6. Clinical studies in biliary tract carcinoma

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Molecular Targets in Biliary Carcinogenesis

8

CONCLUSION While the prognosis is unfavorable in most cases of BTC, progress is being made in the systemic management of this disease.There is no substitution for early detection and improvements in surgical techniques for treating BTC, but opportunities to improve the systemic treatment of this disease abound and should be explored. However, the particular challenges of developing druglike small molecules and antibody-based inhibitors for BTC treatment include (a) targeting DNA methylation and microRNArelated signaling, (b) interactions of multiple signaling pathways, and (c) lack of clinical trials on inhibition of transformation and metastasis. The known alterations of gene expression in BTC are often related to hyper- or hypomethylation of promoters and/or microRNA.Currentinvestigationsmainlyarefocusingonreceptors such as EGFR, HER2,VEGFR, and downstream effectors such as sirolimus and selumetinib inhibition of mTOR and MEK/ERK. The specificity of these drugs targeting downstream events may be low, since the major mediators of BTC are likely related to epigenetics and transcriptional events. Development of

inhibitors targeting BTC-related DNA methylation and microRNA modulation is expected to overcome this obstacle. Epigenetic and transcriptional modulation often impacts multiple signal transduction pathways. Indeed, abnormalities of transformation, cell cycle control, survival, and invasion have been observed in BTC.Targeting a single pathway may not prevent BTC progression. Rational combinations of targeted therapies and targeted therapy with conventional chemotherapy are crucial to future success in treatment. Current clinical trials of inhibitors for BTC treatment are aimed at blocking PI3K/Akt- and MEK/ERK-mediated proliferation and tumor growth. DNA methylation or microRNAaltered cadherin, HOX, and FAK/SRC expression play a critical role in tumorigenesis and metastasis. Therefore, it could be beneficial for BTC patients if specific small molecules or antibodies are developed that attenuate hyperactivation of these molecules associated with transformation and invasion. Inthiseraofpersonalizedmedicine,identificationofpredictive markers, including well-characterized genetic mutations, will help to select specific populations of BTCs for targeted therapy.

AUTHOR CONTRIBUTIONS Conception/Design: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald Provision of study material or patients: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald Collection and/or assembly of data: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald Data analysis and interpretation: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald Manuscript writing: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald Final approval of manuscript: Tolutope Oyasiji, Jianliang Zhang, Boris Kuvshinoff, Renuka Iyer, Steven N. Hochwald

DISCLOSURES Renuka Iyer: Genentech (RF); Steven N. Hochwald: Ethicon Endosurgery (C/A). The other authors indicated no financial relationships. (C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/ inventor/patent holder; (SAB) Scientific advisory board

REFERENCES 1. Hezel AF, Deshpande V, Zhu AX. Genetics of biliary tract cancers and emerging targeted therapies. J Clin Oncol 2010;28:3531–3540. 2. Tonini G, Virz`ı V, Fratto ME et al. Targeted therapy in biliary tract cancer: 2009 update. Future Oncol 2009;5:1675–1684. 3. Marino D, Leone F, Cavalloni G et al. Biliary tract carcinomas: From chemotherapy to targeted therapy. Crit Rev Oncol Hematol 2013;85:136–148. 4. Khan SA, Toledano MB, Taylor-Robinson SD. Epidemiology, risk factors, and pathogenesis of cholangiocarcinoma. HPB (Oxford) 2008;10:77–82.

9. Jarnagin WR, Ruo L, Little SA et al. Patterns of initial disease recurrence after resection of gallbladder carcinoma and hilar cholangiocarcinoma: Implications for adjuvant therapeutic strategies. Cancer 2003;98:1689–1700. 10. Valle J,Wasan H, Palmer DH et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med 2010;362:1273–1281. 11. Okusaka T, Nakachi K, Fukutomi 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: 469–474.

15. Dalmay T, Edwards DR. MicroRNAs and the hallmarks of cancer. Oncogene 2006;25:6170–6175. 16. Kent OA, Mendell JT. A small piece in the cancer puzzle: microRNAs as tumor suppressors and oncogenes. Oncogene 2006;25:6188–6196. 17. Jones PA, Baylin SB.The epigenomics of cancer. Cell 2007;128:683–692. 18. Sriraksa R, Zeller C, Dai W et al. Aberrant DNA methylation at genes associated with a stem cell-like phenotype in cholangiocarcinoma tumors. Cancer Prev Res (Phila) 2013;6:1348–1355.

12. Hezel AF, Zhu AX. Systemic therapy for biliary tract cancers. The Oncologist 2008;13:415–423.

19. Sasaki M, Yamaguchi J, Itatsu K et al. Overexpression of polycomb group protein EZH2 relates to decreased expression of p16 INK4a in cholangiocarcinogenesis in hepatolithiasis. J Pathol 2008;215:175–183.

7. Sirica AE. Cholangiocarcinoma: molecular targeting strategies for chemoprevention and therapy. Hepatology 2005;41:5–15.

13. Karpf AR. Epigenomic reactivation screening to identify genes silenced by DNA hypermethylation in human cancer. Curr Opin Mol Ther 2007;9:231–241.

20. Møller MB. Molecular control of the cell cycle in cancer: Biological and clinical aspects. Dan Med Bull 2003;50:118–138.

8. Berthiaume EP, Wands J. The molecular pathogenesis of cholangiocarcinoma. Semin Liver Dis 2004;24:127–137.

14. Nelson WG, Yegnasubramanian S, Agoston AT et al. Abnormal DNA methylation, epigenetics, and prostate cancer. Front Biosci 2007;12:4254–4266.

21. Agrawal A, Yang J, Murphy RF et al. Regulation of the p14ARF-Mdm2-p53 pathway: An overview in breast cancer. Exp Mol Pathol 2006;81:115–122.

5. Shaib Y, El-Serag HB. The epidemiology of cholangiocarcinoma. Semin Liver Dis 2004;24:115–125. 6. Gores GJ. Cholangiocarcinoma: Current concepts and insights. Hepatology 2003;37:961–969.

OTncologist he

©AlphaMed Press 2015

®

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

group, one partial remission and five cases of stable disease were observed at 3 months, while the CC group had three cases of stable disease.The median survival for HCC patients treated under the protocol was 6.5 months (range: 0.2–36 months) and 7 months (range: 2.6–35 months) for CC patients (supplemental online Table 1) [113]. The Multi-Institutional Phase II Study of the Akt Inhibitor MK-2206 in Refractory Biliary Cancers (ClinicalTrials.gov identifier NCT-01425879) is an ongoing, multi-institutional phase II study of the Akt inhibitor MK-2206 in refractory biliary cancers. This trial is designed to show how effective MD2206 is in treating patients with advanced refractory biliary cancer that cannot be removed by surgery. The findings of another clinical trial (NCT01859182) assessing the response of unresectable gallbladdercancerand bileductcancer tocombinationtherapy with selumetinib (MEK inhibitor) and Akt inhibitor MK-2206 are also pending.

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Oyasiji, Zhang, Kuvshinoff et al.

9 40. Meng F, Wehbe-Janek H, Henson R et al. Epigenetic regulation of microRNA-370 by interleukin6 in malignant human cholangiocytes. Oncogene 2008; 27:378–386.

57. Jiao Y, Pawlik TM, Anders RA et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet 2013;45:1470–1473.

23. Lee S, Kim WH, Jung HY et al. Aberrant CpG island methylation of multiple genes in intrahepatic cholangiocarcinoma. Am J Pathol 2002;161:1015–1022.

41. Meng F, Henson R, Wehbe-Janek H et al. The microRNA let-7a modulates interleukin-6-dependent STAT-3 survival signaling in malignant human cholangiocytes. J Biol Chem 2007;282:8256–8264.

58. Li M, Zhang Z, Li X et al. Whole-exome and targeted gene sequencing of gallbladder carcinoma identifies recurrent mutations in the ErbB pathway. Nat Genet 2014;46:872–876.

42. Kawase H, Fujii K, Miyamoto M et al. Differential LC-MS-based proteomics of surgical human cholangiocarcinoma tissues. J Proteome Res 2009;8: 4092–4103.

59. Simbolo M, Fassan M, Ruzzenente A et al. Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget 2014;5:2839–2852.

43. Yonglitthipagon P, Pairojkul C, Bhudhisawasdi V et al. Proteomics-based identification of a-enolase as a potential prognostic marker in cholangiocarcinoma. Clin Biochem 2012;45:827–834.

60. Ross JS, Wang K, Gay L et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. The Oncologist 2014;19:235–242.

44. Borger DR, Tanabe KK, Fan KC et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. The Oncologist 2012;17:72–79.

61. Arai Y, Totoki Y, Hosoda F et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology 2014;59:1427–1434.

24. Tischoff I, Wittekind C, Tannapfel A. Role of epigenetic alterations in cholangiocarcinoma. J Hepatobiliary Pancreat Surg 2006;13:274–279. 25. Isomoto H, Mott JL, Kobayashi S et al. Sustained IL-6/STAT-3 signaling in cholangiocarcinoma cells due to SOCS-3 epigenetic silencing. Gastroenterology 2007;132:384–396. 26. Sugawara H, Yasoshima M, Katayanagi K et al. Relationship between interleukin-6 and proliferation and differentiation in cholangiocarcinoma. Histopathology 1998;33:145–153. 27. Yasoshima M, Kono N, Sugawara H et al. Increased expression of interleukin-6 and tumor necrosis factor-alpha in pathologic biliary epithelial cells: in situ and culture study. Lab Invest 1998;78:89–100. 28. Hodge DR, Xiao W, Clausen PA et al. Interleukin-6 regulation of the human DNA methyltransferase (HDNMT) gene in human erythroleukemia cells. J Biol Chem 2001;276:39508–39511. 29. Wehbe H, Henson R, Meng F et al. Interleukin-6 contributes to growth in cholangiocarcinoma cells by aberrant promoter methylation and geneexpression. Cancer Res 2006;66:10517–10524. 30. Doki Y, Shiozaki H, Tahara H et al. Correlation between E-cadherin expression and invasiveness in vitro in a human esophageal cancer cell line. Cancer Res 1993;53:3421–3426. 31. Oka H, Shiozaki H, Kobayashi K et al. Expression of E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis. Cancer Res 1993;53:1696–1701. 32. Sommers CL, Thompson EW, Torri JA et al. Cell adhesion molecule uvomorulin expression in human breast cancer cell lines: Relationship to morphology and invasive capacities. Cell Growth Differ 1991;2:365–372. 33. Kim BH, Cho NY, Choi M et al. Methylation profiles of multiple CpG island loci in extrahepatic cholangiocarcinoma versus those of intrahepatic cholangiocarcinomas. Arch Pathol Lab Med 2007; 131:923–930. 34. Brennecke J, Hipfner DR, Stark A et al. bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 2003;113: 25–36. 35. Xu P, Vernooy SY, Guo M et al. The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol 2003;13:790–795. 36. Chen X. A microRNA as a translational repressor of APETALA2 in Arabidopsis flower development. Science 2004;303:2022–2025.

45. Kipp BR, Voss JS, Kerr SE et al. Isocitrate dehydrogenase 1 and 2 mutations in cholangiocarcinoma. Hum Pathol 2012;43:1552–1558. 46. Khansaard W, Techasen A, Namwat N et al. Increased EphB2 expression predicts cholangiocarcinoma metastasis. Tumour Biol 2014;35:10031– 10041. 47. Enkhbold C, Utsunomiya T, Morine Y et al. Loss of FBXW7 expression is associated with poor prognosis in intrahepatic cholangiocarcinoma. Hepatol Res 2014;44:E346–E52. 48. Yi S, Yang ZL, Miao X et al. N-cadherin and P-cadherin are biomarkers for invasion, metastasis, and poor prognosis of gallbladder carcinomas. Pathol Res Pract 2014;210:363–368. 49. Gravdal K, Halvorsen OJ, Haukaas SA et al. A switch from E-cadherin to N-cadherin expression indicates epithelial to mesenchymal transition and is of strong and independent importance for the progress of prostate cancer. Clin Cancer Res 2007; 13:7003–7011. 50. Kubo Y, Aishima S, Tanaka Y et al. Different expression of glucose transporters in the progression of intrahepatic cholangiocarcinoma. Hum Pathol 2014;45:1610–1617. 51. O’Dell MR, Huang JL, Whitney-Miller CL et al. Kras(G12D) and p53 mutation cause primary intrahepatic cholangiocarcinoma. Cancer Res 2012;72: 1557–1567. 52. 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–S681. 53. Isa T, Tomita S, Nakachi A et al. Analysis of microsatellite instability, K-ras gene mutation and p53 protein overexpression in intrahepatic cholangiocarcinoma. Hepatogastroenterology 2002;49: 604–608.

37. Dostie J, Mourelatos Z,Yang M et al. Numerous microRNPs in neuronal cells containing novel microRNAs. RNA 2003;9:180–186.

54. Liu XF, Zhang H, Zhu SG et al. Correlation of p53 gene mutation and expression of P53 protein in cholangiocarcinoma. World J Gastroenterol 2006; 12:4706–4709.

38. Meng F, Henson R, Lang M et al. Involvement of human micro-RNA in growth and response to chemotherapy in human cholangiocarcinoma cell lines. Gastroenterology 2006;130:2113–2129.

55. Kiguchi K, Carbajal S, Chan K et al. Constitutive expression of ErbB-2 in gallbladder epithelium results in development of adenocarcinoma. Cancer Res 2001;61:6971–6976.

39. Mott JL, Kobayashi S, Bronk SF et al. mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene 2007;26:6133–6140.

56. Sekiya S, Suzuki A. Intrahepatic cholangiocarcinoma can arise from Notch-mediated conversion of hepatocytes. J Clin Invest 2012;122:3914–3918.

www.TheOncologist.com

62. Wu YM, Su F, Kalyana-Sundaram S et al. Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov 2013;3:636–647. 63. Borad MJ, Champion MD, Egan JB 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. 64. Philip PA, Mahoney MR, Allmer C et al. Phase II study of erlotinib in patients with advanced biliary cancer. J Clin Oncol 2006;24:3069–3074. 65. Lee J, Park SH, Chang HM 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: 181–188. 66. Ciombor C,Chan E, McClanahanP, Fan K,Flynn J, Young R.. Phase Ib study of gemcitabine and oxaliplatin with erlotinib in patients with advanced biliary tract cancer. J Clin Oncol 2012;30(suppl): e14503. 67. Gruenberger B, Schueller J, Heubrandtner U et al. Cetuximab, gemcitabine, and oxaliplatin in patients with unresectable advanced or metastatic biliary tract cancer: A phase 2 study. Lancet Oncol 2010;11:1142–1148. 68. Malka D, Rousseau V,Trarbach T, Boucher E, De La Fouchardiere C, Faivre S.. Gemcitabine and oxaliplatin alone or in combination with cetuximab as first-line treatment for advanced biliary cancer: Final analysis of a randomized phase II trial (BINGO). J Clin Oncol 2012;30(suppl):4032. 69. Jensen L, Ploen J, Hansen T, Jakobsen A.. Marker driven systemic treatment of inoperable cholangiocarcinomas: Panitumumab and combination chemotherapy in KRAS wild-type tumors. J Clin Oncol 2011;29(suppl):4101. 70. Lim H, Chang H, Kim J.. Phase III study of gemcitabine/oxaliplatin (GEMOX) with or without erlotinib in unresectable, metastatic biliary tract carcinoma. J Clin Oncol 2011;29(suppl):4032. 71. Ferrara N. Molecular and biological properties of vascular endothelial growth factor. J Mol Med (Berl) 1999;77:527–543. 72. Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol 1999;9:211–220. 73. Neufeld G, Cohen T, Gengrinovitch S et al. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 1999;13:9–22.

©AlphaMed Press 2015

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

22. Yang B, House MG, Guo M et al. Promoter methylation profiles of tumor suppressor genes in intrahepatic and extrahepatic cholangiocarcinoma. Mod Pathol 2005;18:412–420.

Published Ahead of Print on May 29, 2015 as 10.1634/theoncologist.2014-0442.

Molecular Targets in Biliary Carcinogenesis

10 74. Brown LF, Berse B, Jackman RW et al. Expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in adenocarcinomas of the gastrointestinal tract. Cancer Res 1993;53:4727–4735. 75. Plate KH, Breier G, Weich HA et al. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 1992;359:845–848. 76. Takahashi Y, Kitadai Y, Bucana CD et al. Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res 1995;55:3964–3968.

78. Tang D, Nagano H, Yamamoto H et al. Angiogenesis in cholangiocellular carcinoma: expression of vascular endothelial growth factor, angiopoietin1/2, thrombospondin-1 and clinicopathological significance. Oncol Rep 2006;15:525–532. 79. Zhu AX, Meyerhardt JA, Blaszkowsky LS 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. 80. Lubner SJ, Mahoney MR, Kolesar JL 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. 81. Bengala C, Bertolini F, Malavasi N et al. Sorafenib in patients with advanced biliary tract carcinoma: A phase II trial. Br J Cancer 2010;102:68–72. 82. Moehler M, Kanzler S, Woerns S, Denzer U, Kolligs F.. A randomized, double-blind, multicenter phase II AIO trial with gemcitabine plus sorafenib versus gemcitabine plus placebo in patients with chemotherapy-naive advanced or metastatic biliary tract cancer: First safety and efficacy data. J Clin Oncol 2011;29(suppl):a4077. 83. Yi J, Doval D, Lee J, Cho M, Park S.. Phase II study of sunitinib as second-line treatment in advanced biliary tract carcinoma: Multicenter, multinational study. J Clin Oncol 2011;29(suppl):a14653. 84. El-Khoueiry AB, Ramanathan RK, Yang DY et al. A randomized phase II of gemcitabine and sorafenib versus sorafenib alone in patients with metastatic pancreatic cancer. Invest New Drugs 2012;30: 1175–1183. 85. Ramanathan R, Belani C, Singh D.. Phase II study of lapatinib, a dual inhibitor of epidermal growth factor receptor (EGFR) tyrosine kinase 1 and 2 (Her2/neu) in patients with advanced biliary tree

86. Siegel-Lakhai WS, Beijnen JH, Vervenne WL et al. Phase I pharmacokinetic study of the safety and tolerability of lapatinib (GW572016) in combination with oxaliplatin/fluorouracil/leucovorin (FOLFOX4) in patients with solid tumors. Clin Cancer Res 2007;13:4495–4502. 87. Peck J, Wei L, Zalupski M et al. HER2/neu may not be an interesting target in biliary cancers: Results of an early phase II study with lapatinib. Oncology 2012;82:175–179. 88. Garrington TP, Johnson GL. Organization and regulation of mitogen-activated protein kinase signaling pathways. Curr Opin Cell Biol 1999;11: 211–218.

99. F¨urstenberger G, Senn HJ. Insulin-like growth factors and cancer. Lancet Oncol 2002;3:298–302. 100. Chan JM, Stampfer MJ, Giovannucci E et al. Plasma insulin-like growth factor-I and prostate cancer risk: a prospective study. Science 1998;279:563–566. 101. Donovan EA, Kummar S. Role of insulin-like growth factor-1R system in colorectal carcinogenesis. Crit Rev Oncol Hematol 2008;66:91–98. 102. Scotlandi K, Picci P. Targeting insulin-like growth factor 1 receptor in sarcomas. Curr Opin Oncol 2008;20:419–427. 103. Gunter MJ, Hoover DR, Yu H et al. A prospective evaluation of insulin and insulin-like growth factor-I as risk factors for endometrial cancer. Cancer Epidemiol Biomarkers Prev 2008;17:921–929.

89. Kato Y, Tapping RI, Huang S et al. Bmk1/Erk5 is required for cell proliferation induced by epidermal growth factor. Nature 1998;395:713–716.

104. Sayer RA, Lancaster JM, Pittman J et al. High insulin-like growth factor-2 (IGF-2) gene expression is an independent predictor of poor survival for patients with advanced stage serous epithelial ovarian cancer. Gynecol Oncol 2005;96:355–361.

90. Bekaii-Saab T, Phelps MA, Li X et al. Multiinstitutional phase II study of selumetinib in patients with metastatic biliary cancers. J Clin Oncol 2011;29: 2357–2363.

105. Wu X, Zhao H,DoKA etal.Serumlevels ofinsulin growth factor (IGF-I) and IGF-binding protein predict risk of second primary tumors in patients with head and neck cancer. Clin Cancer Res 2004;10:3988–3995.

91. Finn RS, Javle MM, Tan BR et al. A phase I study of MEK inhibitor MEK162 (ARRY-438162) in patients with biliary tract cancer. J Clin Oncol 2012;30(suppl 4):a220.

106. Yu H, Spitz MR, Mistry J et al. Plasma levels of insulin-like growth factor-I and lung cancer risk: a case-control analysis. J Natl Cancer Inst 1999;91: 151–156.

92. Yu H, Rohan T. Role of the insulin-like growth factor family in cancer development and progression. J Natl Cancer Inst 2000;92:1472–1489.

107. Bergmann U, Funatomi H, Yokoyama M et al. Insulin-like growth factor I overexpression in human pancreatic cancer: Evidence for autocrine and paracrine roles. Cancer Res 1995;55:2007–2011.

93. Remacle-Bonnet M, Garrouste F, el Atiq F et al. des-(1-3)-IGF-I, an insulin-like growth factor analog used to mimic a potential IGF-II autocrine loop, promotes the differentiation of human coloncarcinoma cells. Int J Cancer 1992;52:910–917. 94. Ma J, Pollak MN, Giovannucci E et al. Prospective studyofcolorectal cancer risk in men and plasma levels of insulin-like growth factor (IGF)-I and IGF-binding protein-3. J Natl Cancer Inst 1999;91:620–625. 95. Harper J, Burns JL, Foulstone EJ et al. Soluble IGF2 receptor rescues Apc(Min/1) intestinal adenoma progression induced by Igf2 loss of imprinting. Cancer Res 2006;66:1940–1948. 96. Adachi Y, Yamamoto H, Imsumran A. Insulinlike growth factor-I receptor as a candidate for a novel molecular target in the gastrointestinal cancer. Dig Endosc 2006;18:245–251. 97. Foulstone E, Prince S, Zaccheo O et al. Insulinlike growth factor ligands, receptors, and binding proteins in cancer. J Pathol 2005;205:145–153. 98. Adachi Y, Li R, Yamamoto H et al. Insulin-like growth factor-I receptor blockade reduces the invasiveness of gastrointestinal cancers via blocking production of matrilysin. Carcinogenesis 2009;30: 1305–1313.

108. Parker A, Cheville JC, Lohse C et al. Expression of insulin-like growth factor I receptor and survival in patients with clear cell renal cell carcinoma. J Urol 2003;170:420–424. 109. Hankinson SE, Willett WC, Colditz GA et al. Circulating concentrations of insulin-like growth factorIandriskofbreastcancer.Lancet1998;351:1393–1396. 110. Ma CX, Suman VJ, Goetz M et al. A phase I trial of the IGF-1R antibody Cixutumumab in combination with temsirolimus in patients with metastatic breast cancer. Breast Cancer Res Treat 2013;139:145–153. 111. Schwartz GK, Tap WD, Qin LX et al. Cixutumumab and temsirolimus for patients with bone and soft-tissue sarcoma: A multicentre, open-label, phase 2 trial. Lancet Oncol 2013;14:371–382. 112. Naing A, LoRusso P, Fu S et al. Insulin growth factor-receptor (IGF-1R) antibody cixutumumab combined with the mTOR inhibitor temsirolimus in patients with refractory Ewing’s sarcoma family tumors. Clin Cancer Res 2012;18:2625–2631. 113. Rizell M, Andersson M, Cahlin C et al. Effects of the mTOR inhibitor sirolimus in patients with hepatocellular and cholangiocellular cancer. Int J Clin Oncol 2008;13:66–70.

See http://www.TheOncologist.com for supplemental material available online.

OTncologist he

©AlphaMed Press 2015

®

Downloaded from http://theoncologist.alphamedpress.org/ at Countway Library of Medicine on July 6, 2015

77. Benckert C, Jonas S,Cramer T etal.Transforming growth factor beta 1 stimulates vascular endothelial growth factor gene transcription in human cholangiocellular carcinoma cells. Cancer Res 2003;63: 1083–1092.

cancer (BTC) or hepatocellular cancer HCC): A California Consortium (CCC-P) trial. J Clin Oncol 2006;24(18S):4010.

Molecular Targets in Biliary Carcinogenesis and Implications for Therapy.

Biliary tract cancers (BTCs) encompass a group of invasive carcinomas, including cholangiocarcinoma (intrahepatic, perihilar, or extrahepatic), and ga...
912KB Sizes 1 Downloads 8 Views