Abstract

Background

Biliary tract cancers (BTCs) are highly fatal malignancies that make up less than 1% of all cancers. BTC is often diagnosed at an unresectable stage; surgical resection remains the only definitive treatment. Brain metastases (BMs) from BTC are extremely rare, and few studies on patients with BMs from BTC exist. The aim of this study was to identify clinical characteristics associated with poor prognosis for patients with BMs from BTC.

Materials and Methods

We performed a retrospective review of electronic medical records for patients with BMs from BTC managed at Mount Sinai Hospital from 2000 to 2017. Data on patient characteristics, magnetic resonance imaging findings, treatment regimens, and clinical outcomes were analyzed.

Results

We identified 1,910 patients with BTC. Nine patients developed BMs, with an incidence of 0.47%. Of these nine patients, six had intrahepatic cholangiocarcinoma, two had extrahepatic cholangiocarcinoma, and one had gallbladder cancer. Six (66.7%) patients had one BM, one (11.1%) patient had two BMs, and two (22.2%) patients had three or more BMs. Four (44.4%) patients underwent BM resection, and seven (77.8%) received BM radiation. Median overall survival from time of BM diagnosis was 3.8 months (95% confidence interval 0.1–16.9).

Conclusion

Development of BMs from BTC is rare; however, prognosis is less than 4 months. BM diagnosis can occur within 2 years of primary diagnosis. As targeted therapeutics emerge, future studies ought to focus on identifying genomic BM markers associated with BTC subtypes.

Implications for Practice

In the largest retrospective study of biliary tract cancer brain metastases, the clinical presentation and outcomes are reported of nine patients with an extremely rare clinical entity. The genomic literature and potential therapeutic targets for these patients with limited treatment options is comprehensively and exhaustively discussed.

Introduction

Biliary tract cancers (BTCs) are highly fatal malignancies that make up less than 1% of all cancers globally [1]. These cancers are generally of the adenocarcinoma histologic subtype and include extrahepatic and intrahepatic cholangiocarcinoma (CC), gallbladder cancer (GBC), and cancer of the ampulla and papilla of Vater [2, 3]. The incidence of BTC varies widely by region of the world, with the lowest incidence in Western countries and the highest in Asia and Latin America [4].

Owing to nonspecific presenting symptoms, BTC is often diagnosed at an advanced, unresectable stage; radical surgical resection remains the only definitive treatment option for BTC and portends the best prognosis [5]. Thus, prognosis for BTC is poor, with 5‐year survival rates of 10% for CC and 5% for GBC [6, 7]. Chemotherapy can improve survival in patients with advanced BTC and is the current standard of care for patients with unresectable disease [3]. First‐line chemotherapy is gemcitabine combined with a platinum agent. Other regimens are not well‐established despite many clinical trials for new chemotherapeutic regimens and targeted therapies [1, 8].

BTC commonly invades adjacent lymph nodes and the liver and less frequently metastasizes to distant sites including lungs and bone. Metastasis to the brain is extremely rare. The incidence of brain metastases (BMs) from BTC is largely unknown. Only a few case reports and two case series have characterized BMs from BTC [914]. The most recent case series reports the incidence of BMs from BTC to be 1.4% [10].

As such, there is an unmet need to understand clinical outcomes of patients with BTC who develop BMs. Our study aims to expand the existing literature by further describing patient outcomes for those who develop BMs from BTC within the context of the genomic era.

Materials and Methods

Patient Cohort

We performed a retrospective review of electronic medical records for patients with BMs from BTC managed at Mount Sinai Hospital from 2000 to 2017. Inclusion criteria were (a) presence of primary CC or GBC confirmed by a board‐certified pathologist and (b) presence of at least one metastatic brain lesion confirmed by computed tomography (CT) or magnetic resonance (MR) imaging. Nine patients met these criteria and were included for analysis. Data on patient characteristics, treatment regimens, and clinical outcomes were extracted and are presented in Table 1. The study was reviewed and approved by the Mount Sinai Institutional Review Board and was in compliance with Health Insurance Portability and Accountability Act guidelines.

Table 1.

Patient characteristics

CharacteristicsPatient no.
123456789
Age at primary diagnosis, years66.863.760.848.164.764.947.454.868.1
SexFFMFFMMFF
Smoking statusFormerNeverFormerNeverCurrentUnknownNeverCurrentFormer
Cardiac diseaseNoYesYesYesNoNoNoYesYes
DiabetesNoYesYesYesNoNoNoNoNo
Primary tumor locationICCICCICCICCECCICCICCGBCECC
Resection of primaryYesNoYesYesNoYesNoYesYes
Metastatic sites prior to BMLiver, lungsLungs, retroperitonealLiver, lungsLiver, lungs, chest wallNoneLungsLungs, boneNoneNone
Chemotherapy prior to BMYesNoYesYesYesNoYesYesYes
Radiation to primary prior to BMNoNoNoYesNoNoNoYesYes
Presenting symptomsHeadache, dizzinessAMS, dizzinessSeizureAMS, weaknessHeadache, weaknessAMSSeizureSeizureDizziness, decreased sensation, facial pain
Number of BMs13+11113+21
Location of BMCerebellumCerebellum, parietalFronto‐parietalTemporalFrontalFrontalCerebellum, all lobesCerebellum, frontalParietal
Resection of BMYesNoNoYesNoYesNoNoYes
BM radiationWBRT, SRSNoneWBRTSRSWBRTNoneWBRTSRSWBRT, SRS
Time between primary tumor diagnosis and BM diagnosis, months66.70.712.336.214.725.216.81616.7
OS from BM diagnosis, months16.90.11.33.11.51.53.83.210
CharacteristicsPatient no.
123456789
Age at primary diagnosis, years66.863.760.848.164.764.947.454.868.1
SexFFMFFMMFF
Smoking statusFormerNeverFormerNeverCurrentUnknownNeverCurrentFormer
Cardiac diseaseNoYesYesYesNoNoNoYesYes
DiabetesNoYesYesYesNoNoNoNoNo
Primary tumor locationICCICCICCICCECCICCICCGBCECC
Resection of primaryYesNoYesYesNoYesNoYesYes
Metastatic sites prior to BMLiver, lungsLungs, retroperitonealLiver, lungsLiver, lungs, chest wallNoneLungsLungs, boneNoneNone
Chemotherapy prior to BMYesNoYesYesYesNoYesYesYes
Radiation to primary prior to BMNoNoNoYesNoNoNoYesYes
Presenting symptomsHeadache, dizzinessAMS, dizzinessSeizureAMS, weaknessHeadache, weaknessAMSSeizureSeizureDizziness, decreased sensation, facial pain
Number of BMs13+11113+21
Location of BMCerebellumCerebellum, parietalFronto‐parietalTemporalFrontalFrontalCerebellum, all lobesCerebellum, frontalParietal
Resection of BMYesNoNoYesNoYesNoNoYes
BM radiationWBRT, SRSNoneWBRTSRSWBRTNoneWBRTSRSWBRT, SRS
Time between primary tumor diagnosis and BM diagnosis, months66.70.712.336.214.725.216.81616.7
OS from BM diagnosis, months16.90.11.33.11.51.53.83.210

Abbreviations: AMS, altered mental status; BM, brain metastasis; ECC, extrahepatic cholangiocarcinoma; F, female; GBC, gallbladder cancer; ICC, intrahepatic cholangiocarcinoma; M, male; OS, overall survival; SRS, stereotactic radiosurgery; WBRT, whole‐brain radiation therapy.

Table 1.

Patient characteristics

CharacteristicsPatient no.
123456789
Age at primary diagnosis, years66.863.760.848.164.764.947.454.868.1
SexFFMFFMMFF
Smoking statusFormerNeverFormerNeverCurrentUnknownNeverCurrentFormer
Cardiac diseaseNoYesYesYesNoNoNoYesYes
DiabetesNoYesYesYesNoNoNoNoNo
Primary tumor locationICCICCICCICCECCICCICCGBCECC
Resection of primaryYesNoYesYesNoYesNoYesYes
Metastatic sites prior to BMLiver, lungsLungs, retroperitonealLiver, lungsLiver, lungs, chest wallNoneLungsLungs, boneNoneNone
Chemotherapy prior to BMYesNoYesYesYesNoYesYesYes
Radiation to primary prior to BMNoNoNoYesNoNoNoYesYes
Presenting symptomsHeadache, dizzinessAMS, dizzinessSeizureAMS, weaknessHeadache, weaknessAMSSeizureSeizureDizziness, decreased sensation, facial pain
Number of BMs13+11113+21
Location of BMCerebellumCerebellum, parietalFronto‐parietalTemporalFrontalFrontalCerebellum, all lobesCerebellum, frontalParietal
Resection of BMYesNoNoYesNoYesNoNoYes
BM radiationWBRT, SRSNoneWBRTSRSWBRTNoneWBRTSRSWBRT, SRS
Time between primary tumor diagnosis and BM diagnosis, months66.70.712.336.214.725.216.81616.7
OS from BM diagnosis, months16.90.11.33.11.51.53.83.210
CharacteristicsPatient no.
123456789
Age at primary diagnosis, years66.863.760.848.164.764.947.454.868.1
SexFFMFFMMFF
Smoking statusFormerNeverFormerNeverCurrentUnknownNeverCurrentFormer
Cardiac diseaseNoYesYesYesNoNoNoYesYes
DiabetesNoYesYesYesNoNoNoNoNo
Primary tumor locationICCICCICCICCECCICCICCGBCECC
Resection of primaryYesNoYesYesNoYesNoYesYes
Metastatic sites prior to BMLiver, lungsLungs, retroperitonealLiver, lungsLiver, lungs, chest wallNoneLungsLungs, boneNoneNone
Chemotherapy prior to BMYesNoYesYesYesNoYesYesYes
Radiation to primary prior to BMNoNoNoYesNoNoNoYesYes
Presenting symptomsHeadache, dizzinessAMS, dizzinessSeizureAMS, weaknessHeadache, weaknessAMSSeizureSeizureDizziness, decreased sensation, facial pain
Number of BMs13+11113+21
Location of BMCerebellumCerebellum, parietalFronto‐parietalTemporalFrontalFrontalCerebellum, all lobesCerebellum, frontalParietal
Resection of BMYesNoNoYesNoYesNoNoYes
BM radiationWBRT, SRSNoneWBRTSRSWBRTNoneWBRTSRSWBRT, SRS
Time between primary tumor diagnosis and BM diagnosis, months66.70.712.336.214.725.216.81616.7
OS from BM diagnosis, months16.90.11.33.11.51.53.83.210

Abbreviations: AMS, altered mental status; BM, brain metastasis; ECC, extrahepatic cholangiocarcinoma; F, female; GBC, gallbladder cancer; ICC, intrahepatic cholangiocarcinoma; M, male; OS, overall survival; SRS, stereotactic radiosurgery; WBRT, whole‐brain radiation therapy.

Imaging Analysis

Axial T1 postcontrast gadolinium‐enhanced MR images were reviewed using OsiriX software. Tumor cross‐sectional area (CSA) was calculated for each case by drawing perpendicular lines on the axial slice that visualized the most prominent tumor size. Presence of vasogenic edema was assessed using axial T2 fluid‐attenuated inversion recovery (FLAIR) images. Contrast‐enhanced CT images were analyzed in cases where an MR was not available. For patients with more than one BM, CSA was calculated for the largest lesion.

Statistical Analysis

Statistical analysis was conducted using statistical database software (JMP version Pro 14, SAS Institute Inc, Cary, NC). Overall survival was calculated as time from BM imaging diagnosis to date of death or last follow‐up using Kaplan‐Meier and chi‐square proportional hazards methodologies; p ≤ .05 was used to denote significance.

Results

There were 1,910 patients with BTC treated at our institution between 2000 and 2017. Of these, nine patients (0.47%) developed BMs (Fig. 1). Six patients (66.7%) were female and three were male (33.3%). Smoking status was reported as never smoker (33.3%), former smoker (33.3%), current smoker (22.2%), and unknown (11.1%). The most common medical comorbidities included cardiac disease (55.6%), type 2 diabetes (33.3%), pulmonary disease (11.1%), and cirrhosis (11.1%). One patient had systemic lupus erythematosus.

Patient with intrahepatic cholangiocarcinoma with a single brain metastasis to the L cerebellum. Hematoxylin and eosin stains (low power [×4]) of primary liver with tumor and adjacent hepatocytes (A) and (high power [×20]) of primary liver adenocarcinoma (B). Hematoxylin and eosin stains of brain metastasis at ×4 (C) and ×20 (D), showing similar histologic features as seen in the liver.
Figure 1.

Patient with intrahepatic cholangiocarcinoma with a single brain metastasis to the L cerebellum. Hematoxylin and eosin stains (low power [×4]) of primary liver with tumor and adjacent hepatocytes (A) and (high power [×20]) of primary liver adenocarcinoma (B). Hematoxylin and eosin stains of brain metastasis at ×4 (C) and ×20 (D), showing similar histologic features as seen in the liver.

Primary tumor diagnosis included six patients (66.7%) with intrahepatic CC (ICC), two (22.2%) with extrahepatic CC (ECC), and one (11.1%) with GBC. Median age at primary diagnosis was 63.7 years (range, 47.4–68.1 years). Six patients (66.7%) underwent resection of their primary disease site, with five of six patients (83.3%) having negative margins upon histological evaluation. Additionally, seven patients (77.8%) received chemotherapy and three (33.3%) received radiation prior to BM diagnosis.

Metastatic sites prior to BM diagnosis included lung in 66.7% as well as liver, chest wall, and bone. All patients underwent an abdominal/pelvic CT prior to BM diagnosis to assess for the presence of vertebral metastases. Median time from abdominal/pelvic CT to BM diagnosis was 46 days (95% confidence interval [CI] 4–113). One (11.1%) patient had vertebral metastases with sclerotic lesions present in the pelvic and iliac bones, sacrum, and L4 vertebral body.

The most common presenting symptoms at BM presentation included seizure (33.3%), dizziness (33.3%), altered mental status (33.3%), weakness (33.3%), headache (22.2%), and cranial neuropathy (11.1%). Median age at BM diagnosis was 63.7 years (range, 48.8–72.4 years). Median time from primary diagnosis to BM diagnosis was 16.7 months (range, 0.7–66.7 months).

Six patients (66.7%) had one BM, one patient (11.1%) had two BMs, and two patients (22.2%) had three or more BMs upon imaging evaluation at initial presentation. Five (55.6%) patients had a supratentorial lesion, one patient (11.1%) had an infratentorial lesion (Fig. 2), and the remaining three (33.3%) patients had both supratentorial and infratentorial lesions. The most common parenchymal lobe with metastatic disease was frontal (n = 5, 55.6%), followed by parietal (n = 4, 44.4%), cerebellar (n = 4, 44.4%), temporal (n = 2, 22.2%), and occipital (n = 1, 11.1%). Hydrocephalus was not appreciated on any scan. Vasogenic edema was visualized on T2 FLAIR sequence in all cases. Mean tumor CSA was 6.9 cm2 ± 7.1 (Fig. 1).

Postcontrast T1 weighted magnetic resonance image demonstrating single brain metastasis in the L cerebellum. (A): Sagittal view and (B): axial view.
Figure 2.

Postcontrast T1 weighted magnetic resonance image demonstrating single brain metastasis in the L cerebellum. (A): Sagittal view and (B): axial view.

Four (44.4%) patients underwent definitive surgical resection, and seven (77.8%) received BM radiation. Radiation modalities included stereotactic radiosurgery (SRS) in two patients, whole‐brain radiation therapy (WBRT) in three patients, and both SRS and WBRT in three patients. Median overall survival from time of BM diagnosis was 3.8 months (95% CI 0.1–16.9). Patients who received BM radiotherapy had significantly improved overall survival of 10.2 months (95% CI 3.8–16.9) compared with 0.8 months (95% CI 0.1–1.5; p = .0039, log‐rank; RR = 0.38; p = .01, chi‐square).

Discussion

Case Series

Limited studies have evaluated brain metastases from biliary tract cancers. Beyond the few case reports of patients with BMs from BTC, two case series of eight [9] patients from Thailand and six [10] patients from Italy have been reported in the literature. In our series of nine patients, the median age at BM diagnosis was 63.7 years, which is similar to that reported in the two case series of 60 years and 68 years, respectively. The incidence of BM in our cohort was 0.47%, compared with 0.15% and 1.4%, respectively. Additionally, we report a median survival of 3.8 months for patients with BMs from BTC, which is similar to 9.5 weeks and 3.7 months, respectively. When compared with BM from other primary cancers, median survival for BTC is similar to the survival seen in non‐small cell lung cancer (4 months) and head/neck cancer (5 months) and shorter than the survival seen in melanoma (6 months) and breast cancer (12 months) [15].

In our cohort, median time from primary diagnosis to BM diagnosis was 16.7 months, compared with 8 months [9] and 13.6 months [10], respectively. The longer time to BM diagnosis in our cohort compared with that seen in the other two case series may reflect differences in clinical outcomes due to wide genomic variability seen in BTC and varying responses to treatment [16, 17].

We identified a significant survival difference between patients who received BM radiotherapy (10.2 months) compared with those who did not receive BM radiotherapy (0.8 months); however, we acknowledge the limitation that only two patients did not receive radiation. Moreover, although there was no statistical difference between patients who received neurosurgical resection compared with those who did not (data not shown), given the underpowered sample, we caution against clinical interpretation of these findings. Indeed, further institutional case series and a meta‐analysis of previously reported cases are warranted to identify prognostic factors that are associated with survival in patients with BMs from BTC.

Genomics of BTC

Treatment of BTC has traditionally relied on gemcitabine‐based chemotherapy. Recently, genomic characterization of patients with BTC has led to the identification of clinically actionable mutations and subsequent development of targeted therapies. Early efforts to characterize the genomics of BTC relied on genotyping of hotspot oncogenes, leading to the identification of recurrent mutations in known oncogenic pathways, such as the epidermal growth factor receptor (EGFR) pathway, the mitogen‐activated protein kinase (MAPK) pathway, and the phosphoinositide‐3‐kinase (PI3K) pathway [18].

Genomic differences between subtypes of BTC have been identified using next‐generation sequencing. For example, mutations in isocitrate dehydrogenase 1 or 2 (IDH1/2) and FGFR2 gene fusions are more likely to be seen in ICC; KRAS and human epidermal growth receptor 2 (HER2) mutations are more likely to be seen in ECC; mutations in EGFR, HER2/neu, and PIK3CA are more likely to be seen in GBC [19].

Furthermore, within ICC, genomic differences have been reported based on ICC etiology. Whole‐exome sequencing of 15 CC cases revealed that non–liver‐fluke ICC are more likely to harbor BAP1 and IDH mutations, whereas liver‐fluke ICC are more likely to harbor TP53 and SMAD4 mutations [20]. Of note, 30% of ICC have FGFR, IDH, or BRAF mutations, all of which have targeted therapies currently in clinical trials [18].

Genomics of Brain Metastases from BTC

To our knowledge, there have been no efforts to characterize the genomic landscape of BMs from BTC, likely because BMs from BTC is such a rare event. Furthermore, patients with BMs are often ineligible from inclusion in clinical trials, which is a major reason why there is a lack of information on how to treat patients with BMs. Given the dismal prognosis of patients with BMs from BTC, it is important to investigate the genomic events that support metastasis to the brain so that more advanced therapeutic options may be elucidated.

Because BMs from BTC have not yet been genomically characterized, the importance of targeted therapies in the treatment of BMs from BTC remains unclear. However, there is evidence to support targeting of known genomic alterations in patients with BMs from other primary cancers, including EGFR inhibitors in lung cancer [2125], PI3K inhibitors in BMs from breast cancer [26], BRAF inhibitors in BMs from melanoma [27], and vascular endothelial growth factor receptor tyrosine kinase inhibitors in BMs from renal cell carcinoma [28]. The role of targeting genomic alterations in patients with BMs is currently under study (Alliance A071701; NCT03994796) in a national cooperative group phase II clinical trial.

One limitation in treatment of BMs is poor penetration of systemic therapies through the blood–brain barrier (BBB), due to factors that include low passive permeability and efflux pumps. However, advanced cancer may alter BBB permeability, thereby supporting susceptibility to certain targeted agents [29]. Furthermore, BBB permeability is heterogeneous and associated with vascular remodeling [30]. Therefore, more work is needed to further explore the ability of targeted agents to penetrate the BBB and elucidate their potential role in treating BMs from BTC.

Clinical Trials in BTC

A number of clinical trials targeting mutations in BTC have been initiated in recent years. These trials and the mutations that they target are summarized below. Many of the trials focus on the treatment of primary BTC and often do not include patients with BM.

MAPK Pathway

Efforts to target the MAPK pathway in patients with BTC have shown mixed results. A phase II study of the MEK inhibitor selumetinib in patients with advanced BTC reported an 80% disease control rate and a median overall survival of 9.8 months for patients receiving selumetinib as first‐line therapy [31]. More recently, in a phase Ib trial of selumetinib (ABC‐04) in combination with gemcitabine and cisplatin, selumetinib did not show clinical benefit in the treatment of advanced BTC [32]. Trials of the multikinase inhibitor sorafenib have shown little success as well [3336]. Selumetinib may have the potential to penetrate the BBB given efficacy in the treatment of pediatric low‐grade glioma [37].

FGFR2 Fusions

Fusions have been observed between the receptor tyrosine kinase FGFR2 and other genes including AHCYL1, BICC1, PARK2, MGEA5, TACC3, KIAA1598, KCTD1, and TXLNA [3841]. A phase II trial of the FGFR inhibitor BGJ398 showed efficacy in controlling disease in patients with advanced or metastatic cholangiocarcinoma harboring FGFR2 fusions or other alterations [42]. Furthermore, a phase II trial of the nonselective FGFR inhibitor ponatinib is ongoing (NCT02272998). Although there are no data regarding BGJ398 or ponatinib and the BBB, the FGFR inhibitor dovitinib is able to cross the BBB with limited efficacy in the treatment of glioblastoma [43].

IDH

There are multiple clinical trials ongoing that target mutations in IDH1/2. Most notably, the IDH1 inhibitor AG‐120 has shown efficacy in stabilizing disease progression in patients with IDH1‐mutant ICC [44], and a phase III trial for patients with IDH1‐mutant cholangiocarcinoma is currently underway (NCT02989857). Certain patients with IDH‐mutant ICC are sensitive to the multikinase inhibitor dasatinib [45]. Finally, the IDH inhibitors BAY1436032 (NCT02746081), IDH305 (NCT02381886), and AG‐881 (NCT02481154) are currently in phase I testing. Emerging preclinical data support the use of the IDH1 inhibitor BAY 1436032 for the treatment of IDH1 mutant astrocytoma in vivo [46]. Clinical studies are underway (NCT03030066, NCT02481154).

HER2/neu

An estimated 15% of GBC have HER2/neu amplification and overexpression [47], which can be targeted with HER2 antagonists including trastuzumab, afatinib, and lapatinib; however, early trials of HER2 antagonists combined with chemotherapy to treat advanced BTC were not impressive [4850]. A phase II trial of trastuzumab for a select group of patients with HER2‐positive BTC is ongoing (NCT02999672). With regard to the BBB, trastuzumab has been shown to cross the BBB [51].

PIK3CA

The PI3K/AKT/mTOR pathway is a target in many cancers, but data are limited on the role of PIK3CA inhibitors in the treatment of BTC. One study of the mTOR inhibitor everolimus in 27 patients with advanced BTC demonstrated a median overall survival of 9.5 months [52]. Further studies are needed to elucidate the role of PI3K pathway inhibitors in patients with BTC. Importantly, PI3K and mTOR inhibitors have shown good BBB penetration [53].

Immunotherapy for BTC

Recently, great advances have occurred in the field of immunotherapy with the rise of checkpoint inhibitors. Checkpoint inhibitors have efficacy in the treatment of untreated or progressive brain metastases from patients with melanoma and non‐small cell lung cancer [54]. However, few studies have evaluated the role of immunotherapy in patients with BTC.

In one study, a unique whole‐exome sequencing approach was used to determine that tumor‐infiltrating lymphocytes (TILs) from a patient with metastatic cholangiocarcinoma contained CD4+ Helper 1 (TH1) cells that recognized a mutation in erbb2 interacting protein (ERBB2IP). Tumor regression was achieved by using adoptive transfer of TILs containing mutation‐specific polyfunctional TH1 cells. The patient was re‐treated with mutation‐reactive TH1 cells upon disease progression, and tumor regression was again seen [55]. Additionally, in a recent study of pembrolizumab in pretreated BTC with programmed death‐ligand 1 expression, the objective response rate was 17% [56].

Conclusion

Development of BMs from BTC is an extremely rare event associated with dismal prognosis, and as such, few case series of patients with BMs from BTC exist. Future prospective cohort studies are necessary to fully understand prognostic factors that may be associated with survival in patients with BMs from BTC as well as to drive clinical treatment paradigms. As our understanding of the underlying genomics of BM improves, clinical trials that target actionable mutations in patients with BM are encouraged.

Acknowledgments

P.K.B. receives grant funding from Damon Runyon Cancer Research Foundation, Breast Cancer Research Foundation, Susan G. Komen, and NIH.

Author Contributions

Conception/design: Megan R. D'Andrea, Corey M. Gill, Priscilla K. Brastianos, Raj K. Shrivastava

Provision of study material or patients: Megan R. D'Andrea, Corey M. Gill, Joshua B. Bederson, Raj K. Shrivastava

Collection and/or assembly of data: Megan R. D'Andrea, Corey M. Gill, Melissa Umphlett, Nadejda M. Tsankova, Mary Fowkes, Joshua B. Bederson, Raj K. Shrivastava

Data analysis and interpretation: Megan R. D'Andrea, Corey M. Gill, Priscilla K. Brastianos, Raj K. Shrivastava

Manuscript writing: Megan R. D'Andrea, Corey M. Gill, Priscilla K. Brastianos, Raj K. Shrivastava

Final approval of manuscript: Megan R. D'Andrea, Corey M. Gill, Melissa Umphlett, Nadejda M. Tsankova, Mary Fowkes, Joshua B. Bederson, Priscilla K. Brastianos, Raj K. Shrivastava

Disclosures

Priscilla K. Brastianos: Eli Lilly and Company, Tesaro, Angiochem, Merck (C/A), Merck, Pfizer (RF), Genentech‐Roche (H). 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

Ghosn
 
M
.
Optimum chemotherapy for the management of advanced biliary tract cancer
.
World J Gastroenterol
 
2015
;
21
:
4121
.

2

de
 
Groen
 
PC
,
Gores
 
GJ
,
Larusso
 
NF
et al.
Biliary tract cancers
.
N Engl J Med
 
1999
;
341
:
1368
1378
.

3

Sasaki
 
T
,
Isayama
 
H
,
Nakai
 
Y
et al.
Current status of chemotherapy for the treatment of advanced biliary tract cancer
.
Korean J Intern Med
 
2013
;
28
:
515
524
.

4

Randi
 
G
,
Malvezzi
 
M
,
Levi
 
F
et al.
Epidemiology of biliary tract cancers: An update
.
Ann Oncol
 
2008
;
20
:
146
159
.

5

Eckel
 
F
,
Brunner
 
T
,
Jelic
 
S
et al.
Biliary cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow‐up
.
Ann Oncol
 
2011
;
22
:
vi40
vi44
.

6

Misra
 
S
,
Chaturvedi
 
A
,
Misra
 
NC
et al.
Carcinoma of the gallbladder
.
Lancet Oncol
 
2003
;
4
:
167
176
.

7

Razumilava
 
N
,
Gores
 
GJ
.
Cholangiocarcinoma
.
Lancet
 
2014
;
383
:
2168
2179
.

8

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
.

9

Chindaprasirt
 
J
,
Sookprasert
 
A
,
Sawanyawisuth
 
K
et al.
Brain metastases from cholangiocarcinoma: A first case series in Thailand
.
Asian Pac J Cancer Prev
 
2012
;
13
:
1995
1997
.

10

Frega
 
G
,
Garajová
 
I
,
Palloni
 
A
et al.
Brain metastases from biliary tract cancer: A monocentric retrospective analysis of 450 patients
.
Oncology
 
2018
;
94
:
7
11
.

11

Kilbourn
 
KJ
,
Aferzon
 
J
,
Menon
 
M
.
Isolated brain metastasis in cholangiocarcinoma: A case report and review of literature
.
Conn Med
 
2014
;
78
:
161
162
.

12

Mirrakhimov
 
AE
,
Nwankwo
 
N
,
Zdunek
 
T
et al.
Cholangiocarcinoma and brain lesions: An extremely rare finding
.
Case Rep
 
2013
;
2013
:bcr2013009235–bcr2013009235.

13

William
 
BM
,
Grem
 
JL
.
Brain metastasis and leptomeningeal carcinomatosis in a patient with cholangiocarcinoma
.
Gastrointest Cancer Res
 
2011
;
4
:
144
145
.

14

Yu
 
Z
,
Xu
 
J
,
Wang
 
J
.
Isolated brain metastases prior to locoregional recurrence in hilar cholangiocarcinoma
.
Mol Clin Oncol
 
2017
;
6
:
899
902
.

15

Cagney
 
DN
,
Martin
 
AM
,
Catalano
 
PJ
et al.
Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: A population‐based study
.
Neuro Oncol
 
2017
;
19
:
1511
1521
.

16

Nakamura
 
H
,
Arai
 
Y
,
Totoki
 
Y
et al.
Genomic spectra of biliary tract cancer
.
Nat Genet
 
2015
;
47
:
1003
1010
.

17

Farshidfar
 
F
,
Zheng
 
S
,
Gingras
 
MC
et al.
Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH ‐mutant molecular profiles
.
Cell Rep
 
2017
;
18
:
2780
2794
.

18

Jain
 
A
,
Kwong
 
LN
,
Javle
 
M
.
Genomic profiling of biliary tract cancers and implications for clinical practice
.
Curr Treat Options Oncol
 
2016
;
17
:
58
.

19

Zhao
 
DY
,
Lim
 
KH
.
Current biologics for treatment of biliary tract cancers
.
J Gastrointest Oncol
 
2017
;
8
:
430
440
.

20

Chan‐On
 
W
,
Nairismägi
 
ML
,
Ong
 
CK
et al.
Exome sequencing identifies distinct mutational patterns in liver fluke–related and non‐infection‐related bile duct cancers
.
Nat Genet
 
2013
;
45
:
1474
1478
.

21

Rosell
 
R
,
Carcereny
 
E
,
Gervais
 
R
et al.
Erlotinib versus standard chemotherapy as first‐line treatment for European patients with advanced EGFR mutation‐positive non‐small‐cell lung cancer (EURTAC): A multicentre, open‐label, randomised phase 3 trial
.
Lancet Oncol
 
2012
;
13
:
239
246
.

22

Soria
 
JC
,
Ohe
 
Y
,
Vansteenkiste
 
J
et al.
Osimertinib in untreated EGFR ‐mutated advanced non–small‐cell lung cancer
.
N Engl J Med
 
2018
;
378
:
113
125
.

23

Mok
 
TS
,
Wu
 
YL
,
Thongprasert
 
S
et al.
Gefitinib or carboplatin‐paclitaxel in pulmonary adenocarcinoma
.
N Engl J Med
 
2009
;
361
:
947
957
.

24

Sequist
 
LV
,
Yang
 
JC
,
Yamamoto
 
N
et al.
Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations
.
J Clin Oncol
 
2013
;
31
:
3327
3334
.

25

Lee
 
CK
,
Davies
 
L
,
Wu
 
YL
et al.
Gefitinib or erlotinib vs chemotherapy for EGFR mutation‐positive lung cancer: Individual patient data meta‐analysis of overall survival
.
J Natl Cancer Inst
 
2017
;
109
.

26

Ippen
 
FM
,
Grosch
 
JK
,
Subramanian
 
M
et al.
Targeting the PI3K/Akt/mTOR pathway with the pan‐Akt inhibitor GDC‐0068 in PIK3CA‐mutant breast cancer brain metastases
.
Neuro Oncol
 
2019
[Epub ahead of print].

27

Davies
 
MA
,
Saiag
 
P
,
Robert
 
C
et al.
Dabrafenib plus trametinib in patients with BRAFV600‐mutant melanoma brain metastases (COMBI‐MB): A multicentre, multicohort, open‐label
,
phase 2 trial. Lancet Oncol
 
2017
;
18
:
863
873
.

28

Juloori
 
A
,
Miller
 
JA
,
Parsai
 
S
et al.
Overall survival and response to radiation and targeted therapies among patients with renal cell carcinoma brain metastases
.
J Neurosurg
 
2019
;
1
9
.

29

Chacko
 
AM
,
Li
 
C
,
Pryma
 
DA
et al.
Targeted delivery of antibody‐based therapeutic and imaging agents to CNS tumors: Crossing the blood–brain barrier divide
.
Expert Opin Drug Deliv
 
2013
;
10
:
907
926
.

30

Lockman
 
PR
,
Mittapalli
 
RK
,
Taskar
 
KS
et al.
Heterogeneous blood‐tumor barrier permeability determines drug efficacy in experimental brain metastases of breast cancer
.
Clin Cancer Res
 
2010
;
16
:
5664
5678
.

31

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

32

Bridgewater
 
J
,
Lopes
 
A
,
Beare
 
S
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
.

33

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
.

34

El‐Khoueiry
 
AB
,
Rankin
 
CJ
,
Ben‐Josef
 
E
et al.
SWOG 0514: A phase II study of sorafenib in patients with unresectable or metastatic gallbladder carcinoma and cholangiocarcinoma
.
Invest New Drugs
 
2012
;
30
:
1646
1651
.

35

Lee
 
JK
,
Capanu
 
M
,
O'Reilly
 
EM
et al.
A phase II study of gemcitabine and cisplatin plus sorafenib in patients with advanced biliary adenocarcinomas
.
Br J Cancer
 
2013
;
109
:
915
919
.

36

Moehler
 
M
,
Maderer
 
A
,
Schimanski
 
C
et al.
Gemcitabine plus sorafenib versus gemcitabine alone in advanced biliary tract cancer: A double‐blind placebo‐controlled multicentre phase II AIO study with biomarker and serum programme
.
Eur J Cancer
 
2014
;
50
:
3125
3135
.

37

Banerjee
 
A
,
Jakacki
 
RI
,
Onar‐Thomas
 
A
et al.
A phase I trial of the MEK inhibitor selumetinib (AZD6244) in pediatric patients with recurrent or refractory low‐grade glioma: A Pediatric Brain Tumor Consortium (PBTC) study
.
Neuro Oncol
 
2017
;
19
:
1135
1144
.

38

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
.

39

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.

40

Churi
 
CR
,
Shroff
 
R
,
Wang
 
Y
et al.
Mutation profiling in cholangiocarcinoma: Prognostic and therapeutic implications
.
PLoS One
 
2014
;
9
:e115383.

41

Wu
 
YM
,
Su
 
F
,
Kalyana‐Sundaram
 
S
et al.
Identification of targetable FGFR gene fusions in diverse cancers
.
Cancer Discov
 
2013
;
3
:
636
647
.

42

Javle
 
M
,
Lowery
 
M
,
Shroff
 
RT
et al.
Phase II study of BGJ398 in patients with FGFR‐altered advanced cholangiocarcinoma
.
J Clin Oncol
 
2018
;
36
:
276
282
.

43

Schäfer
 
N
,
Gielen
 
GH
,
Kebir
 
S
et al.
Phase I trial of dovitinib (TKI258) in recurrent glioblastoma
.
J Cancer Res Clin Oncol
 
2016
;
142
:
1581
1589
.

44

Lowery
 
MA
,
Abou‐Alfa
 
GK
,
Burris
 
HA
et al.
Phase I study of AG‐120, an IDH1 mutant enzyme inhibitor: Results from the cholangiocarcinoma dose escalation and expansion cohorts
.
J Clin Oncol
 
2017
;
35
:
4015
4015
.

45

Saha
 
SK
,
Gordan
 
JD
,
Kleinstiver
 
BP
et al.
Isocitrate dehydrogenase mutations confer dasatinib hypersensitivity and SRC dependence in intrahepatic cholangiocarcinoma
.
Cancer Discov
 
2016
;
6
:
727
739
.

46

Pusch
 
S
,
Krausert
 
S
,
Fischer
 
V
et al.
Pan‐mutant IDH1 inhibitor BAY 1436032 for effective treatment of IDH1 mutant astrocytoma in vivo
.
Acta Neuropathol
 
2017
;
133
:
629
644
.

47

Javle
 
M
,
Bekaii‐Saab
 
T
,
Jain
 
A
et al.
Biliary cancer: Utility of next‐generation sequencing for clinical management
.
Cancer
 
2016
;
122
:
3838
3847
.

48

Kwak
 
EL
,
Shapiro
 
GI
,
Cohen
 
SM
et al.
Phase 2 trial of afatinib, an ErbB family blocker, in solid tumors genetically screened for target activation
.
Cancer
 
2013
;
119
:
3043
3051
.

49

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
.

50

Ramanathan
 
RK
,
Belani
 
CP
,
Singh
 
DA
et al.
A phase II study of lapatinib in patients with advanced biliary tree and hepatocellular cancer
.
Cancer Chemother Pharmacol
 
2009
;
64
:
777
783
.

51

Terrell‐Hall
 
TB
,
Nounou
 
MI
,
El‐Amrawy
 
F
et al.
Trastuzumab distribution in an in‐vivo and in‐vitro model of brain metastases of breast cancer
.
Oncotarget
 
2017
;
8
:
83734
83744
.

52

Lau
 
DK
,
Tay
 
RY
,
Yeung
 
YH
et al.
Phase II study of everolimus (RAD001) monotherapy as first‐line treatment in advanced biliary tract cancer with biomarker exploration: The RADiChol Study
.
Br J Cancer
 
2018
;
118
:
966
971
.

53

Wen
 
PY
,
Lee
 
EQ
,
Reardon
 
DA
et al.
Current clinical development of PI3K pathway inhibitors in glioblastoma
.
Neuro Oncol
 
2012
;
14
:
819
829
.

54

Goldberg
 
SB
,
Gettinger
 
SN
,
Mahajan
 
A
et al.
Pembrolizumab for patients with melanoma or non‐small‐cell lung cancer and untreated brain metastases: Early analysis of a non‐randomised, open‐label, phase 2 trial
.
Lancet Oncol
 
2016
;
17
:
976
983
.

55

Tran
 
E
,
Turcotte
 
S
,
Gros
 
A
et al.
Cancer immunotherapy based on mutation‐specific CD4+ T cells in a patient with epithelial cancer
.
Science
 
2014
;
344
:
641
645
.

56

Bang
 
YJ
,
Doi
 
T
,
Braud
 
FD
et al.
525 Safety and efficacy of pembrolizumab (MK‐3475) in patients (pts) with advanced biliary tract cancer: Interim results of KEYNOTE‐028
.
Eur J Cancer
 
2015
;
51
:
S112
.

Author notes

Disclosures of potential conflicts of interest may be found at the end of this article.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)