201
|
Ilyas SI, Borad MJ. The rise of the FGFR inhibitor in advanced biliary cancer: the next cover of time magazine? J Gastrointest Oncol 2016; 7:789-796. [PMID: 27747092 PMCID: PMC5056253 DOI: 10.21037/jgo.2016.08.12] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/19/2016] [Indexed: 12/27/2022] Open
Abstract
Cholangiocarcinomas (CCAs) are heterogeneous tumors arising from the biliary tract with features of cholangiocyte differentiation. CCAs are aggressive tumors with limited treatment options and poor overall survival. Only a subset of CCA patients with early stage disease can avail potentially curative treatment options. For advanced biliary tract tumors, currently there are limited effective treatment modalities. Recent advances have provided greater insight into the genomic landscape of CCAs. The fibroblast growth factor receptor (FGFR) pathway is involved in key cellular processes essential to survival and differentiation. Accordingly, aberrant FGFR signaling has significant oncogenic potential. Recent discovery of FGFR2 gene fusions in CCA has heightened interest in FGFR inhibition in advanced biliary tract cancer. These findings have served as a catalyst for ongoing clinical investigation of FGFR inhibition in CCA patients with various FGFR signaling abnormalities. Herein, we review FGFR aberrations in CCA and their prognostic implications, FGFR targeting as a viable therapeutic option in advanced biliary tract malignancies, and future directions for development of combination approaches utilizing FGFR inhibition.
Collapse
Affiliation(s)
- Sumera I. Ilyas
- Division of Gastroenterology and Hepatology, Mayo
Clinic, Rochester, Minnesota, USA
| | - Mitesh J. Borad
- Division of Hematology and Oncology, Mayo Clinic,
Scottsdale, Arizona, USA
| |
Collapse
|
202
|
Wang Y, Ding X, Wang S, Moser CD, Shaleh HM, Mohamed EA, Chaiteerakij R, Allotey LK, Chen G, Miyabe K, McNulty MS, Ndzengue A, Barr Fritcher EG, Knudson RA, Greipp PT, Clark KJ, Torbenson MS, Kipp BR, Zhou J, Barrett MT, Gustafson MP, Alberts SR, Borad MJ, Roberts LR. Antitumor effect of FGFR inhibitors on a novel cholangiocarcinoma patient derived xenograft mouse model endogenously expressing an FGFR2-CCDC6 fusion protein. Cancer Lett 2016; 380:163-73. [PMID: 27216979 PMCID: PMC5119950 DOI: 10.1016/j.canlet.2016.05.017] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/15/2022]
Abstract
Cholangiocarcinoma is a highly lethal cancer with limited therapeutic options. Recent genomic analysis of cholangiocarcinoma has revealed the presence of fibroblast growth factor receptor 2 (FGFR2) fusion proteins in up to 13% of intrahepatic cholangiocarcinoma (iCCA). FGFR fusions have been identified as a novel oncogenic and druggable target in a number of cancers. In this study, we established a novel cholangiocarcinoma patient derived xenograft (PDX) mouse model bearing an FGFR2-CCDC6 fusion protein from a metastatic lung nodule of an iCCA patient. Using this PDX model, we confirmed the ability of the FGFR inhibitors, ponatinib, dovitinib and BGJ398, to modulate FGFR signaling, inhibit cell proliferation and induce cell apoptosis in cholangiocarcinoma tumors harboring FGFR2 fusions. In addition, BGJ398 appeared to be superior in potency to ponatinib and dovitinib in this model. Our findings provide a strong rationale for the investigation of FGFR inhibitors, particularly BGJ398, as a therapeutic option for cholangiocarcinoma patients harboring FGFR2 fusions.
Collapse
Affiliation(s)
- Yu Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China; Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Xiwei Ding
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA; Department of Gastroenterology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Shaoqing Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA; Department of Pathology, Qiqihar Medical University, Qiqihar, China
| | - Catherine D Moser
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Hassan M Shaleh
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Essa A Mohamed
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Roongruedee Chaiteerakij
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Loretta K Allotey
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Gang Chen
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Katsuyuki Miyabe
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Melissa S McNulty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Albert Ndzengue
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA
| | - Emily G Barr Fritcher
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Ryan A Knudson
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Patricia T Greipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Karl J Clark
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Michael S Torbenson
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Benjamin R Kipp
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Michael T Barrett
- Division of Hematology and Medical Oncology, Mayo Clinic College of Medicine, Phoenix, AZ, USA
| | - Michael P Gustafson
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Steven R Alberts
- Department of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Mitesh J Borad
- Division of Hematology and Medical Oncology, Mayo Clinic College of Medicine, Phoenix, AZ, USA
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Mayo Clinic Cancer Center, Rochester, MN, USA.
| |
Collapse
|
203
|
Liang J, She Y, Zhu J, Wei L, Zhang L, Gao L, Wang Y, Xing J, Guo Y, Meng X, Li P. Development and validation of an ultra-high sensitive next-generation sequencing assay for molecular diagnosis of clinical oncology. Int J Oncol 2016; 49:2088-2104. [PMID: 27826616 DOI: 10.3892/ijo.2016.3707] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/07/2016] [Indexed: 11/06/2022] Open
Abstract
Dramatic improvements in the understanding of oncogenes have spurred the development of molecular target therapies, which created an exigent need for comprehensive and rapid clinical genotyping. Next-generation sequencing (NGS) assay with increased performance and decreased cost is becoming more widely used in clinical diagnosis. However, the optimization and validation of NGS assay remain a challenge, especially for the detection of somatic variants at low mutant allele fraction (MAF). In the present study, we developed and validated the Novogene Comprehensive Panel (NCP) based on targeted capture for NGS analysis. Due to the high correlation between SNV/INDEL detection performance and target coverage, here we focused on these two types of variants for our deep sequencing strategy. To validate the capability of NCP in single-nucleotide variant (SNV) and small insert and deletion (INDEL) detection, we implemented a practical validation strategy with pooled cell lines, deep sequencing of pooled samples (>2000X average unique coverage across target region) achieving >99% sensitivity and high specificity (positive predictive value, PPV >99%) for all types of variations with expected MAF >5%. Furthermore, given the high sensitivity and that false positive may exist in this assay, we confirmed its accuracy of variants with MAF <5% using 35 formalin-fixed and paraffin-embedded (FFPE) tumor specimens by Quantstudio 3D Digital PCR (dPCR; Life Technologies) and obtained a high consistency (32 of 35 mutations detected by NGS were verified). We also used the amplification refractory mutation system (ARMS) to verify the variants with a MAF in a broad range of 2-63% detected in 33 FFPE samples and reached a 100% PPV for this assay. As a potential clinical diagnosis tool, NCP can robustly and comprehensively analyze clinical-related genes with high sensitivity and low cost.
Collapse
Affiliation(s)
- Jiao Liang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, National Engineering Laboratory for Anti-tumor Therapeutics, Tsinghua University, Beijing 100084, P.R. China
| | - Yaoguang She
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Jiaqi Zhu
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Longgang Wei
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Lanying Zhang
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Lianju Gao
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Yan Wang
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Jing Xing
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Yang Guo
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Xuehong Meng
- Novogene Bioinformatics Institute, Beijing 100083, P.R. China
| | - Peiyu Li
- Department of General Surgery, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
204
|
Lloyd KCK, Khanna C, Hendricks W, Trent J, Kotlikoff M. Precision medicine: an opportunity for a paradigm shift in veterinary medicine. J Am Vet Med Assoc 2016; 248:45-8. [PMID: 26684088 DOI: 10.2460/javma.248.1.45] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
205
|
Hall TG, Yu Y, Eathiraj S, Wang Y, Savage RE, Lapierre JM, Schwartz B, Abbadessa G. Preclinical Activity of ARQ 087, a Novel Inhibitor Targeting FGFR Dysregulation. PLoS One 2016; 11:e0162594. [PMID: 27627808 PMCID: PMC5023172 DOI: 10.1371/journal.pone.0162594] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/25/2016] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of Fibroblast Growth Factor Receptor (FGFR) signaling through amplifications, mutations, and gene fusions has been implicated in a broad array of cancers (e.g. liver, gastric, ovarian, endometrial, and bladder). ARQ 087 is a novel, ATP competitive, small molecule, multi-kinase inhibitor with potent in vitro and in vivo activity against FGFR addicted cell lines and tumors. Biochemically, ARQ 087 exhibited IC50 values of 1.8 nM for FGFR2, and 4.5 nM for FGFR1 and 3. In cells, inhibition of FGFR2 auto-phosphorylation and other proteins downstream in the FGFR pathway (FRS2α, AKT, ERK) was evident by the response to ARQ 087 treatment. Cell proliferation studies demonstrated ARQ 087 has anti-proliferative activity in cell lines driven by FGFR dysregulation, including amplifications, fusions, and mutations. Cell cycle studies in cell lines with high levels of FGFR2 protein showed a positive relationship between ARQ 087 induced G1 cell cycle arrest and subsequent induction of apoptosis. In addition, ARQ 087 was effective at inhibiting tumor growth in vivo in FGFR2 altered, SNU-16 and NCI-H716, xenograft tumor models with gene amplifications and fusions. ARQ 087 is currently being studied in a phase 1/2 clinical trial that includes a sub cohort for intrahepatic cholangiocarcinoma patients with confirmed FGFR2 gene fusions (NCT01752920).
Collapse
Affiliation(s)
- Terence G. Hall
- ArQule, Inc., Burlington, MA, United States of America
- * E-mail:
| | - Yi Yu
- ArQule, Inc., Burlington, MA, United States of America
| | | | - Yunxia Wang
- ArQule, Inc., Burlington, MA, United States of America
| | | | | | | | | |
Collapse
|
206
|
Javle M, Bekaii-Saab T, Jain A, Wang Y, Kelley RK, Wang K, Kang HC, Catenacci D, Ali S, Krishnan S, Ahn D, Bocobo AG, Zuo M, Kaseb A, Miller V, Stephens PJ, Meric-Bernstam F, Shroff R, Ross J. Biliary cancer: Utility of next-generation sequencing for clinical management. Cancer 2016; 122:3838-3847. [PMID: 27622582 DOI: 10.1002/cncr.30254] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Biliary tract cancers (BTCs) typically present at an advanced stage, and systemic chemotherapy is often of limited benefit. METHODS Hybrid capture-based comprehensive genomic profiling (CGP) was performed for 412 intrahepatic cholangiocarcinomas (IHCCAs), 57 extrahepatic cholangiocarcinomas (EHCCAs), and 85 gallbladder carcinomas (GBCAs). The mutational profile was correlated with the clinical outcome of standard and experimental therapies for 321 patients. Clinical variables, detected mutations, and administered therapies were correlated with overall survival (OS) in a Cox regression model. RESULTS The most frequent genetic aberrations (GAs) observed were tumor protein 53 (TP53; 27%), cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B; 27%), KRAS (22%), AT-rich interactive domain-containing protein 1A (ARID1A; 18%), and isocitrate dehydrogenase 1 (IDH1; 16%) in IHCCA; KRAS (42%), TP53 (40%), CDKN2A/B (17%), and SMAD4 (21%) in EHCCA; and TP53 (59%), CDKN2A/B (19%), ARID1A (13%), and ERBB2 (16%) in GBCA. Fibroblast growth factor receptor (FGFR; 11%) and IDH mutations (20%) were mostly limited to IHCCA but appeared to be mutually exclusive. In the IHCCA group, TP53 and KRAS mutations were associated significantly with poor OS, whereas FGFR2 mutations were associated with improved OS (P = .001), a younger age at onset, and female sex. IDH1/2 mutations were not prognostic. In a multivariate model, the effects of TP53 and FGFR GAs remained significant (P < .05). Patients with FGFR GAs had superior OS with FGFR-targeted therapy versus standard regimens (P = .006). Targeted therapy in IHCCA was associated with a numerical OS improvement (P = .07). CONCLUSIONS This is the largest clinically annotated data set of BTC cases with CGP and indicates the potential of CGP for improving outcomes. CGP should be strongly considered in the management of BTC patients. Cancer 2016;122:3838-3847. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Milind Javle
- Department of Gastrointestinal (GI) Medical Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Tanios Bekaii-Saab
- Division of Medical Oncology, Ohio State University Medical Center, Columbus, Ohio
| | - Apurva Jain
- Department of Gastrointestinal (GI) Medical Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Ying Wang
- Department of Bioinformatics & Computational Biology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Robin Katie Kelley
- Department of Medicine (Hematology/Oncology), UCSF, San Francisco, California
| | - Kai Wang
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Hyunseon C Kang
- Department of Diagnostic Radiology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Daniel Catenacci
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, Illinois
| | - Siraj Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | - Sunil Krishnan
- Department of Radiation Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Daniel Ahn
- Division of Medical Oncology, Ohio State University Medical Center, Columbus, Ohio
| | - Andrea Grace Bocobo
- Department of Medicine (Hematology/Oncology), UCSF, San Francisco, California
| | - Mingxin Zuo
- Department of Gastrointestinal (GI) Medical Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Ahmed Kaseb
- Department of Gastrointestinal (GI) Medical Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | | | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, UT-MD Anderson Cancer Center, Houston, Texas
| | - Rachna Shroff
- Department of Gastrointestinal (GI) Medical Oncology, UT-MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Ross
- Department of Pathology, Albany Medical College Albany, New York
| |
Collapse
|
207
|
Goeppert B, Ernst C, Baer C, Roessler S, Renner M, Mehrabi A, Hafezi M, Pathil A, Warth A, Stenzinger A, Weichert W, Bähr M, Will R, Schirmacher P, Plass C, Weichenhan D. Cadherin-6 is a putative tumor suppressor and target of epigenetically dysregulated miR-429 in cholangiocarcinoma. Epigenetics 2016; 11:780-790. [PMID: 27593557 DOI: 10.1080/15592294.2016.1227899] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cholangiocarcinoma (CC) is a rare malignancy of the extrahepatic or intrahepatic biliary tract with an outstanding poor prognosis. Non-surgical therapeutic regimens result in minimally improved survival of CC patients. Global genomic analyses identified a few recurrently mutated genes, some of them in genes involved in epigenetic patterning. In a previous study, we demonstrated global DNA methylation changes in CC, indicating major contribution of epigenetic alterations to cholangiocarcinogenesis. Here, we aimed at the identification and characterization of CC-related, differentially methylated regions (DMRs) in potential microRNA promoters and of genes targeted by identified microRNAs. Twenty-seven hypermethylated and 13 hypomethylated potential promoter regions of microRNAs, known to be associated with cancer-related pathways like Wnt, ErbB, and PI3K-Akt signaling, were identified. Selected DMRs were confirmed in 2 independent patient cohorts. Inverse correlation between promoter methylation and expression suggested miR-129-2 and members of the miR-200 family (miR-200a, miR-200b, and miR-429) as novel tumor suppressors and oncomiRs, respectively, in CC. Tumor suppressor genes deleted in liver cancer 1 (DLC1), F-box/WD-repeat-containing protein 7 (FBXW7), and cadherin-6 (CDH6) were identified as presumed targets in CC. Tissue microarrays of a representative and well-characterized cohort of biliary tract cancers (n=212) displayed stepwise downregulation of CDH6 and association with poor patient outcome. Ectopic expression of CDH6 on the other hand, delayed growth in the CC cell lines EGI-1 and TFK-1, together suggesting a tumor suppressive function of CDH6. Our work represents a valuable repository for the study of epigenetically altered miRNAs in cholangiocarcinogenesis and novel putative, CC-related tumor suppressive miRNAs and oncomiRs.
Collapse
Affiliation(s)
| | - Christina Ernst
- b Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Constance Baer
- b Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | | | - Marcus Renner
- a Institute of Pathology, University Hospital Heidelberg , Germany
| | - Arianeb Mehrabi
- c Department of General , Visceral, and Transplantation Surgery, University Hospital Heidelberg , Germany
| | - Mohammadreza Hafezi
- c Department of General , Visceral, and Transplantation Surgery, University Hospital Heidelberg , Germany
| | - Anita Pathil
- d Department of Internal Medicine IV, Gastroenterology and Hepatology , University Hospital Heidelberg , Germany
| | - Arne Warth
- a Institute of Pathology, University Hospital Heidelberg , Germany
| | | | - Wilko Weichert
- e Technical University of Munich, University Hospital, Institute for General Pathology and Pathological Anatomy , Germany
| | - Marion Bähr
- b Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Rainer Will
- f Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | | | - Christoph Plass
- b Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Dieter Weichenhan
- b Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
208
|
Hu LS, Ning S, Eschbacher JM, Baxter LC, Gaw N, Ranjbar S, Plasencia J, Dueck AC, Peng S, Smith KA, Nakaji P, Karis JP, Quarles CC, Wu T, Loftus JC, Jenkins RB, Sicotte H, Kollmeyer TM, O'Neill BP, Elmquist W, Hoxworth JM, Frakes D, Sarkaria J, Swanson KR, Tran NL, Li J, Mitchell JR. Radiogenomics to characterize regional genetic heterogeneity in glioblastoma. Neuro Oncol 2016; 19:128-137. [PMID: 27502248 DOI: 10.1093/neuonc/now135] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) exhibits profound intratumoral genetic heterogeneity. Each tumor comprises multiple genetically distinct clonal populations with different therapeutic sensitivities. This has implications for targeted therapy and genetically informed paradigms. Contrast-enhanced (CE)-MRI and conventional sampling techniques have failed to resolve this heterogeneity, particularly for nonenhancing tumor populations. This study explores the feasibility of using multiparametric MRI and texture analysis to characterize regional genetic heterogeneity throughout MRI-enhancing and nonenhancing tumor segments. METHODS We collected multiple image-guided biopsies from primary GBM patients throughout regions of enhancement (ENH) and nonenhancing parenchyma (so called brain-around-tumor, [BAT]). For each biopsy, we analyzed DNA copy number variants for core GBM driver genes reported by The Cancer Genome Atlas. We co-registered biopsy locations with MRI and texture maps to correlate regional genetic status with spatially matched imaging measurements. We also built multivariate predictive decision-tree models for each GBM driver gene and validated accuracies using leave-one-out-cross-validation (LOOCV). RESULTS We collected 48 biopsies (13 tumors) and identified significant imaging correlations (univariate analysis) for 6 driver genes: EGFR, PDGFRA, PTEN, CDKN2A, RB1, and TP53. Predictive model accuracies (on LOOCV) varied by driver gene of interest. Highest accuracies were observed for PDGFRA (77.1%), EGFR (75%), CDKN2A (87.5%), and RB1 (87.5%), while lowest accuracy was observed in TP53 (37.5%). Models for 4 driver genes (EGFR, RB1, CDKN2A, and PTEN) showed higher accuracy in BAT samples (n = 16) compared with those from ENH segments (n = 32). CONCLUSION MRI and texture analysis can help characterize regional genetic heterogeneity, which offers potential diagnostic value under the paradigm of individualized oncology.
Collapse
Affiliation(s)
- Leland S Hu
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Shuluo Ning
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Jennifer M Eschbacher
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Leslie C Baxter
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Nathan Gaw
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Sara Ranjbar
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Jonathan Plasencia
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Amylou C Dueck
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Sen Peng
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Kris A Smith
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Peter Nakaji
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - John P Karis
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - C Chad Quarles
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Teresa Wu
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Joseph C Loftus
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Robert B Jenkins
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Hugues Sicotte
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Thomas M Kollmeyer
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Brian P O'Neill
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - William Elmquist
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Joseph M Hoxworth
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - David Frakes
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Jann Sarkaria
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Kristin R Swanson
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Nhan L Tran
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - Jing Li
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| | - J Ross Mitchell
- Department of Radiology, Mayo Clinic, Phoenix, Arizona (L.S.H., T.W., J.M.H.); Department of Biostatistics, Mayo Clinic, Phoenix, Arizona (A.C.D.); Department of Research, Mayo Clinic, Arizona (J.R.M., K.S.); Department of Neurosurgery, Mayo Clinic, Phoenix, Arizona (K.R.S.); Department of Cancer and Cell Biology, Mayo Clinic, Scottsdale, Arizona (J.C.L.); Department of Pathology, Mayo Clinic, Rochester, Minnesota (R.B.J., T.M.K.); Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota (H.S.); Department of Neuro-oncology, Mayo Clinic, Rochester, Minnesota (B.P.O.); Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (J.S.); Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota (W.E.); Department of Cancer and Cell Biology, Translational Genomics Research Institute, Phoenix, Arizona (S.P., N.L.T.); School of Computing, Informatics and Decision Systems Engineering, Arizona State University, Tempe, Arizona (J.L., T.W., S.N., N.G.); Department of Biomedical Informatics, Arizona State University, Tempe, Arizona (S.R.); School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona (J.P., D.F.); Department of Pathology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (J.M.E.); Department of Neurosurgery, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (K.A.S., P.N.); Department of Radiology, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (L.C.B., J.P. K., L.S.H.); Department of Imaging Research, Barrow Neurological Institute - St. Joseph's Hospital and Medical Center, Phoenix, Arizona (C.C.Q.)
| |
Collapse
|
209
|
Lee CH, Wang HE, Seo SY, Kim SH, Kim IH, Kim SW, Lee ST, Kim DG, Han MK, Lee SO. Cancer related gene alterations can be detected with next-generation sequencing analysis of bile in diffusely infiltrating type cholangiocarcinoma. Exp Mol Pathol 2016; 101:150-6. [PMID: 27460275 DOI: 10.1016/j.yexmp.2016.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/25/2016] [Accepted: 07/22/2016] [Indexed: 01/30/2023]
Abstract
Genome-wide association study in diffusely infiltrating type cholangiocarcinoma (CC) can be limited due to the difficulty of obtaining tumor tissue. We aimed to evaluate the genomic alterations of diffusely infiltrating type CC using next-generation sequencing (NGS) of bile and to compare the variations with those of mass-forming type CC. A total of 24 bile samples obtained during endoscopic retrograde cholangiopancreatography (ERCP) and 17 surgically obtained tumor tissue samples were evaluated. Buffy coat and normal tissue samples were used as controls for a somatic mutation analysis. After extraction of genomic DNA, NGS analysis was performed for 48 cancer related genes. There were 27 men and 14 women with a mean age of 65.0±11.8years. The amount of extracted genomic DNA from 3cm(3) of bile was 66.0±84.7μg and revealed a high depth of sequencing coverage. All of the patients had genomic variations, with an average number of 19.4±2.8 and 22.3±3.3 alterations per patient from the bile and tumor tissue, respectively. After filtering process, damaging SNPs (8 sites for each type of CC) were predicted by analyzing tools, and their target genes showed relevant differences between the diffusely infiltrating and mass-forming type CC. Finally, in somatic mutation analysis, tumor-normal paired 14 tissue and 6 bile samples were analyzed, genomic alterations of EGFR, FGFR1, ABL1, PIK3CA, and CDKN2A gene were seen in the diffusely infiltrating type CC, and TP53, KRAS, APC, GNA11, ERBB4, ATM, SMAD4, BRAF, and IDH1 were altered in the mass-forming type CC group. STK11, GNAQ, RB1, KDR, and SMO genes were revealed in both groups. The NGS analysis was feasible with bile sample and diffusely infiltrating type CC revealed genetic differences compared with mass-forming type CC. Genome-wide association study could be performed using bile sample in the patients with CC undergoing ERCP and a different genetic approach for accurate diagnosis, pathogenesis study, and targeted therapy will be needed in diffusely infiltrating type CC.
Collapse
Affiliation(s)
- Chang Hun Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Hong En Wang
- Department of Medical Science, The Graduate School, Chonbuk National University, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seung Young Seo
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Seong Hun Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - In Hee Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Sang Wook Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Soo Teik Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Dae Ghon Kim
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Myung Kwan Han
- Department of Microbiology, Chonbuk National University Medical School, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea.
| | - Seung Ok Lee
- Division of Gastroenterology, Department of Internal Medicine, Chonbuk National University Medical School and Hospital, Jeonju, Republic of Korea; Biomedical Research Institute, Chonbuk National University Hospital, Jeonju, Republic of Korea; Research Institute of Clinical Medicine, Chonbuk National University, Jeonju, Republic of Korea.
| |
Collapse
|
210
|
Wang J, Zhang K, Wang J, Wu X, Liu X, Li B, Zhu Y, Yu Y, Cheng Q, Hu Z, Guo C, Hu S, Mu B, Tsai CH, Li J, Smith L, Yang L, Liu Q, Chu P, Chang V, Zhang B, Wu M, Jiang X, Yen Y. Underexpression of LKB1 tumor suppressor is associated with enhanced Wnt signaling and malignant characteristics of human intrahepatic cholangiocarcinoma. Oncotarget 2016; 6:18905-20. [PMID: 26056085 PMCID: PMC4662463 DOI: 10.18632/oncotarget.4305] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/13/2015] [Indexed: 11/25/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a rare and highly aggressive malignancy. In this study, we identified the presence of gene deletion and missense mutation leading to inactivation or underexpression of liver kinase B1 (LKB1) tumor suppressor and excluded the involvement of LKB1 gene hypermethylation in ICC tissues. Immunohistochemical analysis showed that LKB1 was underexpressed in a portion of 326 ICC tissues compared to their adjacent normal tissues. By statistical analysis underexpression of LKB1 in ICC tissues significantly correlated with poor survival and malignant disease characteristics in ICC patients. Moreover, we showed that knockdown of LKB1 significantly enhanced growth, migration, and invasion of three LKB1-competent ICC cell lines. Global transcriptional profiling analysis identified multiple malignancy-promoting genes, such as HIF-1α, CD24, Talin1, Vinculin, Wnt5, and signaling pathways including Hedgehog, Wnt/β-catenin, and cell adhesion as novel targets of LKB1 underexpression in ICC cells. Furthermore, knockdown of LKB1 gene expression dramatically enhanced Wnt/β-catenin signaling in ICC cells, while an inverse correlation between LKB1 and nuclear β-catenin was observed in ICC tissues. Our findings suggest a novel mechanism for ICC carcinogenesis in which LKB1 underexpression enhances multiple signaling pathways including Wnt/β-catenin to promote disease progression.
Collapse
Affiliation(s)
- Jinghan Wang
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China.,Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Keqiang Zhang
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Jinhui Wang
- The Integrative Genomics Core lab of Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Xiwei Wu
- The Integrative Genomics Core lab of Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Xiyong Liu
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Bin Li
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Yan Zhu
- Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Yong Yu
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Qingbao Cheng
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Zhenli Hu
- Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Chao Guo
- The Integrative Genomics Core lab of Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Shuya Hu
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Bing Mu
- The Integrative Genomics Core lab of Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, School of Medicine, China Medical University, Taichung, Taiwan
| | - Jie Li
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Lynne Smith
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Lu Yang
- The Integrative Genomics Core lab of Department of Molecular Medicine, City of Hope National Medical Center, Duarte, California, USA
| | - Qi Liu
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center; Duarte, California, USA
| | - Vincent Chang
- Program for Translation Medicine, Taipei Medical University, Taipei, Taiwan
| | - Baihe Zhang
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Mengchao Wu
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoqing Jiang
- The First Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, The Second Military Medical University, Shanghai, China
| | - Yun Yen
- Department of Molecular Pharmacology, City of Hope National Medical Center, Duarte, California, USA.,PhD Program of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
211
|
Rosenquist TA, Grollman AP. Mutational signature of aristolochic acid: Clue to the recognition of a global disease. DNA Repair (Amst) 2016; 44:205-211. [PMID: 27237586 DOI: 10.1016/j.dnarep.2016.05.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mutational signatures associated with specific forms of DNA damage have been identified in several forms of human cancer. Such signatures provide information regarding mechanisms of tumor induction which, in turn, can reduce exposure to carcinogens by shaping public health policy. Using a molecular epidemiologic approach that takes advantage of recent advances in genome sequencing while applying sensitive and specific analytical methods to characterize DNA damage, it has become increasingly possible to establish causative linkages between certain environmental mutagens and disease risk. In this perspective, we use aristolochic acid, a human carcinogen and nephrotoxin found in Aristolochia herbs, to illustrate the power and effectiveness of this multidisciplinary approach. The genome-wide mutational signature for this toxin, detected initially in cancers of the upper urinary tract, has subsequently been associated with cancers of the liver and kidney. These findings have significant implications for global public health, especially in China, where millions of individuals have used Aristolochia herbal remedies as part of traditional Chinese medicine and, thus, are at risk of developing aristolochic acid nephropathy and/or upper urinary tract carcinomas. The studies reported here set the stage for research into prevention and early detection, both of which will be required to manage a potentially devastating global disease.
Collapse
Affiliation(s)
- Thomas A Rosenquist
- Stony Brook University School of Medicine, Department of Pharmacological Sciences, Laboratory of Chemical Biology, Stony Brook, NY, 11794, United States
| | - Arthur P Grollman
- Stony Brook University School of Medicine, Department of Pharmacological Sciences, Laboratory of Chemical Biology, Stony Brook, NY, 11794, United States.
| |
Collapse
|
212
|
Banales JM, Cardinale V, Carpino G, Marzioni M, Andersen JB, Invernizzi P, Lind GE, Folseraas T, Forbes SJ, Fouassier L, Geier A, Calvisi DF, Mertens JC, Trauner M, Benedetti A, Maroni L, Vaquero J, Macias RIR, Raggi C, Perugorria MJ, Gaudio E, Boberg KM, Marin JJG, Alvaro D. Expert consensus document: Cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat Rev Gastroenterol Hepatol 2016; 13:261-80. [PMID: 27095655 DOI: 10.1038/nrgastro.2016.51] [Citation(s) in RCA: 892] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of malignancies with features of biliary tract differentiation. CCA is the second most common primary liver tumour and the incidence is increasing worldwide. CCA has high mortality owing to its aggressiveness, late diagnosis and refractory nature. In May 2015, the "European Network for the Study of Cholangiocarcinoma" (ENS-CCA: www.enscca.org or www.cholangiocarcinoma.eu) was created to promote and boost international research collaboration on the study of CCA at basic, translational and clinical level. In this Consensus Statement, we aim to provide valuable information on classifications, pathological features, risk factors, cells of origin, genetic and epigenetic modifications and current therapies available for this cancer. Moreover, future directions on basic and clinical investigations and plans for the ENS-CCA are highlighted.
Collapse
Affiliation(s)
- Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis 6, 00135, Rome, Italy
| | - Marco Marzioni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Jesper B Andersen
- Biotech Research and Innovation Centre, University of Copenhagen, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Pietro Invernizzi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
- Program for Autoimmune Liver Diseases, International Center for Digestive Health, Department of Medicine and Surgery, University of Milan-Bicocca, Via Cadore 48, 20900, Monza, Italy
| | - Guro E Lind
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310, Oslo, Norway
| | - Trine Folseraas
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Stuart J Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 49 Little France Crescent, EH16 4SB, Edinburgh, United Kingdom
| | - Laura Fouassier
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Andreas Geier
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstrasse 6, D-97080, Würzburg, Germany
| | - Diego F Calvisi
- Institute of Pathology, Universitätsmedizin Greifswald, Friedrich-Löffler-Strasse 23e, 17489, Greifswald, Germany
| | - Joachim C Mertens
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, 8091, Zürich, Switzerland
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, A-1090, Vienna, Austria
| | - Antonio Benedetti
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Luca Maroni
- Department of Clinic and Molecular Sciences, Polytechnic University of Marche, Via Tronto 10, 60020, Ancona, Italy
| | - Javier Vaquero
- INSERM UMR S938, Centre de Recherche Saint-Antoine, 184 rue du Faubourg Saint-Antoine, 75571, Paris cedex 12, Fondation ARC, 9 rue Guy Môquet 94803 Villejuif, France
| | - Rocio I R Macias
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Chiara Raggi
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital, Ikerbasque, CIBERehd, Paseo del Dr. Begiristain s/n, E-20014, San Sebastian, Spain
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Via Alfonso Borelli 50, 00161, Rome, Italy
| | - Kirsten M Boberg
- Department of Transplantation Medicine, Division of Cancer Medicine, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Pb. 4950 Nydalen, N-0424, Oslo, Norway
| | - Jose J G Marin
- Department of Physiology and Pharmacology, Experimental Hepatology and Drug Targeting (HEVEFARM), Campus Miguel de Unamuno, E.I.D. S-09, University of Salamanca, IBSAL, CIBERehd, 37007, Salamanca, Spain
| | - Domenico Alvaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| |
Collapse
|
213
|
Hallinan N, Finn S, Cuffe S, Rafee S, O’Byrne K, Gately K. Targeting the fibroblast growth factor receptor family in cancer. Cancer Treat Rev 2016; 46:51-62. [DOI: 10.1016/j.ctrv.2016.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 02/08/2023]
|
214
|
Brandi G, Farioli A, Astolfi A, Biasco G, Tavolari S. Genetic heterogeneity in cholangiocarcinoma: a major challenge for targeted therapies. Oncotarget 2016; 6:14744-53. [PMID: 26142706 PMCID: PMC4558112 DOI: 10.18632/oncotarget.4539] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/11/2015] [Indexed: 02/07/2023] Open
Abstract
Cholangiocarcinoma (CC) encompasses a group of related but distinct malignancies whose lack of a stereotyped genetic signature makes challenging the identification of genomic landscape and the development of effective targeted therapies. Accumulated evidences strongly suggest that the remarkable genetic heterogeneity of CC may be the result of a complex interplay among different causative factors, some shared by most human cancers while others typical of this malignancy. Currently, considerable efforts are ongoing worldwide for the genetic characterization of CC, also using advanced technologies such as next-generation sequencing (NGS). Undoubtedly this technology could offer an unique opportunity to broaden our understanding on CC molecular pathogenesis. Despite this great potential, however, the high complexity in terms of factors potentially contributing to genetic variability in CC calls for a more cautionary application of NGS to this malignancy, in order to avoid possible biases and criticisms in the identification of candidate actionable targets. This approach is further justified by the urgent need to develop effective targeted therapies in this disease. A multidisciplinary approach integrating genomic, functional and clinical studies is therefore mandatory to translate the results obtained by NGS into effective targeted therapies for this orphan disease.
Collapse
Affiliation(s)
- Giovanni Brandi
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,"G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), University of Bologna, Bologna, Italy.,GICO- Italian Group of Cholangiocarcinoma, Italy
| | - Andrea Farioli
- Department of Medical and Surgical Sciences, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Annalisa Astolfi
- "G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), University of Bologna, Bologna, Italy
| | - Guido Biasco
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,"G. Prodi" Interdepartmental Center for Cancer Research (C.I.R.C.), University of Bologna, Bologna, Italy
| | - Simona Tavolari
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, Bologna, Italy.,Center for Applied Biomedical Research (C.R.B.A.), S. Orsola- Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
215
|
Ilyas SI, Yamada D, Hirsova P, Bronk SF, Werneburg NW, Krishnan A, Salim W, Zhang L, Trushina E, Truty MJ, Gores GJ. A Hippo and Fibroblast Growth Factor Receptor Autocrine Pathway in Cholangiocarcinoma. J Biol Chem 2016; 291:8031-47. [PMID: 26826125 PMCID: PMC4825008 DOI: 10.1074/jbc.m115.698472] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Indexed: 12/15/2022] Open
Abstract
Herein, we have identified cross-talk between the Hippo and fibroblast growth factor receptor (FGFR) oncogenic signaling pathways in cholangiocarcinoma (CCA). Yes-associated protein (YAP) nuclear localization and up-regulation of canonical target genes was observed in CCA cell lines and a patient-derived xenograft (PDX). Expression of FGFR1, -2, and -4 was identified in human CCA cell lines, driven, in part, by YAP coactivation of TBX5. In turn, FGFR signaling in a cell line with minimal basal YAP expression induced its cellular protein expression and nuclear localization. Treatment of YAP-positive CCA cell lines with BGJ398, a pan-FGFR inhibitor, resulted in a decrease in YAP activation. FGFR activation of YAP appears to be driven largely by FGF5 activation of FGFR2, as siRNA silencing of this ligand or receptor, respectively, inhibited YAP nuclear localization. BGJ398 treatment of YAP-expressing cells induced cell death due to Mcl-1 depletion. In a YAP-associated mouse model of CCA, expression of FGFR 1, 2, and 4 was also significantly increased. Accordingly, BGJ398 treatment was tumor-suppressive in this model and in a YAP-positive PDX model. These preclinical data suggest not only that the YAP and Hippo signaling pathways culminate in an Mcl-1-regulated tumor survival pathway but also that nuclear YAP expression may be a biomarker to employ in FGFR-directed therapy.
Collapse
Affiliation(s)
- Sumera I Ilyas
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Daisaku Yamada
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Petra Hirsova
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Steven F Bronk
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Nathan W Werneburg
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Anuradha Krishnan
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Warda Salim
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905
| | - Liang Zhang
- the Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905
| | - Eugenia Trushina
- the Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905, the Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, and
| | - Mark J Truty
- the Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Gregory J Gores
- From the Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota 55905,
| |
Collapse
|
216
|
Byron SA, Van Keuren-Jensen KR, Engelthaler DM, Carpten JD, Craig DW. Translating RNA sequencing into clinical diagnostics: opportunities and challenges. Nat Rev Genet 2016; 17:257-71. [PMID: 26996076 PMCID: PMC7097555 DOI: 10.1038/nrg.2016.10] [Citation(s) in RCA: 452] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RNA-based measurements have the potential for application across diverse areas of human health, including disease diagnosis, prognosis and therapeutic selection. Current clinical applications include infectious diseases, cancer, transplant medicine and fetal monitoring. RNA sequencing (RNA-seq) allows for the detection of a wide variety of RNA species, including mRNA, non-coding RNA, pathogen RNA, chimeric gene fusions, transcript isoforms and splice variants, and provides the capability to quantify known, pre-defined RNA species and rare RNA transcript variants within a sample. In addition to differential expression and detection of novel transcripts, RNA-seq also supports the detection of mutations and germline variation for hundreds to thousands of expressed genetic variants, facilitating assessment of allele-specific expression of these variants. Circulating RNAs and small regulatory RNAs, such as microRNAs, are very stable. These RNA species are vigorously being tested for their potential as biomarkers. However, there are currently few agreed upon methods for isolation or quantitative measurements and a current lack of quality controls that can be used to test platform accuracy and sample preparation quality. Analytical, bioinformatic and regulatory challenges exist, and ongoing efforts toward the establishment of benchmark standards, assay optimization for clinical conditions and demonstration of assay reproducibility are required to expand the clinical utility of RNA-seq.
RNA sequencing (RNA-seq) is a powerful approach for comprehensive analyses of transcriptomes. This Review describes the widespread potential applications of RNA-seq in clinical medicine, such as detecting disease-associated mutations and gene expression disruptions, as well as characteristic non-coding RNAs, circulating extracellular RNAs or pathogen RNAs. The authors also highlight the challenges in adopting RNA-seq routinely into clinical practice. With the emergence of RNA sequencing (RNA-seq) technologies, RNA-based biomolecules hold expanded promise for their diagnostic, prognostic and therapeutic applicability in various diseases, including cancers and infectious diseases. Detection of gene fusions and differential expression of known disease-causing transcripts by RNA-seq represent some of the most immediate opportunities. However, it is the diversity of RNA species detected through RNA-seq that holds new promise for the multi-faceted clinical applicability of RNA-based measures, including the potential of extracellular RNAs as non-invasive diagnostic indicators of disease. Ongoing efforts towards the establishment of benchmark standards, assay optimization for clinical conditions and demonstration of assay reproducibility are required to expand the clinical utility of RNA-seq.
Collapse
Affiliation(s)
- Sara A Byron
- Center for Translational Innovation, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | | | - David M Engelthaler
- Pathogen Genomics Division, Translational Genomics Research Institute, Flagstaff, Arizona 86001, USA
| | - John D Carpten
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - David W Craig
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| |
Collapse
|
217
|
Comprehensive analysis of targetable oncogenic mutations in chinese cervical cancers. Oncotarget 2016; 6:4968-75. [PMID: 25669975 PMCID: PMC4467127 DOI: 10.18632/oncotarget.3212] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 12/27/2014] [Indexed: 11/25/2022] Open
Abstract
Mutations in 16 targetable oncogenic genes were examined using reverse transcription polymerase chain reaction (RT-PCR) and direct sequencing in 285 Chinese cervical cancers. Their clinicopathological relevance and prognostic significance was assessed. Ninety-two nonsynonymous somatic mutations were identified in 29.8% of the cancers. The mutation rates were as follows: PIK3CA (12.3%), KRAS (5.3%), HER2 (4.2%), FGFR3-TACC3 fusions (3.9%), PTEN (2.8%), FGFR2 (1.8%), FGFR3 (0.7%), NRAS (0.7%), HRAS (0.4%) and EGFR (0.4%). No mutations were detected in AKT1 or BRAF, and the fusions FGFR1-TACC1, EML4-ALK, CCDC6-RET and KIF5B-RET were not found in any of the cancers. RTK and RAS mutations were more common in non-squamous carcinomas than in squamous carcinomas (P=0.043 and P=0.042, respectively). RAS mutations were more common in young patients (<45 years) (13.7% vs. 7.7%, P=0.027). RTK mutations tended to be more common in young patients, whereas PIK3CA/PTEN/AKT mutations tended to be more common in old patients. RAS mutations were significantly associated with disease relapse. To our knowledge, this is the first comprehensive analysis of major targetable oncogenic mutations in a large cohort of cervical cancer cases. Our data reveal that a considerable proportion of patients with cervical cancers harbor known druggable mutations and might benefit from targeted therapy.
Collapse
|
218
|
Beadling C, Wald AI, Warrick A, Neff TL, Zhong S, Nikiforov YE, Corless CL, Nikiforova MN. A Multiplexed Amplicon Approach for Detecting Gene Fusions by Next-Generation Sequencing. J Mol Diagn 2016; 18:165-75. [DOI: 10.1016/j.jmoldx.2015.10.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/25/2015] [Accepted: 10/30/2015] [Indexed: 01/05/2023] Open
|
219
|
Xie D, Ren Z, Fan J, Gao Q. Genetic profiling of intrahepatic cholangiocarcinoma and its clinical implication in targeted therapy. Am J Cancer Res 2016; 6:577-586. [PMID: 27152236 PMCID: PMC4851838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 02/12/2016] [Indexed: 06/05/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a treatment-refractory primary liver cancer with an increasing incidence and mortality worldwide in recent years. Lack of a stereotyped genetic signature and limited understanding of genomic landscape make the development of effective targeted therapies challenging. Recent application of advanced technologies such as next-generation sequencing (NGS) has broadened our understanding of genetic heterogeneity in iCCA and many potentially actionable genetic alterations have been identified. This review explores the recent advances in defining genetic alterations in iCCAs, which may present potent therapeutic targets. Chromatin remodeling genes and genes encoding isocitrate dehydrogenase and tyrosine kinase receptors as well as their downstream effectors are among the most frequently altered genes. Clinical trials testing the effect of new targeted agents on iCCA patients, especially those with the above genetic markers are under way. However, the complex interplay of environmental and evolutionary factors contributing to the genetic variability in iCCA calls for a more cautionary use of NGS in tailoring targeted regimen to the patients. Next-generation functional testing may complement NGS to execute precision medicine in future.
Collapse
Affiliation(s)
- Diyang Xie
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai 200032, P. R. China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai 200032, P. R. China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai 200032, P. R. China
- Institute of Biomedical Sciences, Fudan UniversityShanghai 200032, P. R. China
| | - Qiang Gao
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan UniversityShanghai 200032, P. R. China
| |
Collapse
|
220
|
Jensen LH. Clinical aspects and perspectives of erlotinib in the treatment of patients with biliary tract cancer. Expert Opin Investig Drugs 2016; 25:359-65. [PMID: 26781267 DOI: 10.1517/13543784.2016.1142973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Patients with non-resectable biliary tract cancer have a poor prognosis even if treated with systemic chemotherapy. One hope for improving treatment is through molecular biology and the characterization of specific cancer driving alterations followed by the design of targeted drugs. The epidermal growth factor receptor system is upregulated in many cancers and can be targeted by the protein kinase inhibitor erlotinib. Erlotinib has demonstrated a clinically applicable effect in pancreatic and lung cancer Areas covered: In this review, the author presents the published clinical data about erlotinib in biliary tract cancer. The data is interpreted with respect to its clinical value and in regards to its future development. EXPERT OPINION Erlotinib has low activity as a monotherapy, but has shown synergistic effects when combined with bevacizumab. The only phase III trial with erlotinib was negative, but suggested improved progression free survival in cholangiocarcinoma patients when added to gemcitabine and oxaliplatin. There is no clinical, radiological or molecular marker to guide therapy, but genomic profiling and basket or umbrella trials may be useful in identifying the subset of patients benefitting from erlotinib. Until this subgroup has been defined, erlotinib has no value to biliary tract cancer patients in the daily clinic.
Collapse
Affiliation(s)
- Lars Henrik Jensen
- a Department of Oncology , Vejle Hospital and University of Southern Denmark , Vejle , Denmark
| |
Collapse
|
221
|
Hovelson DH, McDaniel AS, Cani AK, Johnson B, Rhodes K, Williams PD, Bandla S, Bien G, Choppa P, Hyland F, Gottimukkala R, Liu G, Manivannan M, Schageman J, Ballesteros-Villagrana E, Grasso CS, Quist MJ, Yadati V, Amin A, Siddiqui J, Betz BL, Knudsen KE, Cooney KA, Feng FY, Roh MH, Nelson PS, Liu CJ, Beer DG, Wyngaard P, Chinnaiyan AM, Sadis S, Rhodes DR, Tomlins SA. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia 2016; 17:385-99. [PMID: 25925381 PMCID: PMC4415141 DOI: 10.1016/j.neo.2015.03.004] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022] Open
Abstract
Next-generation sequencing (NGS) has enabled genome-wide personalized oncology efforts at centers and companies with the specialty expertise and infrastructure required to identify and prioritize actionable variants. Such approaches are not scalable, preventing widespread adoption. Likewise, most targeted NGS approaches fail to assess key relevant genomic alteration classes. To address these challenges, we predefined the catalog of relevant solid tumor somatic genome variants (gain-of-function or loss-of-function mutations, high-level copy number alterations, and gene fusions) through comprehensive bioinformatics analysis of >700,000 samples. To detect these variants, we developed the Oncomine Comprehensive Panel (OCP), an integrative NGS-based assay [compatible with < 20 ng of DNA/RNA from formalin-fixed paraffin-embedded (FFPE) tissues], coupled with an informatics pipeline to specifically identify relevant predefined variants and created a knowledge base of related potential treatments, current practice guidelines, and open clinical trials. We validated OCP using molecular standards and more than 300 FFPE tumor samples, achieving >95% accuracy for KRAS, epidermal growth factor receptor, and BRAF mutation detection as well as for ALK and TMPRSS2:ERG gene fusions. Associating positive variants with potential targeted treatments demonstrated that 6% to 42% of profiled samples (depending on cancer type) harbored alterations beyond routine molecular testing that were associated with approved or guideline-referenced therapies. As a translational research tool, OCP identified adaptive CTNNB1 amplifications/mutations in treated prostate cancers. Through predefining somatic variants in solid tumors and compiling associated potential treatment strategies, OCP represents a simplified, broadly applicable targeted NGS system with the potential to advance precision oncology efforts.
Collapse
Affiliation(s)
- Daniel H Hovelson
- Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew S McDaniel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andi K Cani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kate Rhodes
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | | | - Paul Choppa
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | - Guoying Liu
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | | | - Catherine S Grasso
- Department of Pathology, Oregon Health and Sciences University, Portland, OR, USA
| | - Michael J Quist
- Department of Pathology, Oregon Health and Sciences University, Portland, OR, USA
| | - Venkata Yadati
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anmol Amin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Bryan L Betz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA; Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA; Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen A Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael H Roh
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chia-Jen Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David G Beer
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Seth Sadis
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | - Daniel R Rhodes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Thermo Fisher Scientific, Ann Arbor, MI, USA
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
222
|
Abstract
Understanding the molecular landscape of cancer has facilitated the development of diagnostic, prognostic, and predictive biomarkers for clinical oncology. Developments in next-generation DNA sequencing technologies have increased the speed and reduced the cost of sequencing the nucleic acids of cancer cells. This has unlocked opportunities to characterize the genomic and transcriptomic landscapes of cancer for basic science research through projects like The Cancer Genome Atlas. The cancer genome includes DNA-based alterations, such as point mutations or gene duplications. The cancer transcriptome involves RNA-based alterations, including changes in messenger RNAs. Together, the genome and transcriptome can provide a comprehensive view of an individual patient's cancer that is beginning to impact real-time clinical decision-making. The authors discuss several opportunities for translating this basic science knowledge into clinical practice, including a molecular classification of cancer, heritable risk of cancer, eligibility for targeted therapies, and the development of innovative, genomic-based clinical trials. In this review, key applications and new directions are outlined for translating the cancer genome and transcriptome into patient care in the clinic.
Collapse
Affiliation(s)
- Sameek Roychowdhury
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210 USA
- Department of Pharmacology, The Ohio State University, Columbus, Ohio 43210 USA
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
- Center for Computational Medicine and Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
223
|
Fornaro L, Vivaldi C, Cereda S, Leone F, Aprile G, Lonardi S, Silvestris N, Santini D, Milella M, Caparello C, Musettini G, Pasquini G, Falcone A, Brandi G, Sperduti I, Vasile E. Second-line chemotherapy in advanced biliary cancer progressed to first-line platinum-gemcitabine combination: a multicenter survey and pooled analysis with published data. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:156. [PMID: 26693938 PMCID: PMC4689003 DOI: 10.1186/s13046-015-0267-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/07/2015] [Indexed: 02/09/2023]
Abstract
Background After progression to a standard first-line platinum and gemcitabine combination (GP), there is no established second-line therapy for patients with advanced biliary tract cancers (aBTC). Indeed, literature data suggest limited activity of most second-line agents evaluated so far. Methods We collected a large retrospective series of aBTC patients treated with second-line chemotherapy after progression to a first-line GP regimen at different Italian institutions. We then pooled the data with those reported in previous studies, which were identified with a Medline search and the on-line abstract datasets of major international oncology meetings. Results A total of 174 patients were included in the multicenter survey: response rate (RR) with second-line chemotherapy was low (3.4 %), with median PFS and OS of 3.0 months and 6.6 months, respectively. At multivariate analysis, preserved performance status, low CA19.9 levels and absence of distant metastases were favorable prognostic factors. Data from other five presented or published series were identified, for a total of 499 patients included in the pooled analysis. The results confirmed marginal activity of second-line chemotherapy (RR: 10.2 %), with limited efficacy in unselected patient populations (median PFS: 3.1 months; median OS: 6.3 months). Conclusions The current analysis highlights the limited value of second-line chemotherapy after a first-line GP combination in aBTC. While waiting for effective biologic agents in this setting, ongoing randomized trials will identify the optimal second-line chemotherapy regimen and validate prognostic factors for individual patient management.
Collapse
Affiliation(s)
- Lorenzo Fornaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Caterina Vivaldi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Stefano Cereda
- Department of Medical Oncology, San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Francesco Leone
- Unit of Medical Oncology, Institute for Cancer Research and Treatment IRCCS, Strada provinciale 142, 10060, Candiolo, Italy.
| | - Giuseppe Aprile
- Department of Oncology, University and General Hospital, P.le S. Maria della Misericordia 15, 33100, Udine, Italy.
| | - Sara Lonardi
- Unit of Medical Oncology 1, Istituto Oncologico Veneto, IRCCS, Via Gattamelata 64, 35138, Padova, Italy.
| | - Nicola Silvestris
- Unit of Medical Oncology, National Cancer Institute Giovanni Paolo II, V.le Orazio Flacco 65, 70124, Bari, Italy.
| | - Daniele Santini
- Department of Medical Oncology, University Campus Bio-Medico, via Alvaro del Portillo 21, 00128, Rome, Italy.
| | - Michele Milella
- Medical Oncology A, Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| | - Chiara Caparello
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Gianna Musettini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Giulia Pasquini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Alfredo Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Giovanni Brandi
- Unit of Medical Oncology, Sant'Orsola Malpighi Hospital, University of Bologna, Via Albertoni 15, 40138, Bologna, Italy.
| | - Isabella Sperduti
- Biostatistical Unit, Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| | - Enrico Vasile
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | | |
Collapse
|
224
|
Kasaian K, Wiseman SM, Walker BA, Schein JE, Zhao Y, Hirst M, Moore RA, Mungall AJ, Marra MA, Jones SJM. The genomic and transcriptomic landscape of anaplastic thyroid cancer: implications for therapy. BMC Cancer 2015; 15:984. [PMID: 26680454 PMCID: PMC4683857 DOI: 10.1186/s12885-015-1955-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/25/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Anaplastic thyroid carcinoma is the most undifferentiated form of thyroid cancer and one of the deadliest of all adult solid malignancies. Here we report the first genomic and transcriptomic profile of anaplastic thyroid cancer including those of several unique cell lines and outline novel potential drivers of malignancy and targets of therapy. METHODS We describe whole genomic and transcriptomic profiles of 1 primary anaplastic thyroid tumor and 3 authenticated cell lines. Those profiles augmented by the transcriptomes of 4 additional and unique cell lines were compared to 58 pairs of papillary thyroid carcinoma and matched normal tissue transcriptomes from The Cancer Genome Atlas study. RESULTS The most prevalent mutations were those of TP53 and BRAF; repeated alterations of the epigenetic machinery such as frame-shift deletions of HDAC10 and EP300, loss of SMARCA2 and fusions of MECP2, BCL11A and SS18 were observed. Sequence data displayed aneuploidy and large regions of copy loss and gain in all genomes. Common regions of gain were however evident encompassing chromosomes 5p and 20q. We found novel anaplastic gene fusions including MKRN1-BRAF, FGFR2-OGDH and SS18-SLC5A11, all expressed in-frame fusions involving a known proto-oncogene. Comparison of the anaplastic thyroid cancer expression datasets with the papillary thyroid cancer and normal thyroid tissue transcriptomes suggested several known drug targets such as FGFRs, VEGFRs, KIT and RET to have lower expression levels in anaplastic specimens compared with both papillary thyroid cancers and normal tissues, confirming the observed lack of response to therapies targeting these pathways. Further integrative data analysis identified the mTOR signaling pathway as a potential therapeutic target in this disease. CONCLUSIONS Anaplastic thyroid carcinoma possessed heterogeneous and unique profiles revealing the significance of detailed molecular profiling of individual tumors and the treatment of each as a unique entity; the cell line sequence data promises to facilitate the more accurate and intentional drug screening studies for anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Sam M Wiseman
- Department of Surgery, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada.
| | - Blair A Walker
- Department of Pathology and Laboratory Medicine, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada.
| | - Jacqueline E Schein
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Yongjun Zhao
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Martin Hirst
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada.
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada. .,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada. .,, 570 West 7th Ave, Vancouver, British Columbia, V5Z 4S6, Canada.
| |
Collapse
|
225
|
Maggi E, Montagna C. AACR precision medicine series: Highlights of the integrating clinical genomics and cancer therapy meeting. Mutat Res 2015; 782:44-51. [PMID: 26554403 DOI: 10.1016/j.mrfmmm.2015.10.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 10/23/2015] [Accepted: 10/28/2015] [Indexed: 06/05/2023]
Abstract
The American Association for Cancer Research (AACR) Precision Medicine Series "Integrating Clinical Genomics and Cancer Therapy" took place June 13-16, 2015 in Salt Lake City, Utah. The conference was co-chaired by Charles L. Sawyers form Memorial Sloan Kettering Cancer Center in New York, Elaine R. Mardis form Washington University School of Medicine in St. Louis, and Arul M. Chinnaiyan from University of Michigan in Ann Arbor. About 500 clinicians, basic science investigators, bioinformaticians, and postdoctoral fellows joined together to discuss the current state of Clinical Genomics and the advances and challenges of integrating Next Generation Sequencing (NGS) technologies into clinical practice. The plenary sessions and panel discussions covered current platforms and sequencing approaches adopted for NGS assays of cancer genome at several national and international institutions, different approaches used to map and classify targetable sequence variants, and how information acquired with the sequencing of the cancer genome is used to guide treatment options. While challenges still exist from a technological perspective, it emerged that there exists considerable need for the development of tools to aid the identification of the therapy most suitable based on the mutational profile of the somatic cancer genome. The process to match patients to ongoing clinical trials is still complex. In addition, the need for centralized data repositories, preferably linked to well annotated clinical records, that aid sharing of sequencing information is central to begin understanding the contribution of variants of unknown significance to tumor etiology and response to therapy. Here we summarize the highlights of this stimulating four-day conference with a major emphasis on the open problems that the clinical genomics community is currently facing and the tools most needed for advancing this field.
Collapse
Affiliation(s)
- Elaine Maggi
- Department of Genetics, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA
| | - Cristina Montagna
- Department of Genetics, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA; Department Pathology, Albert Einstein College of Medicine, Yeshiva University, Bronx, NY 10461, USA.
| |
Collapse
|
226
|
Marquardt JU, Andersen JB, Thorgeirsson SS. Functional and genetic deconstruction of the cellular origin in liver cancer. Nat Rev Cancer 2015; 15:653-67. [PMID: 26493646 DOI: 10.1038/nrc4017] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During the past decade, research on primary liver cancers has particularly highlighted the uncommon plasticity of differentiated parenchymal liver cells (that is, hepatocytes and cholangiocytes (also known as biliary epithelial cells)), the role of liver progenitor cells in malignant transformation, the importance of the tumour microenvironment and the molecular complexity of liver tumours. Whereas other reviews have focused on the landscape of genetic alterations that promote development and progression of primary liver cancers and the role of the tumour microenvironment, the crucial importance of the cellular origin of liver cancer has been much less explored. Therefore, in this Review, we emphasize the importance and complexity of the cellular origin in tumour initiation and progression, and attempt to integrate this aspect with recent discoveries in tumour genomics and the contribution of the disrupted hepatic microenvironment to liver carcinogenesis.
Collapse
Affiliation(s)
- Jens U Marquardt
- Department of Medicine I, Johannes Gutenberg University, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen N, Denmark
| | - Snorri S Thorgeirsson
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
227
|
Zhang YA, Shen XZ, Zhu JM, Liu TT. Extensive Metastatic Cholangiocarcinoma Associated With IgG4-Related Sclerosing Cholangitis Misdiagnosed as Isolated IgG4-Related Sclerosing Cholangitis: A Case Report and Literature Review. Medicine (Baltimore) 2015; 94:e2052. [PMID: 26559312 PMCID: PMC4912306 DOI: 10.1097/md.0000000000002052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As cholangiographic features of IgG4-related sclerosing cholangitis (IgG4-SC) resemble those of cholangiocarcinoma, it is highly confusing between the 2 conditions on the basis of cholangiographic findings. This study presents a case of extensive metastatic cholangiocarcinoma with IgG4-SC misdiagnosed as isolated IgG4-SC, and reviews recent studies of the 2 diseases.A 56-year-old man with no family history of malignant tumors or liver diseases presented with recurrent mild abdominal pain and distention for 3 months. Magnetic resonance cholangiopancreatography showed a 3.7 cm nodular lesion with unclear boundary in segment VI of the liver. Serum IgG4 and CA19-9 were slightly elevated. Histopathological examination was consistent with the consensus statement on the pathology of IgG4-SC. IgG4-SC was thus considered. Due to his mild symptoms, glucocorticoid was not given at first. However, 3 months after his first admission, he had more severe abdominal pain and further elevated serum CA19-9. Actually he was found suffering from extensive metastatic cholangiocarcinoma with IgG4-SC by exploratory laparotomy.The present case serves as a reminder that extensive metastatic cholangiocarcinoma with or without IgG4-SC may be misdiagnosed as an isolated IgG4-SC case if one relies solely on elevated serum and tissue IgG4 levels. We emphasize on the importance of repeated core needle biopsy or exploratory laparoscopy/laparotomy before immunosuppressive drugs are given, and on follow-up of imaging findings and serum CA19-9 once immunosuppressive therapy is started.
Collapse
Affiliation(s)
- Yi-An Zhang
- From Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, China (YAZ, XZS, JMZ, TTL); Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, China (XZS); and Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, Shanghai, China (XZS)
| | | | | | | |
Collapse
|
228
|
Juntermanns B, Sydor S, Kaiser GM, Jaradat D, Mertens JC, Sotiropoulos GC, Swoboda S, Neuhaus JP, Meng W, Mathé Z, Baba HA, Canbay A, Paul A, Fingas CD. Polo-like kinase 3 is associated with improved overall survival in cholangiocarcinoma. Liver Int 2015; 35:2448-57. [PMID: 25818805 DOI: 10.1111/liv.12839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/25/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Cholangiocarcinomas (CCA) paradoxically express the death ligand TRAIL and thus, are dependent on effective survival signals to circumvent apoptosis. Hedgehog signalling exerts major survival signals in CCA by regulating serine/threonine kinase polo-like kinase (PLK)2. We here aimed to examine the role of PLK1/2/3 expression for CCA tumour biology. METHODS We employed CCA samples from 73 patients and human HUCCT-1/Mz-CHA1/KMCH-1 CCA cells. Immunohistochemistry for PLK1/2/3 was performed using tissue microarrays from representative tumour areas. RESULTS PLK1/2/3-immunoreactive cancer cells were present in most of the CCA samples. However, only PLK1 and especially PLK3 were expressed in higher amounts within CCA cells as compared to normal liver. Given that fibroblast growth factor (FGF) can induce PLK3 expression and also is present in CCA, we examined the effect of FGF on PLK3 in vitro. Indeed, rhFGF rapidly increased PLK3 mRNA expression all three CCA cell lines giving an explanation for the abundant PLK3 presence in the tissue samples. Clinicopathologically, PLK3 expression was associated with decreased tumour cell migration and lymph/blood vessel infiltration whereas higher levels of PLK1 were correlated with larger tumour sizes. Moreover, strong PLK3 expression was associated with prolonged overall survival. CONCLUSIONS The results suggest that PLK3 predominantly is expressed in CCA cells and that high PLK3 expression correlates with prolonged overall survival. These observations might have implications for prognosis prediction of human CCA as well as the potential therapeutic use of polo-like kinase inhibitors (i.e., PLK1/2 specifity).
Collapse
Affiliation(s)
- Benjamin Juntermanns
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Svenja Sydor
- Division of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany
| | - Gernot M Kaiser
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Derar Jaradat
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Joachim C Mertens
- Division of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Georgios C Sotiropoulos
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Sandra Swoboda
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Jan P Neuhaus
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Wei Meng
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Zoltan Mathé
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, Essen, Germany
| | - Ali Canbay
- Division of Gastroenterology and Hepatology, University Hospital Essen, Essen, Germany
| | - Andreas Paul
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | - Christian D Fingas
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| |
Collapse
|
229
|
Ilyas SI, Eaton JE, Gores GJ. Primary Sclerosing Cholangitis as a Premalignant Biliary Tract Disease: Surveillance and Management. Clin Gastroenterol Hepatol 2015; 13:2152-65. [PMID: 26051390 PMCID: PMC4618039 DOI: 10.1016/j.cgh.2015.05.035] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/20/2015] [Accepted: 05/22/2015] [Indexed: 02/07/2023]
Abstract
Primary sclerosing cholangitis (PSC) is a premalignant biliary tract disease that confers a significant risk for the development of cholangiocarcinoma (CCA). The chronic biliary tract inflammation of PSC promotes pro-oncogenic processes such as cellular proliferation, induction of DNA damage, alterations of the extracellular matrix, and cholestasis. The diagnosis of malignancy in PSC can be challenging because inflammation-related changes in PSC may produce dominant biliary tract strictures mimicking CCA. Biomarkers such as detection of methylated genes in biliary specimens represent noninvasive techniques that may discriminate malignant biliary ductal changes from PSC strictures. However, conventional cytology and advanced cytologic techniques such as fluorescence in situ hybridization for polysomy remain the practice standard for diagnosing CCA in PSC. Curative treatment options of malignancy arising in PSC are limited. For a subset of patients selected by using stringent criteria, liver transplantation after neoadjuvant chemoradiation is a potential curative therapy. However, most patients have advanced malignancy at the time of diagnosis. Advances directed at identifying high-risk patients, early cancer detection, and development of chemopreventive strategies will be essential to better manage the cancer risk in this premalignant disease. A better understanding of dysplasia definition and especially its natural history is also needed in this disease. Herein, we review recent developments in our understanding of the risk factors, pathogenic mechanisms of PSC associated with CCA, as well as advances in early detection and therapies.
Collapse
Affiliation(s)
- Sumera I Ilyas
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - John E Eaton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
230
|
Anastasi S, Lamberti D, Alemà S, Segatto O. Regulation of the ErbB network by the MIG6 feedback loop in physiology, tumor suppression and responses to oncogene-targeted therapeutics. Semin Cell Dev Biol 2015; 50:115-24. [PMID: 26456277 DOI: 10.1016/j.semcdb.2015.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/02/2015] [Indexed: 01/08/2023]
Abstract
The ErbB signaling network instructs the execution of key cellular programs, such as cell survival, proliferation and motility, through the generation of robust signals of defined strength and duration. In contrast, unabated ErbB signaling disrupts tissue homeostasis and leads to cell transformation. Cells oppose the threat inherent in excessive ErbB activity through several mechanisms of negative feedback regulation. Inducible feedback inhibitors (IFIs) are expressed in the context of transcriptional responses triggered by ErbB signaling, thus being uniquely suited to regulate ErbB activity during the execution of complex cellular programs. This review focuses on MIG6, an IFI that restrains ErbB signaling by mediating ErbB kinase suppression and receptor down-regulation. We will review key issues in MIG6 function, regulation and tumor suppressor activity. Subsequently, the role for MIG6 loss in the pathogenesis of tumors driven by ErbB oncogenes as well as in the generation of cellular addiction to ErbB signaling will be discussed. We will conclude by analyzing feedback inhibition by MIG6 in the context of therapies directed against ErbB and non-ErbB oncogenes.
Collapse
Affiliation(s)
- Sergio Anastasi
- Laboratory of Cell Signaling, Regina Elena National Cancer Institute, via E. Chianesi, 53, 00144 Rome, Italy.
| | - Dante Lamberti
- Laboratory of Cell Signaling, Regina Elena National Cancer Institute, via E. Chianesi, 53, 00144 Rome, Italy.
| | - Stefano Alemà
- Institute of Cell Biology and Neurobiology, CNR, 00016 Monterotondo, Italy.
| | - Oreste Segatto
- Laboratory of Cell Signaling, Regina Elena National Cancer Institute, via E. Chianesi, 53, 00144 Rome, Italy.
| |
Collapse
|
231
|
Graham RP, Garcia JJ, Greipp PT, Barr Fritcher EG, Kipp BR, Torbenson MS. FGFR1andFGFR2in fibrolamellar carcinoma. Histopathology 2015; 68:686-92. [DOI: 10.1111/his.12799] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/05/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Rondell P Graham
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN USA
| | - Joaquin J Garcia
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN USA
| | - Patricia T Greipp
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN USA
| | | | - Benjamin R Kipp
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN USA
| | - Michael S Torbenson
- Department of Laboratory Medicine and Pathology; Mayo Clinic; Rochester MN USA
| |
Collapse
|
232
|
Moeini A, Sia D, Bardeesy N, Mazzaferro V, Llovet JM. Molecular Pathogenesis and Targeted Therapies for Intrahepatic Cholangiocarcinoma. Clin Cancer Res 2015; 22:291-300. [PMID: 26405193 DOI: 10.1158/1078-0432.ccr-14-3296] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/06/2015] [Indexed: 12/17/2022]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a molecularly heterogeneous hepatobiliary neoplasm with poor prognosis and limited therapeutic options. The incidence of this neoplasm is growing globally. One third of iCCA tumors are amenable to surgical resection, but most cases are diagnosed at advanced stages with chemotherapy as the only established standard of practice. No molecular therapies are currently available for the treatment of this neoplasm. The poor understanding of the biology of iCCA and the lack of known oncogenic addiction loops has hindered the development of effective targeted therapies. Studies with sophisticated animal models defined IDH mutation as the first gatekeeper in the carcinogenic process and led to the discovery of striking alternative cellular origins. RNA- and exome-sequencing technologies revealed the presence of recurrent novel fusion events (FGFR2 and ROS1 fusions) and somatic mutations in metabolic (IDH1/2) and chromatin-remodeling genes (ARID1A, BAP1). These latest advancements along with known mutations in KRAS/BRAF/EGFR and 11q13 high-level amplification have contributed to a better understanding of the landscape of molecular alterations in iCCA. More than 100 clinical trials testing molecular therapies alone or in combination with chemotherapy including iCCA patients have not reported conclusive clinical benefits. Recent discoveries have shown that up to 70% of iCCA patients harbor potential actionable alterations that are amenable to therapeutic targeting in early clinical trials. Thus, the first biomarker-driven trials are currently underway.
Collapse
Affiliation(s)
- Agrin Moeini
- Liver Cancer Translational Research Laboratory, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, CIBERehd, Universitat de Barcelona, Barcelona, Catalonia, Spain. Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Daniela Sia
- Liver Cancer Translational Research Laboratory, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, CIBERehd, Universitat de Barcelona, Barcelona, Catalonia, Spain. Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Gastrointestinal Surgery and Liver Transplantation Unit, Department of Surgery, National Cancer Institute IRCCS Foundation, Milan, Italy
| | - Nabeel Bardeesy
- Cancer Center, Center for Regenerative Medicine, and Department of Molecular Biology, Massachusetts General Hospital, Harvard University, Boston, Massachusetts
| | - Vincenzo Mazzaferro
- Gastrointestinal Surgery and Liver Transplantation Unit, Department of Surgery, National Cancer Institute IRCCS Foundation, Milan, Italy
| | - Josep M Llovet
- Liver Cancer Translational Research Laboratory, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, CIBERehd, Universitat de Barcelona, Barcelona, Catalonia, Spain. Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Institució Catalana de Recerca i Estudis Avançats, Barcelona, Catalonia, Spain.
| |
Collapse
|
233
|
An Omics Perspective on Molecular Biomarkers for Diagnosis, Prognosis, and Therapeutics of Cholangiocarcinoma. Int J Genomics 2015; 2015:179528. [PMID: 26421274 PMCID: PMC4572471 DOI: 10.1155/2015/179528] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/09/2015] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive biliary tract malignancy arising from the epithelial bile duct. The lack of early diagnostic biomarkers as well as therapeutic measures results in severe outcomes and poor prognosis. Thus, effective early diagnostic, prognostic, and therapeutic biomarkers are required to improve the prognosis and prolong survival rates in CCA patients. Recent advancement in omics technologies combined with the integrative experimental and clinical validations has provided an insight into the underlying mechanism of CCA initiation and progression as well as clues towards novel biomarkers. This work highlights the discovery and validation of molecular markers in CCA identified through omics approaches. The possible roles of these molecules in various cellular pathways, which render CCA carcinogenesis and progression, will also be discussed. This paper can serve as a reference point for further investigations to yield deeper understanding in the complex feature of this disease, potentially leading to better approaches for diagnosis, prognosis, and therapeutics.
Collapse
|
234
|
Abstract
Tumors are typically sequenced to depths of 75-100× (exome) or 30-50× (whole genome). We demonstrate that current sequencing paradigms are inadequate for tumors that are impure, aneuploid or clonally heterogeneous. To reassess optimal sequencing strategies, we performed ultra-deep (up to ~312×) whole genome sequencing (WGS) and exome capture (up to ~433×) of a primary acute myeloid leukemia, its subsequent relapse, and a matched normal skin sample. We tested multiple alignment and variant calling algorithms and validated ~200,000 putative SNVs by sequencing them to depths of ~1,000×. Additional targeted sequencing provided over 10,000× coverage and ddPCR assays provided up to ~250,000× sampling of selected sites. We evaluated the effects of different library generation approaches, depth of sequencing, and analysis strategies on the ability to effectively characterize a complex tumor. This dataset, representing the most comprehensively sequenced tumor described to date, will serve as an invaluable community resource (dbGaP accession id phs000159).
Collapse
|
235
|
Razumilava N, Gores GJ. Building a staircase to precision medicine for biliary tract cancer. Nat Genet 2015; 47:967-8. [DOI: 10.1038/ng.3386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
236
|
Squillace RM, Frampton GM, Stephens PJ, Ross JS, Miller VA. Comparing two assays for clinical genomic profiling: the devil is in the data. Onco Targets Ther 2015; 8:2237-42. [PMID: 26346763 PMCID: PMC4556029 DOI: 10.2147/ott.s88908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
|
237
|
EXP CLIN TRANSPLANTExp Clin Transplant 2015; 13. [DOI: 10.6002/ect.2015.0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
238
|
Nahm JH, Kim H, Lee H, Cho JY, Choi YR, Yoon YS, Han HS, Park YN. Transforming acidic coiled-coil-containing protein 3 (TACC3) overexpression in hepatocellular carcinomas is associated with “stemness” and epithelial-mesenchymal transition-related marker expression and a poor prognosis. Tumour Biol 2015. [DOI: 10.1007/s13277-015-3810-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
239
|
Ang C. Role of the fibroblast growth factor receptor axis in cholangiocarcinoma. J Gastroenterol Hepatol 2015; 30:1116-22. [PMID: 25678238 DOI: 10.1111/jgh.12916] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2015] [Indexed: 02/06/2023]
Abstract
Advanced cholangiocarcinoma (CCA) is a highly lethal disease with limited therapeutic options beyond cytotoxic chemotherapy. Molecular profiling of CCA has provided insights into the pathogenesis of this disease and identified potential therapeutic targets. The fibroblast growth factor receptor (FGFR) axis is important for maintaining tissue homeostasis. Aberrations in FGFR activity have been implicated in the development and progression of CCA and other malignancies, which has generated significant interest in exploring FGFR's therapeutic potential. FGFR2 fusion events are present in up to 17% of intrahepatic CCAs and appear to predict sensitivity to FGFR inhibitors even after progression on chemotherapy. These observations have led to a clinical trial evaluating FGFR inhibition in patients with CCA enriched for FGFR alterations. This review summarizes current knowledge about the role of the FGFR pathway in cholangiocarcinogenesis and ongoing work in developing FGFR-directed therapies as an antineoplastic strategy for CCA.
Collapse
Affiliation(s)
- Celina Ang
- Division of Medicine, Hematology, and Medical Oncology, Mount Sinai School of Medicine, New York, New York, USA
| |
Collapse
|
240
|
Tanizaki J, Ercan D, Capelletti M, Dodge M, Xu C, Bahcall M, Tricker EM, Butaney M, Calles A, Sholl LM, Hammerman PS, Oxnard GR, Wong KK, Jänne PA. Identification of Oncogenic and Drug-Sensitizing Mutations in the Extracellular Domain of FGFR2. Cancer Res 2015; 75:3139-46. [PMID: 26048680 DOI: 10.1158/0008-5472.can-14-3771] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/28/2015] [Indexed: 11/16/2022]
Abstract
The discovery of oncogenic driver mutations and the subsequent developments in targeted therapies have led to improved outcomes for subsets of lung cancer patients. The identification of additional oncogenic and drug-sensitive alterations may similarly lead to new therapeutic approaches for lung cancer. We identify and characterize novel FGFR2 extracellular domain insertion mutations and demonstrate that they are both oncogenic and sensitive to inhibition by FGFR kinase inhibitors. We demonstrate that the mechanism of FGFR2 activation and subsequent transformation is mediated by ligand-independent dimerization and activation of FGFR2 kinase activity. Both FGFR2-mutant forms are predominantly located in the endoplasmic reticulum and Golgi but nevertheless can activate downstream signaling pathways through their interactions with fibroblast growth factor receptor substrate 2 (FRS2). Our findings provide a rationale for therapeutically targeting this unique subset of FGFR2-mutant cancers as well as insight into their oncogenic mechanisms.
Collapse
Affiliation(s)
- Junko Tanizaki
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Dalia Ercan
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Marzia Capelletti
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Michael Dodge
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Chunxiao Xu
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Magda Bahcall
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Erin M Tricker
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Mohit Butaney
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Antonio Calles
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Peter S Hammerman
- Department of Medical Oncology, Boston, Massachusetts. The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Geoffrey R Oxnard
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts. Department of Medicine, Boston, Massachusetts
| | - Kwok-Kin Wong
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts. Department of Medicine, Boston, Massachusetts. Belfer Institute for Applied Cancer Science, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Boston, Massachusetts. Department of Medical Oncology, Boston, Massachusetts. Department of Medicine, Boston, Massachusetts. Belfer Institute for Applied Cancer Science, Dana Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
241
|
Lozada ME, Chaiteerakij R, Roberts LR. Screening for hepatocellular carcinoma and cholangiocarcinoma: Can biomarkers replace imaging? CURRENT HEPATOLOGY REPORTS 2015; 14:128-138. [PMID: 26328266 PMCID: PMC4551404 DOI: 10.1007/s11901-015-0261-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Maria E. Lozada
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905
| | - Roongruedee Chaiteerakij
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905 and Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok 10330, Thailand
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905
| |
Collapse
|
242
|
Oyasiji T, Zhang J, Kuvshinoff B, Iyer R, Hochwald SN. Molecular Targets in Biliary Carcinogenesis and Implications for Therapy. Oncologist 2015; 20:742-51. [PMID: 26025932 DOI: 10.1634/theoncologist.2014-0442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/27/2015] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED 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. 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.
Collapse
Affiliation(s)
- Tolutope Oyasiji
- Departments of Surgical Oncology and Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Jianliang Zhang
- Departments of Surgical Oncology and Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Boris Kuvshinoff
- Departments of Surgical Oncology and Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Renuka Iyer
- Departments of Surgical Oncology and Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Steven N Hochwald
- Departments of Surgical Oncology and Medicine, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
243
|
Flippot R, Kone M, Magné N, Vignot S. [FGF/FGFR signalling: Implication in oncogenesis and perspectives]. Bull Cancer 2015; 102:516-26. [PMID: 25986739 DOI: 10.1016/j.bulcan.2015.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/10/2015] [Indexed: 02/02/2023]
Abstract
Deregulation of FGF (fibroblast growth factor)/FGFR (fibroblast growth factor receptor) signalling leads to the promotion of several oncogenic mechanisms: proliferation, epithelial-mesenchymal transition, cytoskeleton modifications, migration and angiogenesis. Deregulation of this pathway is reported in various cancers at early stages, and can therefore be responsible for the emergence of the hallmarks of cancer. It is necessary to precise downstream pathways of FGFR signalling to understand its oncogenic potential. We will then describe its implications in different cancer types. Oncogenic mechanisms will be studied through the example of melanoma, in which deregulation of FGF/FGFR pathway is considered as a driver event and occurs in nearly 90% of cases. The FGF/FGFR signalling pathway is a putative therapeutic target. Numerous agents are in active development, operating through a selective or multi-targeted approach. Recent studies have shown rather disappointing results in non-selected patients, but promising results in patients with FGF/FGFR pathway alterations. A careful screening of patients is the key to a valuable evaluation of these new targeted molecular therapies.
Collapse
Affiliation(s)
- Ronan Flippot
- Gustave-Roussy, département d'innovations thérapeutiques essais précoces, 94800 Villejuif-Grand Paris, France
| | - Moumini Kone
- Hôpital Louis-Pasteur, service d'oncologie-hématologie, 28630 Chartres-Le-Coudray, France
| | - Nicolas Magné
- Institut de cancérologie Lucien-Neuwirth, département de radiothérapie, 42270 Saint-Priest-en-Jarez, France
| | - Stéphane Vignot
- Hôpital Louis-Pasteur, service d'oncologie-hématologie, 28630 Chartres-Le-Coudray, France.
| |
Collapse
|
244
|
Huang Y, Li X, Zhao Y. Progression of targeted therapy in advanced cholangiocarcinoma. Chin J Cancer Res 2015; 27:122-7. [PMID: 25937773 DOI: 10.3978/j.issn.1000-9604.2015.04.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/03/2015] [Indexed: 01/07/2023] Open
Abstract
It is necessary to establish an effective therapy to improve the survival of patients with advanced cholangiocarcinoma (CCA). Recently, with the development of pathology research in CCA, a lot of special bio-markers such as EGFR, VEGF, HER2, and MEK et al. could be over expression or mutations in CCA patients. According to their changes, combinations of targeted therapy plus chemotherapy are now recognized as effective therapies for advanced CCA. The aim of this paper is to analyze recent promising studies about targeted therapy alone or combination with each other or with chemotherapies.
Collapse
Affiliation(s)
- Yingying Huang
- 1 Oncology Department, Beijing Hospital of the Ministry of Health, Beijing 100038, China ; 2 Oncology Department, Chinese PLA General hospital, Beijing 100853, China
| | - Xiaomei Li
- 1 Oncology Department, Beijing Hospital of the Ministry of Health, Beijing 100038, China ; 2 Oncology Department, Chinese PLA General hospital, Beijing 100853, China
| | - Yunbo Zhao
- 1 Oncology Department, Beijing Hospital of the Ministry of Health, Beijing 100038, China ; 2 Oncology Department, Chinese PLA General hospital, Beijing 100853, China
| |
Collapse
|
245
|
Ornitz DM, Itoh N. The Fibroblast Growth Factor signaling pathway. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2015; 4:215-66. [PMID: 25772309 PMCID: PMC4393358 DOI: 10.1002/wdev.176] [Citation(s) in RCA: 1360] [Impact Index Per Article: 151.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/23/2014] [Accepted: 01/08/2015] [Indexed: 12/13/2022]
Abstract
The signaling component of the mammalian Fibroblast Growth Factor (FGF) family is comprised of eighteen secreted proteins that interact with four signaling tyrosine kinase FGF receptors (FGFRs). Interaction of FGF ligands with their signaling receptors is regulated by protein or proteoglycan cofactors and by extracellular binding proteins. Activated FGFRs phosphorylate specific tyrosine residues that mediate interaction with cytosolic adaptor proteins and the RAS-MAPK, PI3K-AKT, PLCγ, and STAT intracellular signaling pathways. Four structurally related intracellular non-signaling FGFs interact with and regulate the family of voltage gated sodium channels. Members of the FGF family function in the earliest stages of embryonic development and during organogenesis to maintain progenitor cells and mediate their growth, differentiation, survival, and patterning. FGFs also have roles in adult tissues where they mediate metabolic functions, tissue repair, and regeneration, often by reactivating developmental signaling pathways. Consistent with the presence of FGFs in almost all tissues and organs, aberrant activity of the pathway is associated with developmental defects that disrupt organogenesis, impair the response to injury, and result in metabolic disorders, and cancer. For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of MedicineSt. Louis, MO, USA
- *
Correspondence to:
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto UniversitySakyo, Kyoto, Japan
| |
Collapse
|
246
|
Borad MJ, Gores GJ, Roberts LR. Fibroblast growth factor receptor 2 fusions as a target for treating cholangiocarcinoma. Curr Opin Gastroenterol 2015; 31:264-8. [PMID: 25763789 PMCID: PMC4750878 DOI: 10.1097/mog.0000000000000171] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This review will cover the role of the fibroblast growth factor pathway in the pathogenesis, targeted therapy potential and prognostic value in patients with cholangiocarcinoma (CCA). RECENT FINDINGS Recent studies that have identified fibroblast growth factor receptor 2 (FGFR2) fusions, prognostic implications of FGFR2 fusions, treatment strategies that target FGFR2 in CCA and future directions for understanding and targeting the FGFR2 pathway in this disease, will be discussed. SUMMARY Understanding the role of the FGFR2 pathway as a disease pathogenetic mechanism and the ability to develop targeted therapies and diagnostics surrounding this concept are critical elements toward developing novel targeted approaches in CCA.
Collapse
Affiliation(s)
- Mitesh J. Borad
- Division of Hematology/Oncology, Mayo Clinic, Scottsdale, Arizona, USA,Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
247
|
Abstract
Biliary tract cancers are a heterogeneous group of cancers that arise in either the intra- or extrahepatic bile ducts or the gallbladder. Local therapy with surgical resection and perhaps radiation therapy is used for localized disease. There is no known effective adjuvant therapy, although various combinations have been used clinically without definitive data showing a benefit. The most standard chemotherapy for metastatic disease is gemcitabine plus cisplatin based on a single positive randomized trial. Genetic mutations that may lead to better, targeted therapy choices are being identified, albeit with variable frequency. Early studies of targeted agents have been negative, but these were in unselected patients where it was unknown whether the target was activated in any individual patient. Careful selection of patients enrolling onto trials of targeted agents will make the subsets of biliary tract cancers even smaller but is likely necessary to improve outcomes from these deadly diseases.
Collapse
Affiliation(s)
- Emily Chan
- All authors: Vanderbilt University, Nashville, TN.
| | | |
Collapse
|
248
|
Gallo LH, Nelson KN, Meyer AN, Donoghue DJ. Functions of Fibroblast Growth Factor Receptors in cancer defined by novel translocations and mutations. Cytokine Growth Factor Rev 2015; 26:425-49. [PMID: 26003532 DOI: 10.1016/j.cytogfr.2015.03.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 03/25/2015] [Indexed: 11/25/2022]
Abstract
The four receptor tyrosine kinases (RTKs) within the family of Fibroblast Growth Factor Receptors (FGFRs) are critical for normal development but also play an enormous role in oncogenesis. Mutations and/or abnormal expression often lead to constitutive dimerization and kinase activation of FGFRs, and represent the primary mechanism for aberrant signaling. Sequencing of human tumors has revealed a plethora of somatic mutations in FGFRs that are frequently identical to germline mutations in developmental syndromes, and has also identified novel FGFR fusion proteins arising from chromosomal rearrangements that contribute to malignancy. This review details approximately 200 specific point mutations in FGFRs and 40 different fusion proteins created by translocations involving FGFRs that have been identified in human cancer. This review discusses the effects of these genetic alterations on downstream signaling cascades, and the challenge of drug resistance in cancer treatment with antagonists of FGFRs.
Collapse
Affiliation(s)
- Leandro H Gallo
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - Katelyn N Nelson
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - April N Meyer
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093-0367, United States.
| |
Collapse
|
249
|
Future directions in the treatment of cholangiocarcinoma. Best Pract Res Clin Gastroenterol 2015; 29:355-61. [PMID: 25966434 DOI: 10.1016/j.bpg.2015.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 02/11/2015] [Indexed: 01/31/2023]
Abstract
Cholangiocarcinoma (CCA) comprises a heterogeneous group of cancers with pathologic features of biliary tract differentiation, and is best classified anatomically as intrahepatic CCA (ICC), perihilar (pCCA), or distal (dCCA) CCA. They represent a clinically and genetically diverse collection of cancers. Surgical resection represents the only curative modality for CCA, although there are encouraging data with liver transplantation for early stage pCCA. There is no established adjuvant therapy for CCA. Unfortunately, most patients with CCA will present with unresectable or metastatic disease with poor prognosis. Currently the combination of gemcitabine and cisplatin remains the standard therapy for advanced CCA. No second line therapy has definitely demonstrated improved survival benefits. Development of molecularly targeted therapies in advanced CCA remains challenging. However, recent efforts with targeted and whole exome sequencing have defined the landscape of mutations underlying CCA, particularly ICC. The identification of novel molecular signatures in CCA coupled with molecularly targeted therapy development provides the potential for developing novel therapeutic options in this intractable disease.
Collapse
|
250
|
Bruix J, Han KH, Gores G, Llovet JM, Mazzaferro V. Liver cancer: Approaching a personalized care. J Hepatol 2015; 62:S144-56. [PMID: 25920083 PMCID: PMC4520430 DOI: 10.1016/j.jhep.2015.02.007] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 12/04/2022]
Abstract
The knowledge and understanding of all aspects of liver cancer [this including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA)] have experienced a major improvement in the last decades. New laboratory technologies have identified several molecular abnormalities that, at the very end, should provide an accurate stratification and optimal treatment of patients diagnosed with liver cancer. The seminal discovery of the TP53 hotspot mutation [1 ,2 ] was an initial landmark step for the future classification and treatment decision using conventional clinical criteria blended with molecular data. At the same time, the development of ultrasound, computed tomography (CT) and magnetic resonance (MR) has been instrumental for earlier diagnosis, accurate staging and treatment advances. Several treatment options with proven survival benefit if properly applied are now available. Major highlights include: i) acceptance of liver transplantation for HCC if within the Milan criteria [3 ], ii) recognition of ablation as a potentially curative option [4 ,5 ], iii) proof of benefit of chemoembolization (TACE), [6 ] and iv) incorporation of sorafenib as an effective systemic therapy [7 ]. These options are part of the widely endorsed BCLC staging and treatment model (Fig. 1 ) [8 ,9 ]. This is clinically useful and it will certainly keep evolving to accommodate new scientific evidence. This review summarises the data which are the basis for the current recommendations for clinical practice, while simultaneously exposes the areas where more research is needed to fulfil the still unmet needs (Table 1 ).
Collapse
Affiliation(s)
- Jordi Bruix
- Barcelona Clinic Liver Cancer Group (BCLC), Liver Unit, IDIBAPS, CIBERehd, Hospital Clínic, Universitat de Barcelona, Catalonia, Spain.
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gregory Gores
- Mayo Clinic, Mayo College of Medicine, Rochester, MN, USA
| | - Josep Maria Llovet
- Barcelona Clinic Liver Cancer Group (BCLC), Liver Unit, IDIBAPS, CIBERehd, Hospital Clínic, Universitat de Barcelona, Catalonia, Spain; Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
| | - Vincenzo Mazzaferro
- Gastrointestinal Surgery and Liver Transplantation, Istituto Nazionale Tumori IRCCS (National Cancer Institute), Milan 20133, Italy
| |
Collapse
|