1
|
Alrouji M, Yasmin S, Alhumaydhi FA, Sharaf SE, Shahwan M, Shamsi A. ROS1 kinase inhibition reimagined: identifying repurposed drug via virtual screening and molecular dynamics simulations for cancer therapeutics. Front Chem 2024; 12:1392650. [PMID: 39136033 PMCID: PMC11317403 DOI: 10.3389/fchem.2024.1392650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/01/2024] [Indexed: 08/15/2024] Open
Abstract
Precision medicine has revolutionized modern cancer therapeutic management by targeting specific molecular aberrations responsible for the onset and progression of tumorigenesis. ROS proto-oncogene 1 (ROS1) is a receptor tyrosine kinase (RTK) that can induce tumorigenesis through various signaling pathways, such as cell proliferation, survival, migration, and metastasis. It has emerged as a promising therapeutic target in various cancer types. However, there is very limited availability of specific ROS1 inhibitors for therapeutic purposes. Exploring repurposed drugs for rapid and effective treatment is a useful approach. In this study, we utilized an integrated approach of virtual screening and molecular dynamics (MD) simulations of repurposing existing drugs for ROS1 kinase inhibition. Using a curated library of 3648 FDA-approved drugs, virtual screening identified drugs capable of binding to ROS1 kinase domain. The results unveil two hits, Midostaurin and Alectinib with favorable binding profiles and stable interactions with the active site residues of ROS1. These hits were subjected to stability assessment through all-atom MD simulations for 200 ns. MD results showed that Midostaurin and Alectinib were stable with ROS1. Taken together, the study showed a rational framework for the selection of repurposed Midostaurin and Alectinib with ROS1 inhibitory potential for therapeutic management after further validation.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Sharaf E. Sharaf
- Pharmaceutical Sciences Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
2
|
Khan MS, Altwaijry N, Al-Bagmi MS, Alafaleq NO, Alokail MS, Shahwan M, Shamsi A. Structure-guided identification of potent inhibitors of ROS1 kinase for therapeutic development against non-small cell lung cancer. J Biomol Struct Dyn 2024; 42:3837-3847. [PMID: 37254309 DOI: 10.1080/07391102.2023.2217450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/12/2023] [Indexed: 06/01/2023]
Abstract
Proto-oncogene tyrosine-protein kinase ROS (ROS1) is a member of the sevenless receptor, which affects epithelial cell differentiation and is highly expressed in a variety of tumor cells. The elevated expression and dysfunction of ROS1 have been involved in various malignancies, such as non-small cell lung cancer (NSCLC), stomach cancer, ovarian, breast cancer, cholangiocarcinoma, colorectal cancer, adenosarcoma, oesophageal cancer, etc. ROS1 has been postulated as a potential drug target in anticancer therapeutics. In this study, we carried out a virtual screening of phytochemicals against ROS1 to identify its potential inhibitors. The virtual screening process was performed on the ROS1 structure, where two phytochemicals, Helioscopinolide C and Taiwanin C, were identified. These compounds resulted from filters like Lipinski rule of five, PAINS filter, binding affinities values, and all-atom molecular dynamics (MD) simulations followed by principal component analysis (PCA) and essential dynamics. The findings of this study highlight the role of ROS1 in multiple physiological candidates and its therapeutic targeting using phytochemicals. This study suggests Helioscopinolide C and Taiwanin C as potential compounds for therapeutic development targeting ROS1-associated non-small cell lung cancer for clinical applications. Further in vitro and in vivo experiments are required to validate these findings.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Nojood Altwaijry
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Moneera Saud Al-Bagmi
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Nouf Omar Alafaleq
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Majed S Alokail
- Department of Biochemistry, College of Science, King Saud University, KSA, Riyadh, Saudi Arabia
| | - Moyad Shahwan
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Anas Shamsi
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
3
|
Speckart J, Rasmusen V, Talib Z, GnanaDev DA, Rahnemai-Azar AA. Emerging Therapies in Management of Cholangiocarcinoma. Cancers (Basel) 2024; 16:613. [PMID: 38339363 PMCID: PMC10854763 DOI: 10.3390/cancers16030613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Cholangiocarcinoma is a heterogeneous group of biliary tract cancers that has a poor prognosis and globally increasing incidence and mortality. While surgical resection remains the only curative option for the treatment of cholangiocarcinoma, the majority of cancers are unresectable at the time of diagnosis. Additionally, the prognosis of cholangiocarcinoma remains poor even with the current first-line systemic therapy regimens, highlighting the difficulty of treating locally advanced, metastatic, or unresectable cholangiocarcinoma. Through recent developments, targetable oncogenic driver mutations have been identified in the pathogenesis of cholangiocarcinoma, leading to the utilization of molecular targeted therapeutics. In this review, we comprehensively discuss the latest molecular therapeutics for the treatment of cholangiocarcinoma, including emerging immunotherapies, highlighting promising developments and strategies.
Collapse
Affiliation(s)
- Jessica Speckart
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (J.S.); (V.R.)
| | - Veronica Rasmusen
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA; (J.S.); (V.R.)
| | - Zohray Talib
- Department of Medicine, Arrowhead Regional Medical Center, California University of Science and Medicine, Colton, CA 92324, USA;
| | - Dev A. GnanaDev
- Department of Surgery, Arrowhead Regional Medical Center, Colton, CA 92324, USA
| | - Amir A. Rahnemai-Azar
- Division of Surgical Oncology, Department of Surgery, Arrowhead Regional Cancer Center, California University of Science and Medicine, Colton, CA 92324, USA
| |
Collapse
|
4
|
Fountzilas C, Adjei A, Opyrchal M, Evans R, Ghasemi M, Attwood K, Groman A, Bshara W, Goey A, Wilton J, Ma WW, Iyer R. A phase I study of the anaplastic lymphoma kinase inhibitor ceritinib in combination with gemcitabine-based chemotherapy in patients with advanced solid tumors. Int J Cancer 2021; 149:2063-2074. [PMID: 34319586 DOI: 10.1002/ijc.33754] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 11/06/2022]
Abstract
In this phase I, dose-escalation study, we sought to determine the maximum tolerated dose (MTD) of the anaplastic lymphoma kinase/c-ROS oncogene 1 receptor (ALK/ROS1) inhibitor ceritinib in combination with gemcitabine-based chemotherapy in patients with advanced solid tumors. Secondary objectives were characterization of the safety profile, pharmacokinetics and preliminary efficacy of these combinations, and identification of potential biomarkers of efficacy. Ceritinib was combined with gemcitabine (Arm 1), gemcitabine/nab-paclitaxel (Arm 2) or gemcitabine/cisplatin (Arm 3). Drug concentrations in plasma were measured by tandem mass spectrometric detection (LC-MS/MS). We analyzed archival tumor tissue for ALK, ROS1, hepatocyte growth factor receptor (c-MET) and c-Jun N-terminal kinase (JNK) expression by immunohistochemistry. Arm 2 closed early secondary to toxicity. Twenty-one patients were evaluable for dose-limiting toxicity (DLT). There was one DLT in Arm 1 (grade 3 ALT increase) and three DLTs in Arm 3 (grade 3 acute renal failure, grade 3 thrombocytopenia, grade 3 dyspnea). The MTD of ceritinib was determined to be 600 mg (Arm 1) and 450 mg orally daily (Arm 3). Main toxicities were hematologic, constitutional and gastrointestinal as expected by the chemotherapy backbone. The apparent clearance for ceritinib decreased substantially after repeated dosing; cisplatin did not significantly affect the pharmacokinetics of ceritinib. The overall response rate was 20%; the median progression-free survival was 4.8 months. Three out of five response-evaluable cholangiocarcinoma patients had clinical benefit. Increased expression of c-MET was associated with a lack of clinical benefit. Ceritinib in combination with gemcitabine and gemcitabine/cisplatin has a manageable toxicity profile. Further development of this strategy in tumors with ALK or ROS1 fusions is warranted.
Collapse
Affiliation(s)
- Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Alex Adjei
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mateusz Opyrchal
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rachel Evans
- Clinical Research Services, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Mohammad Ghasemi
- Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Adrienne Groman
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Wiam Bshara
- Pathology Resource Network, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Andrew Goey
- Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - John Wilton
- Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Wen Wee Ma
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Renuka Iyer
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| |
Collapse
|
5
|
Mazzoni F, Petreni P, Vasile E, Panebianco M, Casadei-Gardini A, Negri F, Lunghi A, Pillozzi S, Vivaldi C, Gervasi E, Frassineti GL, Messerini L, Jocollé G, Bisagni A, Antonuzzo L, Rossi G. ROS1 rearrangements are uncommon in biliary tract cancers. Oncol Lett 2020; 20:316. [PMID: 33133252 DOI: 10.3892/ol.2020.12179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/31/2020] [Indexed: 11/06/2022] Open
Abstract
Biliary tract cancers (BTCs) are a pool of diseases with poor prognosis and there is no orphan drug available. Currently, no molecular targets have been tested as druggable oncogenic drivers. C-ros oncogene 1 (ROS1) rearrangements have been previously described in various tumors, including BTCs; however, data regarding their incidence and biological significance are controversial. Therefore, a retrospective multicenter study was performed to assess the incidence of ROS1 rearrangements in BTCs by means of immunohistochemistry and fluorescence in situ hybridization (FISH). The present study failed to demonstrate ROS1 expression in a multicenter series of 150 cases with BTCs and revealed that D4D6 was the most specific clone compared with other ROS1 primary antibodies, namely PA1-30318 and EPMGHR2. Notably, negative results obtained with D4D6 completely matched to data sorted out by FISH analysis, thus confirming a lack of ROS1 gene rearrangements in BTCs and false positive results when PA1-30318 and EPMGHR2 clones were used. These results suggest that ROS1 rearrangements may not be targets for molecular therapy of BTCs with specific inhibitors.
Collapse
Affiliation(s)
- Francesca Mazzoni
- Medical Oncology Unit, AOU Careggi Hospital, I-50134 Florence, Italy
| | - Paolo Petreni
- Medical Oncology Unit, Alta Val d'Elsa Hospital, I-53036 Poggibonsi, Italy
| | - Enrico Vasile
- Medical Oncology Unit, AOU Pisana Hospital, I-56126 Pisa, Italy
| | | | | | - Francesca Negri
- Medical Oncology Unit, AOU Parma Hospital, I-43126 Parma, Italy
| | - Alice Lunghi
- Medical Oncology Unit, San Luca Hospital, I-55100 Lucca, Italy
| | - Serena Pillozzi
- Medical Oncology Unit, AOU Careggi Hospital, I-50134 Florence, Italy.,Department of Experimental and Clinical Medicine, University of Florence, I-50134 Florence, Italy
| | | | | | | | - Luca Messerini
- Anatomical Pathology Unit, AOU Careggi Hospital, I-50134 Florence, Italy
| | | | | | - Lorenzo Antonuzzo
- Medical Oncology Unit, AOU Careggi Hospital, I-50134 Florence, Italy
| | - Giulio Rossi
- Operative Unit of Pathologic Anatomy, Santa Maria delle Croci Hospital, I-48121 Ravenna, Italy
| |
Collapse
|
6
|
Jakubowski CD, Mohan AA, Kamel IR, Yarchoan M. Response to Crizotinib in ROS1 Fusion-Positive Intrahepatic Cholangiocarcinoma. JCO Precis Oncol 2020; 4:825-828. [PMID: 35050759 PMCID: PMC9797238 DOI: 10.1200/po.20.00116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Christopher D. Jakubowski
- Bloomberg-Kimmel Institute for Cancer
Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins
University School of Medicine, Baltimore, MD
| | - Aditya A. Mohan
- Bloomberg-Kimmel Institute for Cancer
Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins
University School of Medicine, Baltimore, MD
| | - Ihab R. Kamel
- Russell H. Morgan Department of Radiology
and Radiological Sciences, Johns Hopkins Hospital, Baltimore, MD
| | - Mark Yarchoan
- Bloomberg-Kimmel Institute for Cancer
Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins
University School of Medicine, Baltimore, MD,Mark Yarchoan, Johns Hopkins University School of Medicine, 1450
Orleans St, Baltimore, MD 21287; Twitter: @MarkYarchoan; e-mail:
| |
Collapse
|
7
|
Palmieri LJ, Lavolé J, Dermine S, Brezault C, Dhooge M, Barré A, Chaussade S, Coriat R. The choice for the optimal therapy in advanced biliary tract cancers: Chemotherapy, targeted therapies or immunotherapy. Pharmacol Ther 2020; 210:107517. [PMID: 32109491 DOI: 10.1016/j.pharmthera.2020.107517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/18/2020] [Indexed: 01/09/2023]
Abstract
Biliary tract cancers (BTCs) represent a heterogeneous group that includes intrahepatic cholangiocarcinomas (CCAs), perihilar-CCAs or Klatskin tumors, extrahepatic-CCAs, and gallbladder adenocarcinoma. These entities have distinct demographics, risk factors, clinical presentation, and molecular characteristics. In advanced BTCs, the recommendations are mainly supporting a doublet chemotherapy regimen using cisplatin/gemcitabine (CisGem) with a 5-year overall survival rate close to 5% and median overall survival (mOS) of less than a year. The lack of overall efficacy stresses the need for personalized therapies. Recently, whole-genome and transcriptome sequencing highlighted the diversity of BTCs' subtypes. Distinct genetic alterations were retrieved according to the localization, with a high rate of potentially actionable alterations. Targeted therapies and immunotherapy have since then been tested for BTCs, trying to propose a more personalized treatment. This review describes the different therapeutic options, validated and in development, for patients with advanced BTCs.
Collapse
Affiliation(s)
- L-J Palmieri
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Unité INSERM U1016, University of Paris, France.
| | - J Lavolé
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - S Dermine
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Unité INSERM U1016, University of Paris, France
| | - C Brezault
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - M Dhooge
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - A Barré
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Unité INSERM U1016, University of Paris, France
| | - S Chaussade
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Unité INSERM U1016, University of Paris, France
| | - R Coriat
- Gastroenterology and Digestive Oncology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris 75014, France; Unité INSERM U1016, University of Paris, France
| |
Collapse
|
8
|
Qiao J, Li M, Sun D, Li W, Xin Y. Knockdown of ROS proto-oncogene 1 inhibits migration and invasion in gastric cancer cells by targeting the PI3K/Akt signaling pathway. Onco Targets Ther 2019; 12:8569-8582. [PMID: 31802893 PMCID: PMC6801563 DOI: 10.2147/ott.s213421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Objectives Gastric cancer ranks the fourth most common cancer and the third leading cause of cancer mortality in the world. ROS proto-oncogene 1 (ROS1) is an oncogene and ROS1 rearrangement has been reported in many cancers. Our study aimed to investigate the potential function and the precise mechanisms of ROS1 in gastric cancer. Methods In our study, the analysis of ROS1 expression and clinical pathologic factors of gastric cancer in gastric cancer using TCGA database demonstrated that ROS1 expression was elevated in gastric cancer and related to T, N, M and TNM staging. High expression of ROS1 predicted poor survival in patients with gastric cancer. Then, we measured ROS1 expression in four human gastric cancer cell lines and knocked down ROS1 expression in BGC-823 and SGC-7901 cells by specific shRNA transfection via Lipofectamine 2000. The effect of ROS1 knockdown on cell proliferation, cell cycle distribution, cell apoptosis and metastasis in vitro was evaluated by MTT, colony formation, flow cytometric analysis, wound healing and Transwell invasion assays. The levels of apoptosis-related proteins, EMT markers and the PI3K/Akt signaling pathway members were measured by Western blotting. Results We demonstrated that shROS1 transfection markedly downregulated ROS1 expression in BGC-823 and SGC-7901 cells. Knockdown of ROS1 inhibited cell survival, clonogenic growth, migration, invasion and epithelial–mesenchymal transition (EMT), as well as induced cell cycle arrest and apoptosis in gastric cancer cells. Furthermore, ROS1 knockdown inhibited the phosphorylation of PI3K and Akt. Conclusion Collectively, our data suggest that ROS1 may serve as a promising therapeutic target in gastric cancer treatment.
Collapse
Affiliation(s)
- Jingjing Qiao
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, People's Republic of China.,Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116027, People's Republic of China
| | - Man Li
- Department of Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province 116027, People's Republic of China
| | - Dan Sun
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Wenhui Li
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, People's Republic of China
| | - Yan Xin
- Laboratory of Gastrointestinal Onco-Pathology, Cancer Institute and General Surgery Institute, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, People's Republic of China
| |
Collapse
|
9
|
Nyambura LW, Muñoz AA, le Coutre P, Walden P. HLA class I-restricted T cell epitopes isolated and identified from myeloid leukemia cells. Sci Rep 2019; 9:14029. [PMID: 31575892 PMCID: PMC6773711 DOI: 10.1038/s41598-019-50341-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/11/2019] [Indexed: 12/19/2022] Open
Abstract
Leukemia-associated antigens (LAAs) and HLA-I epitopes published previously have shown promise in inducing leukemia-specific T cell responses. However, the clinical responses are limited, and clinical effectiveness is yet to be achieved. Limitations, among others, being the LAAs themselves, the indirect approach to HLA-I epitope identification by reverse immunology, and the use of single or few LAAs and HLA-I epitopes, which limits the spectrum of inducible tumor-specific T cells. Use of a direct approach to identify naturally processed and presented HLA-I epitopes from LAAs, and higher numbers of antigens for T cell-mediated immunotherapy for leukemia may enhance clinical responses and broaden clinical effectiveness. In a prior study we used immunoaffinity purification of HLA-I peptide complexes from the differentiated myeloid tumor cell lines MUTZ3 and THP1 coupled to high-performance liquid chromatography tandem mass spectrometry (LC-MS/MS). From this we identified in the current study seven new HLA-I epitopes and the corresponding LAAs for myeloid leukemia. In comparison, the myeloid HLA-I epitopes reported here were generally stronger HLA-binders that induce stronger T cell responses than those previously published, and their source LAAs had higher immunogenicity, higher expression levels in myeloid tumors cells compared to normal hemopoietin and other major normal tissues, and more protein interaction partners, and they are targeted by CD8 T cells in CML patients. This study analyses and compares the LAAs and HLA-I epitopes based on various immunotherapeutic targets selection criteria, and highlights new targets for T cell-mediated immunotherapy for leukemia.
Collapse
Affiliation(s)
- Lydon Wainaina Nyambura
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Alejandro Azorin Muñoz
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Philipp le Coutre
- Medical Department, Division of Hematology and Oncology, Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany
| | - Peter Walden
- Department of Dermatology, Venerology and Allergology, Clinical Research Group 'Tumor Immunology', Charité - Universitätsmedizin Berlin corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10098, Berlin, Germany.
| |
Collapse
|
10
|
Tessier-Cloutier B, Cai E, Schaeffer DF. Off-label use of common predictive biomarkers in gastrointestinal malignancies: a critical appraisal. Diagn Pathol 2019; 14:62. [PMID: 31221175 PMCID: PMC6587260 DOI: 10.1186/s13000-019-0843-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/11/2019] [Indexed: 12/13/2022] Open
Abstract
The use of immunohistochemistry (IHC) as a companion diagnostic is an increasingly important part of the case workup by pathologists and is often central to clinical decision making. New predictive molecular markers are constantly sought for to improve treatment stratification parallel to drug development. Unfortunately, official biomarker guidelines lag behind, and pathologists are often left hesitating when medical oncologists request off-labelled biomarker testing. We performed a literature review of five commonly requested off-label IHC predictive biomarkers in gastrointestinal tract (GIT) malignancies: HER2, mismatch repair (MMR), PD-L1, BRAF V600E and ROS1. We found that HER2 amplification is rare and poorly associated to IHC overexpression in extracolonic and extragastric GIT cancers; however in KRAS wild type colorectal cancers, which fail conventional treatment, HER2 IHC may be useful and should be considered. For MMR testing, more evidence is needed to recommend reflex testing in GIT cancers for treatment purposes. MMR testing should not be discouraged in patients considered for second line checkpoint inhibitor therapy. With the exception of gastric tumors, PD-L1 IHC is a weak predictor of checkpoint inhibitor response in the GIT and should be replaced by MMR in this context. BRAF inhibitors showed activity in BRAF V600E mutated cholangiocarcinomas and pancreatic carcinomas in non-first line settings. ROS1 translocation is extremely rare and poorly correlated to ROS1 IHC expression in the GIT; currently there is no role for ROS1 IHC testing in GIT cancers. Overall, the predictive biomarker literature has grown exponentially, and official guidelines need to be updated more regularly to support pathologists’ testing decisions.
Collapse
Affiliation(s)
- Basile Tessier-Cloutier
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 910 West 10th Ave, Vancouver, BC, Canada
| | - Ellen Cai
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 910 West 10th Ave, Vancouver, BC, Canada
| | - David F Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada. .,Department of Pathology and Laboratory Medicine, Vancouver General Hospital, 910 West 10th Ave, Vancouver, BC, Canada.
| |
Collapse
|
11
|
Bogenberger JM, DeLeon TT, Arora M, Ahn DH, Borad MJ. Emerging role of precision medicine in biliary tract cancers. NPJ Precis Oncol 2018; 2:21. [PMID: 30302397 PMCID: PMC6170410 DOI: 10.1038/s41698-018-0064-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 12/14/2022] Open
Abstract
Biliary tracts cancers (BTCs) are a diverse group of aggressive malignancies with an overall poor prognosis. Genomic characterization has uncovered many putative clinically actionable aberrations that can also facilitate the prognostication of patients. As such, comprehensive genomic profiling is playing a growing role in the clinical management of BTCs. Currently however, there is only one precision medicine approved by the US Food and Drug Administration (FDA) for the treatment of BTCs. Herein, we highlight the prevalence and prognostic, diagnostic, and predictive significance of recurrent mutations and other genomic aberrations with current clinical implications or emerging relevance to clinical practice. Some ongoing clinical trials, as well as future areas of exploration for precision oncology in BTCs are highlighted.
Collapse
Affiliation(s)
- James M. Bogenberger
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Thomas T. DeLeon
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Mansi Arora
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Daniel H. Ahn
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ USA
| | - Mitesh J. Borad
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN USA
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ USA
| |
Collapse
|
12
|
Wu YL, Yang JCH, Kim DW, Lu S, Zhou J, Seto T, Yang JJ, Yamamoto N, Ahn MJ, Takahashi T, Yamanaka T, Kemner A, Roychowdhury D, Paolini J, Usari T, Wilner KD, Goto K. Phase II Study of Crizotinib in East Asian Patients With ROS1-Positive Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2018; 36:1405-1411. [PMID: 29596029 DOI: 10.1200/jco.2017.75.5587] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Approximately 1% to 2% of non-small-cell lung cancers (NSCLCs) harbor a c-ros oncogene 1 ( ROS1) rearrangement. Crizotinib, an inhibitor of anaplastic lymphoma kinase (ALK), ROS1, and MET, has shown marked antitumor activity in a small expansion cohort of patients with ROS1-positive advanced NSCLC from an ongoing phase I study. We assessed the efficacy and safety of crizotinib in the largest cohort of patients with ROS1-positive advanced NSCLC. Patients and Methods This phase II, open-label, single-arm trial enrolled East Asian patients with ROS1-positive (assessed through validated AmoyDx assay [Amoy Diagnostics, Xiamen, China] at three regional laboratories) advanced NSCLC who had received three or fewer lines of prior systemic therapies. Patients were to receive oral crizotinib at a starting dose of 250 mg twice daily and continued treatment until Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1-defined progression (by independent radiology review [IRR]), unacceptable toxicity, or withdrawal of consent. The primary end point was objective response rate (ORR) by IRR. Results In the efficacy and safety analyses, 127 patients were included, with 49.6% still receiving treatment at data cutoff. ORR by IRR was 71.7% (95% CI, 63.0% to 79.3%), with 17 complete responses and 74 partial responses. ORRs were similar irrespective of the number of prior lines of therapy, and responses were durable (median duration of response, 19.7 months; 95% CI, 14.1 months to not reached). Median progression-free survival by IRR was 15.9 months (95% CI, 12.9 to 24.0 months). No new safety signals associated with crizotinib were reported. Conclusion This study demonstrated clinically meaningful benefit and durable responses with crizotinib in East Asian patients with ROS1-positive advanced NSCLC. Crizotinib was generally well tolerated, with a safety profile consistent with previous reports.
Collapse
Affiliation(s)
- Yi-Long Wu
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - James Chih-Hsin Yang
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Dong-Wan Kim
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Shun Lu
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Jianying Zhou
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Takashi Seto
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Jin-Ji Yang
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Noboru Yamamoto
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Myung-Ju Ahn
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Toshiaki Takahashi
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Takeharu Yamanaka
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Allison Kemner
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Debasish Roychowdhury
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Jolanda Paolini
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Tiziana Usari
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Keith D Wilner
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| | - Koichi Goto
- Yi-Long Wu and Jin-Ji Yang, Guangdong General Hospital and Guangdong Academy of Medical Sciences, Guangzhou; Shun Lu, Jiao Tong University, Shanghai; Jianying Zhou, First Affiliated Hospital of Zhejiang University, Hangzhou, People's Republic of China; James Chih-Hsin Yang, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Republic of China; Dong-Wan Kim, Seoul National University Hospital; Myung-Ju Ahn, Samsung Medical Center, Seoul, South Korea; Takashi Seto, National Kyushu Cancer Center, Fukuoka; Noboru Yamamoto, National Cancer Center Hospital, Tokyo; Toshiaki Takahashi, Shizuoka Cancer Center, Shizuoka; Takeharu Yamanaka, Yokohama City University School of Medicine, Yokohama; Koichi Goto, National Cancer Center Hospital East, Kashiwa, Japan; Allison Kemner and Debasish Roychowdhury, OxOnc Development, Princeton, NJ; Nirvan Consultants, Lexington, MA; Jolanda Paolini and Tiziana Usari, Pfizer, Milan, Italy; and Keith D. Wilner, Pfizer, La Jolla, CA
| |
Collapse
|
13
|
Lombardi P, Marino D, Fenocchio E, Chilà G, Aglietta M, Leone F. Emerging molecular target antagonists for the treatment of biliary tract cancer. Expert Opin Emerg Drugs 2018; 23:63-75. [PMID: 29468924 DOI: 10.1080/14728214.2018.1444749] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Biliary tract cancers (BTCs) are a heterogeneous group of cancers, characterized by low incidence but poor prognosis. Even after complete surgical resection for early stage, relapse is frequent and the lack of effective treatments contributes to the dismal prognosis. To date, the only standard treatment in first-line is cisplatin/gemcitabine combination, whereas no standard in 2nd-line has been defined. Hence, the current goal is to better understand the biology of BTCs, discovering new treatment methods and improving clinical outcomes. Areas covered: The development of next-generation-sequencing has unveiled the picture of the molecular signatures characterizing BTCs, leading to the identification of actionable mutations in biomarker-driven clinical trials. In this review we will cover the genetic landscape of BTC, focusing on the efficacy of existing treatments. Furthermore, we will discuss emerging molecular targets and evaluate the findings of pre-clinical studies. Finally, the encouraging results of clinical trials involving targeted therapies or immunotherapy will be reviewed. Expert opinion: FGFR fusion rearrangements and IDH1 or IDH2 mutations are the most promising targeted treatments under evaluation. In addition, innovative trial design will allow to offer a chance for tailored medicine to infrequent subgroups of BTCs patients based on their molecular features rather than their histology.
Collapse
Affiliation(s)
- Pasquale Lombardi
- a Department of Oncology , University of Turin Medical School , Turin , Italy
| | - Donatella Marino
- b Medical Oncology , Candiolo Cancer Institute - FPO- IRCCS , Candiolo , Italy
| | | | - Giovanna Chilà
- a Department of Oncology , University of Turin Medical School , Turin , Italy
| | - Massimo Aglietta
- a Department of Oncology , University of Turin Medical School , Turin , Italy.,b Medical Oncology , Candiolo Cancer Institute - FPO- IRCCS , Candiolo , Italy
| | - Francesco Leone
- a Department of Oncology , University of Turin Medical School , Turin , Italy
| |
Collapse
|
14
|
Abstract
ROS1 is a receptor tyrosine kinase that has recently been shown to undergo gene rearrangements in~1%-2% of non-small cell lung carcinoma (NSCLC) and in a variety of other tumours including cholangiocarcinoma, gastric carcinoma, colorectal carcinoma and in spitzoid neoplasms, glioblastoma and inflammatory myofibroblastic tumours. The ROS1 gene fusion undergoes constitutive activation, regulates cellular proliferation and is implicated in carcinogenesis. ROS1 fusions can be detected by fluorescence in situ hybridisation, real-time PCR, sequencing-based techniques and immunohistochemistry-based methods in clinical laboratories. The small molecule tyrosine kinase inhibitor, crizotinib has been shown to be an effective inhibitor of ROS1 and has received Food and Drug Administration approval for treatment of advanced NSCLC. The current review is an update on the clinical findings and detection methods of ROS1 in clinical laboratories in NSCLC and other tumours.
Collapse
Affiliation(s)
- Prodipto Pal
- Department of Laboratory Medicine and Pathobiology, University Health Network - University of Toronto, Toronto, Canada
| | - Zanobia Khan
- Department of Laboratory Medicine and Pathobiology, University Health Network - Lakeridge Regional Health Center, Toronto, Canada
| |
Collapse
|
15
|
Valle JW, Lamarca A, Goyal L, Barriuso J, Zhu AX, Knittel G, Leeser U, van Oers J, Edelmann W, Heukamp LC, Reinhardt HC. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov 2017. [PMID: 28818953 DOI: 10.1158/2159-8290] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biliary tract cancers (BTC), including cholangiocarcinoma and gallbladder cancer, are poor-prognosis and low-incidence cancers, although the incidence of intrahepatic cholangiocarcinoma is rising. A minority of patients present with resectable disease but relapse rates are high; benefit from adjuvant capecitabine chemotherapy has been demonstrated. Cisplatin/gemcitabine combination chemotherapy has emerged as the reference first-line treatment regimen; there is no standard second-line therapy. Selected patients may be suitable for liver-directed therapy (e.g., radioembolization or external beam radiation), pending confirmation of benefit in randomized studies. Initial trials targeting the epithelial growth factor receptor and angiogenesis pathways have failed to deliver new treatments. Emerging data from next-generation sequencing analyses have identified actionable mutations (e.g., FGFR fusion rearrangements and IDH1 and IDH2 mutations), with several targeted drugs entering clinical development with encouraging results. The role of systemic therapies, including targeted therapies and immunotherapy for BTC, is rapidly evolving and is the subject of this review.Significance: The authors address genetic drivers and molecular biology from a translational perspective, in an intent to offer a clear view of the recent past, present, and future of BTC. The review describes a state-of-the-art update of the current status and future directions of research and therapy in advanced BTC. Cancer Discov; 7(9); 943-62. ©2017 AACR.
Collapse
Affiliation(s)
- Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK. .,Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK
| | - Lipika Goyal
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jorge Barriuso
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK.,Faculty of Medical, Biological and Human Sciences, University of Manchester, Rumford Street, Manchester, UK
| | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| | | | | | | | | | | | | |
Collapse
|
16
|
Valle JW, Lamarca A, Goyal L, Barriuso J, Zhu AX. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov 2017; 7:943-962. [PMID: 28818953 DOI: 10.1158/2159-8290.cd-17-0245] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 05/24/2017] [Accepted: 07/10/2017] [Indexed: 02/06/2023]
Abstract
Biliary tract cancers (BTC), including cholangiocarcinoma and gallbladder cancer, are poor-prognosis and low-incidence cancers, although the incidence of intrahepatic cholangiocarcinoma is rising. A minority of patients present with resectable disease but relapse rates are high; benefit from adjuvant capecitabine chemotherapy has been demonstrated. Cisplatin/gemcitabine combination chemotherapy has emerged as the reference first-line treatment regimen; there is no standard second-line therapy. Selected patients may be suitable for liver-directed therapy (e.g., radioembolization or external beam radiation), pending confirmation of benefit in randomized studies. Initial trials targeting the epithelial growth factor receptor and angiogenesis pathways have failed to deliver new treatments. Emerging data from next-generation sequencing analyses have identified actionable mutations (e.g., FGFR fusion rearrangements and IDH1 and IDH2 mutations), with several targeted drugs entering clinical development with encouraging results. The role of systemic therapies, including targeted therapies and immunotherapy for BTC, is rapidly evolving and is the subject of this review.Significance: The authors address genetic drivers and molecular biology from a translational perspective, in an intent to offer a clear view of the recent past, present, and future of BTC. The review describes a state-of-the-art update of the current status and future directions of research and therapy in advanced BTC. Cancer Discov; 7(9); 943-62. ©2017 AACR.
Collapse
Affiliation(s)
- Juan W Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK. .,Institute of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester, UK
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK
| | - Lipika Goyal
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Jorge Barriuso
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, UK.,Faculty of Medical, Biological and Human Sciences, University of Manchester, Rumford Street, Manchester, UK
| | - Andrew X Zhu
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Buettner S, van Vugt JLA, IJzermans JN, Groot Koerkamp B. Intrahepatic cholangiocarcinoma: current perspectives. Onco Targets Ther 2017; 10:1131-1142. [PMID: 28260927 PMCID: PMC5328612 DOI: 10.2147/ott.s93629] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is the second most common malignancy arising from the liver. ICC makes up about 10% of all cholangiocarcinomas. It arises from the peripheral bile ducts within the liver parenchyma, proximal to the secondary biliary radicals. Histologically, the majority of ICCs are adenocarcinomas. Only a minority of patients (15%) present with resectable disease, with a median survival of less than 3 years. Multidisciplinary management of ICC is complicated by large differences in disease course for individual patients both across and within tumor stages. Risk models and nomograms have been developed to more accurately predict survival of individual patients based on clinical parameters. Predictive risk factors are necessary to improve patient selection for systemic treatments. Molecular differences between tumors, such as in the epidermal growth factor receptor status, are promising, but their clinical applicability should be validated. For patients with locally advanced disease, several treatment strategies are being evaluated. Both hepatic arterial infusion chemotherapy with floxuridine and yttrium-90 embolization aim to downstage locally advanced ICC. Selected patients have resectable disease after downstaging, and other patients might benefit because of postponing widespread dissemination and biliary obstruction.
Collapse
Affiliation(s)
- Stefan Buettner
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jeroen LA van Vugt
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jan Nm IJzermans
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Bas Groot Koerkamp
- Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
18
|
Wu J, Lin Y, He X, Yang H, He P, Fu X, Li G, Gu X. Comparison of detection methods and follow-up study on the tyrosine kinase inhibitors therapy in non-small cell lung cancer patients with ROS1 fusion rearrangement. BMC Cancer 2016; 16:599. [PMID: 27488371 PMCID: PMC4973062 DOI: 10.1186/s12885-016-2582-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 07/19/2016] [Indexed: 01/15/2023] Open
Abstract
Background The screening of ROS proto-oncogene 1, receptor tyrosine kinase(ROS1) fusion rearrangement might be potentially beneficial for an effective therapy against non-small cell lung cancer (NSCLC). However, the three main ROS1 rearrangement detection methods have limitations, and no routine protocol for the detection of ROS1 rearrangement in NSCLC is available. In this study, our aims were to compare immunohistochemistry (IHC), fluorescent in situ hybridization (FISH) and quantitative real-time polymerase chain reaction (qRT-PCR) in their ability to detect ROS1 rearrangement in NSCLC, and discuss the clinical characteristics and histopathology of the patients with ROS1 rearrangement. Moreover, the effects of tyrosine kinase inhibitors (TKIs) therapy on the patients with ROS1 rearrangement and advanced stage disease (III b–IV) were investigated. Methods Patients with a previously diagnosed NSCLC were recruited in this study from November 2013 to October 2015. IHC was performed using the D4D6 monoclonal antibody (mAb) in an automatic IHC instrument, while FISH and qRT-PCR were carried out to confirm the IHC results. FISH and qRT-PCR positive cases underwent direct sequencing. After detection, patients with advanced ROS1 rearranged NSCLC had received TKI therapy. Results Two hundred and thirty-eight patients were included in this study. ROS1 rearrangement was detected in 10 patients. The concordant rate of FISH and qRT-PCR results was 100 %, while in the FISH and IHC results high congruence was present when IHC showed a diffusely (≥60 % tumor cells) 2–3+ cytoplasmic reactivity pattern. Patients harboring ROS1 rearrangement were mostly young (8/10), females (7/10) and non-smokers (7/10) with adenocarcinoma (10/10) and acinar pattern. Most of their tumor were in intermediate grade (6/8). Among these 10 patients, three of them in stage IV with ROS1 rearrangement gained benefits from ROS1 TKI therapy. Conclusions IHC, FISH and qRT-PCR can reliably detect ROS1 rearrangement in NSCLC, while IHC can be used as a preliminary screening tool. These results supported the efficacy of ROS1 TKI therapy in treating advanced NSCLC patients with ROS1 rearrangement. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2582-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jieyu Wu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Yunen Lin
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xinming He
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Haihong Yang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ping He
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xinge Fu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Guangqiu Li
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China
| | - Xia Gu
- Department of Pathology, the First Affiliated Hospital of Guangzhou Medical University, No. 151, Yanjiangxi Road, Guangzhou, 510120, China.
| |
Collapse
|
19
|
Chiang NJ, Hsu C, Chen JS, Tsou HH, Shen YY, Chao Y, Chen MH, Yeh TS, Shan YS, Huang SF, Chen LT. Expression levels of ROS1/ALK/c-MET and therapeutic efficacy of cetuximab plus chemotherapy in advanced biliary tract cancer. Sci Rep 2016; 6:25369. [PMID: 27136744 PMCID: PMC4853728 DOI: 10.1038/srep25369] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/15/2016] [Indexed: 12/26/2022] Open
Abstract
Aberrant expression of ROS1, ALK or c-MET (RAM) is implicated in carcinogenesis and cancer drug resistance. We retrospectively evaluated the effect of RAM expression on outcomes for advanced biliary tract cancer patients, who were treated with gemcitabine plus oxaliplatin (GEMOX), with or without cetuximab, in a randomized phase II trial. RAM expression levels on archived tissue sections were scored using immunohistochemistry (IHC). Of 110 tumors with IHC staining for all three markers, 18 were RAMhigh (IHC intensity 3+ for any markers). Ninety-two tumors were RAMlow (IHC intensity <3+ for all markers). All RAMhigh tumors were intra-hepatic cholangiocarcinomas (IHCC). Of the patients with IHCC (n = 80), median overall survival (OS) of RAMhigh group was inferior to that of the RAMlow group (5.7 vs. 11.7 months, p = 0.021). In multivariate analysis RAMhigh remained an independently adverse prognostic factor, with a hazard ratio of 2.01 (p = 0.039). In the RAMlow group, GEMOX treatment with cetuximab significantly improved the disease control rate (68% vs. 41%, p = 0.044), median progression-free survival (7.3 vs. 4.9 months, p = 0.026), and marginally prolonged median OS (14.1 vs 9.6 months, p = 0.056), compared to GEMOX treatment alone. Future trials of anti-EGFR inhibitors for IHCC may consider RAM expression as a patient stratification factor.
Collapse
Affiliation(s)
- Nai-Jung Chiang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, and National Taiwan University Cancer Center, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jen-Shi Chen
- Division of Hematology and Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Hsiao-Hui Tsou
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan.,Graduate Institute of Biostatistics, College of Public Health, China Medical University, Taichung, Taiwan
| | - Ying-Ying Shen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yee Chao
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Huang Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan.,Institute of Clinical Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Lim SM, Yoo JE, Lim KH, Meng Tai DW, Cho BC, Park YN. Rare Incidence of ROS1 Rearrangement in Cholangiocarcinoma. Cancer Res Treat 2016; 49:185-192. [PMID: 27121721 PMCID: PMC5266400 DOI: 10.4143/crt.2015.497] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/02/2016] [Indexed: 12/23/2022] Open
Abstract
Purpose The recent discovery and characterization of an oncogenic ROS1 gene rearrangement has raised significant interest because small molecule inhibitors are effective in these tumors. The aim of this study was to determine frequency and clinicopathological features associated with ROS1 rearrangement in patients with cholangiocarcinoma (CCA). Materials and Methods A total of 261 patients who underwent surgery for CCA between October 1997 and August 2013 were identified from an international, multi-institutional database. ROS1 rearrangement was evaluated by break-apart fluorescence in situ hybridization using tissue microarrays of these patients. Results Of 261 CCA evaluated, three cases (1.1%) showed ROS1 rearrangement by fluorescence in situ hybridization (FISH), all of which were derived from intrahepatic origin. ROS1 protein expression was observed in 38 samples (19.1%). Significantly larger tumor size was observed in ROS1 immunohistochemistry (IHC)–negative patients compared with ROS1 IHC–positive patients. ROS1 FISH–positive patients had a single tumor with a median size of 4 cm and well-to-moderate differentiation. Overall, there was no difference in terms of baseline characteristics, overall survival, and recurrence-free survival between ROS1-positive and -negative patients. Conclusion ROS1 rearrangement was detected in 1.1% of CCA patients. Although rare, conduct of clinical trials using ROS1 inhibitors in these genetically unique patients is warranted.
Collapse
Affiliation(s)
- Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jeong Eun Yoo
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Singapore
| | - David Wai Meng Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Young Nyun Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Integrated Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|