1
|
Tsilimigras DI, Stecko H, Ntanasis-Stathopoulos I, Pawlik TM. Racial and Sex Differences in Genomic Profiling of Intrahepatic Cholangiocarcinoma. Ann Surg Oncol 2024; 31:9071-9078. [PMID: 39251514 PMCID: PMC11549159 DOI: 10.1245/s10434-024-16141-8] [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: 05/24/2024] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Racial and sex disparities in the incidence and outcomes of patients with intrahepatic cholangiocarcinoma (iCCA) exist, yet potential genomic variations of iCCA based on race and sex that might be contributing to disparate outcomes have not been well studied. METHODS Data from the American Association for Cancer Research Project GENIE registry (version 15.0) were analyzed to assess genetic variations in iCCA. Adult patients (age >18 years) with histologically confirmed iCCA who underwent next-generation sequencing were included in the analytic cohort. Racial and sex variations in genomic profiling of iCCA were examined. RESULTS The study enrolled 1068 patients from 19 centers (White, 71.9%; Black, 5.1%; Asian, 8.4%, other, 14.6%). The male-to-female ratio was 1:1. The majority of the patients had primary tumors (73.7%), whereas 23.0% had metastatic disease sequenced. While IDH1 mutations occurred more frequently in White versus Black patients (20.8% vs. 5.6%; p = 0.021), FGFR2 mutations tended to be more common among Black versus White populations (27.8% vs. 16.1%; p = 0.08). Males were more likely to have TP53 mutations than females (24.3% vs. 18.2%, p = 0.016), whereas females more frequently had IDH1 (23.3% vs 16.0 %), FGFR2 (21.0% vs. 11.3%), and BAP1 (23.4% vs. 14.5%) mutations than males (all p < 0.05). Marked variations in the prevalence of other common genomic alterations in iCCA were noted across different races and sexes. CONCLUSION Distinct genomic variations exist in iCCA across race and sex. Differences in mutational profiles of iCCA patients highlight the importance of including a diverse patient population in iCCA clinical trials as well as the importance of recognizing different genetic drivers that may be targetable to treat distinct patient cohorts.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA.
| | - Hunter Stecko
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
2
|
Supradit K, Wongprasert K, Tangphatsornruang S, Yoocha T, Sonthirod C, Pootakham W, Thitapakorn V, Butthongkomvong K, Phanaksri T, Kunjantarachot A, Klongprateeppon H, Sattavacharavech P, Prasopdee S. microRNA profiling of exosomes derived from plasma and their potential as biomarkers for Opisthorchis viverrini-associated cholangiocarcinoma. Acta Trop 2024; 258:107362. [PMID: 39151716 DOI: 10.1016/j.actatropica.2024.107362] [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: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Cholangiocarcinoma (CCA) is a life-threatening disease that impacts patients worldwide. In Southeast Asian countries, the liver fluke Opisthorchis viverrini plays a major role in inducing carcinogenesis of the bile ducts. Due to its asymptomatic nature, O. viverrini infections are rarely treated, consequently leading to the development of advanced stages of CCA before diagnosis. Despite the current use of exosomal microRNAs (miRNA) as diagnostic biomarkers for the early detection of many types of cancer, the applications for miRNA remain limited with CCA. Circulating exosomes, membranous vesicles essential for intercellular communication, were found to contain unique miRNA. In this study, we conducted next-generation sequencing (Ion Torrent PGM) and bioinformatics to characterize and compare the contents of exosomal miRNA derived from the plasma of CCA patients, O. viverrini-infected patients, and healthy individuals, as well as to identify and validate key molecules as markers for screening the diagnosis of CCA and O. viverrini infection. The obtained results showed the success of using NGS technology in discovering exosomal miRNAs, specifically miR-194-5p and miR-192-5p, both of which were upregulated in the O. viverrini-infected group. Interestingly, miR-192-5p was upregulated while miR-194-5p was downregulated in CCA, suggesting their potential use as biomarkers for screening CCA and O. viverrini infection, especially in O. viverrini-endemic areas.
Collapse
Affiliation(s)
- Kittiya Supradit
- Department of Radiological technology, Faculty of Science, Ramkhamhaeng University, Bangkok, 10240, Thailand; Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kanokpan Wongprasert
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sithichoke Tangphatsornruang
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Thippawan Yoocha
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Chutima Sonthirod
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Wirulda Pootakham
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani, 12120, Thailand
| | - Veerachai Thitapakorn
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand; Research Unit in Opisthorchiasis, Cholangiocarcinoma, and Neglected Parasitic Diseases, Thammasat University, Pathum Thani, 12120, Thailand
| | | | - Teva Phanaksri
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | - Anthicha Kunjantarachot
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand
| | | | | | - Sattrachai Prasopdee
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, 12120, Thailand; Research Unit in Opisthorchiasis, Cholangiocarcinoma, and Neglected Parasitic Diseases, Thammasat University, Pathum Thani, 12120, Thailand.
| |
Collapse
|
3
|
Porreca V, Barbagallo C, Corbella E, Peres M, Stella M, Mignogna G, Maras B, Ragusa M, Mancone C. Unveil Intrahepatic Cholangiocarcinoma Heterogeneity through the Lens of Omics and Multi-Omics Approaches. Cancers (Basel) 2024; 16:2889. [PMID: 39199659 PMCID: PMC11352949 DOI: 10.3390/cancers16162889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is recognized worldwide as the second leading cause of morbidity and mortality among primary liver cancers, showing a continuously increasing incidence rate in recent years. iCCA aggressiveness is revealed through its rapid and silent intrahepatic expansion and spread through the lymphatic system leading to late diagnosis and poor prognoses. Multi-omics studies have aggregated information derived from single-omics data, providing a more comprehensive understanding of the phenomena being studied. These approaches are gradually becoming powerful tools for investigating the intricate pathobiology of iCCA, facilitating the correlation between molecular signature and phenotypic manifestation. Consequently, preliminary stratifications of iCCA patients have been proposed according to their "omics" features opening the possibility of identifying potential biomarkers for early diagnosis and developing new therapies based on personalized medicine (PM). The focus of this review is to provide new and advanced insight into the molecular pathobiology of the iCCA, starting from single- to the latest multi-omics approaches, paving the way for translating new basic research into therapeutic practices.
Collapse
Affiliation(s)
- Veronica Porreca
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Cristina Barbagallo
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Eleonora Corbella
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Marco Peres
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| | - Michele Stella
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Giuseppina Mignogna
- Department of Biochemistry Science, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Bruno Maras
- Department of Biochemistry Science, Sapienza University of Rome, 00185 Rome, Italy; (G.M.); (B.M.)
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (C.B.); (M.S.); (M.R.)
| | - Carmine Mancone
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy; (E.C.); (M.P.)
| |
Collapse
|
4
|
Demir T, Moloney C, Mahalingam D. Emerging targeted therapies and strategies to overcome resistance in biliary tract cancers. Crit Rev Oncol Hematol 2024; 199:104388. [PMID: 38754771 DOI: 10.1016/j.critrevonc.2024.104388] [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: 01/22/2024] [Revised: 04/14/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024] Open
Abstract
In the last decade, targeted therapies have shown rapid advancement in biliary tract cancer (BTC). Today, many targeted agents are available and under investigation for patients with BTC. More recently, immune checkpoint inhibitors (ICI) such as durvalumab and pembrolizumab in combination with gemcitabine plus cisplatin (gem/cis) have resulted in improved overall survival and progression-free survival in the first-line setting. However, the efficacy benefit of these novel therapeutics is often short-lived, with literature outlining concerns about both primary and secondary resistance to these agents. Investigators also need to consider toxicity profiles that can emerge using this strategy. There have been efforts to reduce evolving resistance through combinatory approaches, both pre-clinically and in early clinical settings. This review summarizes the emerging targeted therapies in BTC, evolving biomarkers of resistance, strategies to overcome them, and an analysis of ongoing clinical trials of patients with advanced BTC.
Collapse
Affiliation(s)
- Tarik Demir
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA.
| | - Carolyn Moloney
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA
| | - Devalingam Mahalingam
- Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine1, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Ferrell LD, Kakar S, Terracciano LM, Wee A. Tumours and Tumour-Like Lesions. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:842-946. [DOI: 10.1016/b978-0-7020-8228-3.00013-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Khizar H, Hu Y, Wu Y, Yang J. The role and implication of autophagy in cholangiocarcinoma. Cell Death Discov 2023; 9:332. [PMID: 37666811 PMCID: PMC10477247 DOI: 10.1038/s41420-023-01631-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor that originates from the biliary epithelial cells. It is characterized by a difficult diagnosis and limited treatment options. Autophagy is a cellular survival mechanism that maintains nutrient and energy homeostasis and eliminates intracellular pathogens. It is involved in various physiological and pathological processes, including the development of cancer. However, the role, mechanism, and potential therapeutic targets of autophagy in CCA have not been thoroughly studied. In this review, we introduce the classification, characteristics, process, and related regulatory genes of autophagy. We summarize the regulation of autophagy on the progression of CCA and collect the latest research progress on some autophagy modulators with clinical potential in CCA. In conclusion, combining autophagy modulators with immunotherapy, chemotherapy, and targeted therapy has great potential in the treatment of CCA. This combination may be a potential therapeutic target for CCA in the future.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Oncology, The Fourth Affiliated Hospital, International Institute of Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufei Hu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Gastroenterology, The Fourth School of Clinical medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanhua Wu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Gastroenterology, The Fourth School of Clinical medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, 310006, Hangzhou, Zhejiang, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, 310006, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Digestive Diseases, 310006, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Ong KH, Lai HY, Sun DP, Chen TJ, Huang SKH, Tian YF, Chou CL, Shiue YL, Chan TC, Li CF, Kuo YH. Prognostic Significance of DNA Topoisomerase II Alpha (TOP2A) in Cholangiocarcinoma. FRONT BIOSCI-LANDMRK 2023; 28:75. [PMID: 37114547 DOI: 10.31083/j.fbl2804075] [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: 12/24/2022] [Revised: 03/03/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023]
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is a malignant tumor with an increasing incidence worldwide. Although radiation therapy has improved the therapeutic efficiency of CCA treatment, differential expression of genes among cholangiocarcinoma subtypes has been revealed through precise sequencing. However, no specific molecular therapeutic targets or biomarkers have been figured out for use in precision medicine, and the exact mechanism by which antitumorigenic effects occur is still unclear. Therefore, it is necessary to conduct further studies on the development and mechanisms associated with CCA. METHODS We examined the clinical data and pathological features of patients with cholangiocarcinomas. We investigated the associations between DNA Topoisomerase II Alpha (TOP2A) expression and patient outcomes, such as metastasis-free survival (MFS) and disease-specific survival (DSS), as well as clinical characteristics and pathological results. RESULTS TOP2A expression was shown to be upregulated in CCA tissue sections by immunohistochemistry staining and data mining. Moreover, we observed that the TOP2A expression correlated with clinical features, such as the primary tumor stage, histological variants, and patients with hepatitis. Furthermore, high expression of TOP2A was associated with worse survival outcomes in terms of the overall survival (p < 0.0001), disease-specific survival (p < 0.0001), and metastasis-free survival (p < 0.0001) compared with patients in the low TOP2A expression group. This indicates that a high level of TOP2A expression is related to an unfavorable prognosis. CONCLUSIONS Our results show that TOP2A is highly expressed in CCA tissues, and its upregulation is correlated with the primary disease stage and poor prognosis significantly. Consequently, TOP2A is a prognostic biomarker and a novel therapeutic target for the treatment of CCA.
Collapse
Affiliation(s)
- Khaa Hoo Ong
- Division of Gastroenterology & General Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
- Department of Medical Technology, Chung Hwa University of Medical Technology, 717 Tainan, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
| | - Hong-Yue Lai
- Department of Pharmacology, School of Medicine, China Medical University, 404333 Taichung, Taiwan
| | - Ding-Ping Sun
- Division of Gastroenterology & General Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
| | - Tzu-Ju Chen
- Department of Medical Technology, Chung Hwa University of Medical Technology, 717 Tainan, Taiwan
- Department of Clinical Pathology, Chi Mei Medical Center, 710 Tainan, Taiwan
| | - Steven Kuan-Hua Huang
- Division of Urology, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, 711 Tainan, Taiwan
| | - Yu-Feng Tian
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
| | - Chia-Lin Chou
- Department of Medical Technology, Chung Hwa University of Medical Technology, 717 Tainan, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chi Mei Medical Center, 710 Tainan, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Sciences, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
- Institute of Precision Medicine, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, 710 Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, 704 Tainan, Taiwan
| | - Chien-Feng Li
- Institute of Precision Medicine, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
- Department of Medical Research, Chi Mei Medical Center, 710 Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, 704 Tainan, Taiwan
- Trans-Omic Laboratory for Precision Medicine, Chi Mei Medical Center, 710 Tainan, Taiwan
| | - Yu-Hsuan Kuo
- Institute of Biomedical Sciences, National Sun Yat-sen University, 804 Kaohsiung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, 71004 Tainan, Taiwan
- College of Pharmacy and Science, Chia Nan University, 71710 Tainan, Taiwan
| |
Collapse
|
8
|
Choi WJ, Ivanics T, Gravely A, Gallinger S, Sapisochin G, O'Kane GM. Optimizing Circulating Tumour DNA Use in the Perioperative Setting for Intrahepatic Cholangiocarcinoma: Diagnosis, Screening, Minimal Residual Disease Detection and Treatment Response Monitoring. Ann Surg Oncol 2023; 30:3849-3863. [PMID: 36808320 DOI: 10.1245/s10434-023-13126-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/23/2023]
Abstract
In this review, we present the current evidence and future perspectives on the use of circulating tumour DNA (ctDNA) in the diagnosis, management and understanding the prognosis of patients with intrahepatic cholangiocarcinoma (iCCA) undergoing surgery. Liquid biopsies or ctDNA maybe utilized to: (1) determine the molecular profile of the tumour and therefore guide the selection of molecular targeted therapy in the neoadjuvant setting, (2) form a surveillance tool for the detection of minimal residual disease or cancer recurrence after surgery, and (3) diagnose and screen for early iCCA detection in high-risk populations. The potential for ctDNA can be tumour-informed or -uninformed depending on the goals of its use. Future studies will require ctDNA extraction technique validations, with standardizations of both the platforms and the timing of ctDNA collections.
Collapse
Affiliation(s)
- Woo Jin Choi
- HBP and Multi Organ Transplant Program, Division of General Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada.,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.,HPB Surgical Oncology, University Health Network, Toronto, Ontario, Canada
| | - Tommy Ivanics
- Department of Surgery, Henry Ford Hospital, Detroit, MI, USA.,Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Annabel Gravely
- HPB Surgical Oncology, University Health Network, Toronto, Ontario, Canada
| | - Steven Gallinger
- HBP and Multi Organ Transplant Program, Division of General Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada.,HPB Surgical Oncology, University Health Network, Toronto, Ontario, Canada
| | - Gonzalo Sapisochin
- HBP and Multi Organ Transplant Program, Division of General Surgery, Department of Surgery, University Health Network, University of Toronto, Toronto, Ontario, Canada. .,HPB Surgical Oncology, University Health Network, Toronto, Ontario, Canada.
| | - Grainne M O'Kane
- Department of Medical Oncology, Trinity St. James's Cancer Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
9
|
Brown ZJ, Patwardhan S, Bean J, Pawlik TM. Molecular diagnostics and biomarkers in cholangiocarcinoma. Surg Oncol 2022; 44:101851. [PMID: 36126350 DOI: 10.1016/j.suronc.2022.101851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/26/2022] [Accepted: 09/09/2022] [Indexed: 10/14/2022]
Abstract
Regardless of anatomic origin, cholangiocarcinoma is generally an aggressive malignancy with a relatively high case fatality. Surgical resection with curative intent remains the best opportunity to achieve meaningful long-term survival. Most patients present, however, with advanced disease and less than 20% of patients are candidates for surgical resection. Unfortunately, even patients who undergo resection have a 5-year survival that ranges from 20 to 40%. Biomarkers are indicators of normal, pathologic, or biologic responses to an intervention and can range from a characteristic (i.e., blood pressure reading which can detect hypertension) to specific genetic mutations or proteins (i.e., carcinoembryonic antigen level). Novel biomarkers and improved molecular diagnostics represent an attractive opportunity to improve detection as well as to identify novel therapeutic targets for patients with cholangiocarcinoma. We herein review the latest advances in molecular diagnostics and biomarkers related to the early detection and treatment of patients with cholangiocarcinoma.
Collapse
Affiliation(s)
- Zachary J Brown
- Department of Surgery, The State Wexner Medical Center, Columbus, OH, USA.
| | - Satyajit Patwardhan
- Dept of HPB Surgery and Liver Transplantation, Global Hospital, Mumbai, India
| | - Joal Bean
- Department of Surgery, The State Wexner Medical Center, Columbus, OH, USA
| | - Timothy M Pawlik
- Department of Surgery, The State Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
10
|
Carotenuto M, Sacco A, Forgione L, Normanno N. Genomic alterations in cholangiocarcinoma: clinical significance and relevance to therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:200-223. [PMID: 36046845 PMCID: PMC9400790 DOI: 10.37349/etat.2022.00079] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/22/2022] [Indexed: 11/22/2022] Open
Abstract
Improving the survival of patients with cholangiocarcinoma (CCA) has long proved challenging, although the treatment of this disease nowadays is on advancement. The historical invariability of survival outcomes and the limited number of agents known to be effective in the treatment of this disease has increased the number of studies designed to identify genetic targetable hits that can be efficacious for novel therapies. In this respect, the increasing feasibility of molecular profiling starting either from tumor tissue or circulating cell-free DNA (cfDNA) has led to an increased understanding of CCA biology. Intrahepatic CCA (iCCA) and extrahepatic CCA (eCCA) display different and typical patterns of actionable genomic alterations, which offer opportunity for therapeutic intervention. This review article will summarize the current knowledge on the genomic alterations of iCCA and eCCA, provide information on the main technologies for genomic profiling using either tumor tissue or cfDNA, and briefly discuss the main clinical trials with targeted agents in this disease.
Collapse
Affiliation(s)
- Marianeve Carotenuto
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Laura Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| |
Collapse
|
11
|
DNA Damage Response Inhibitors in Cholangiocarcinoma: Current Progress and Perspectives. Cells 2022; 11:cells11091463. [PMID: 35563769 PMCID: PMC9101358 DOI: 10.3390/cells11091463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/24/2022] [Indexed: 12/27/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a poorly treatable type of cancer and its incidence is dramatically increasing. The lack of understanding of the biology of this tumor has slowed down the identification of novel targets and the development of effective treatments. Based on next generation sequencing profiling, alterations in DNA damage response (DDR)-related genes are paving the way for DDR-targeting strategies in CCA. Based on the notion of synthetic lethality, several DDR-inhibitors (DDRi) have been developed with the aim of accumulating enough DNA damage to induce cell death in tumor cells. Observing that DDRi alone could be insufficient for clinical use in CCA patients, the combination of DNA-damaging regimens with targeted approaches has started to be considered, as evidenced by many emerging clinical trials. Hence, novel therapeutic strategies combining DDRi with patient-specific targeted drugs could be the next level for treating cholangiocarcinoma.
Collapse
|
12
|
Díaz-Muñoz M, Hernández-Muñoz R, Butanda-Ochoa A. Structure-activity features of purines and their receptors: implications in cell physiopathology. MOLECULAR BIOMEDICINE 2022; 3:5. [PMID: 35079944 PMCID: PMC8789959 DOI: 10.1186/s43556-022-00068-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/19/2022] [Indexed: 11/21/2022] Open
Abstract
The purine molecular structure consists of fused pyrimidine and imidazole rings. Purines are main pieces that conform the structure of nucleic acids which rule the inheritance processes. Purines also work as metabolic intermediates in different cell functions and as messengers in the signaling pathways throughout cellular communication. Purines, mainly ATP and adenosine (ADO), perform their functional and pharmacological properties because of their structural/chemical characteristics that make them either targets of mutagenesis, mother frameworks for designing molecules with controlled effects (e.g. anti-cancer), or chemical donors (e.g., of methyl groups, which represent a potential chemoprotective action against cancer). Purines functions also come from their effect on specific receptors, channel-linked and G-protein coupled for ATP, and exclusively G-coupled receptors for ADO (also known as ADORAs), which are involved in cell signaling pathways, there, purines work as chemical messengers with autocrine, paracrine, and endocrine actions that regulate cell metabolism and immune response in tumor progression which depends on the receptor types involved in these signals. Purines also have antioxidant and anti-inflammatory properties and participate in the cell energy homeostasis. Therefore, purine physiology is important for a variety of functions relevant to cellular health; thus, when these molecules present a homeostatic imbalance, the stability and survival of the cellular systems become compromised.
Collapse
Affiliation(s)
- Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular Y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM, Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - Rolando Hernández-Muñoz
- Departamento de Biología Celular Y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Ciudad Universitaria/Circuito Exterior, C.P. 04510, Ciudad de México, México
| | - Armando Butanda-Ochoa
- Departamento de Biología Celular Y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, UNAM, Ciudad Universitaria/Circuito Exterior, C.P. 04510, Ciudad de México, México.
| |
Collapse
|
13
|
Yang R, Wang D, Han S, Gu Y, Li Z, Deng L, Yin A, Gao Y, Li X, Yu Y, Wang X. MiR-206 suppresses the deterioration of intrahepatic cholangiocarcinoma and promotes sensitivity to chemotherapy by inhibiting interactions with stromal CAFs. Int J Biol Sci 2022; 18:43-64. [PMID: 34975317 PMCID: PMC8692143 DOI: 10.7150/ijbs.62602] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Intrahepatic cholangiocarcinoma (iCCA) is a highly malignant subtype of cholangiocarcinoma (CCA) with poor prognosis. In iCCA, the interplay between the stroma and tumor cells results in resistance to adjuvant chemotherapy. Increasing evidence indicates that miR-206 participates in tumor progression, but its role in iCCA is still unclear. The aim of this study was to identify dysregulated miR-206 expression in iCCA and to further explore the underlying mechanism. Methods: MiR-206 expression was proven to be downregulated in iCCA tissues by qPCR, and its correlation with clinical characteristics and prognosis was investigated. iCCA-derived cancer-associated fibroblast cells (CAFs) and normal fibroblast cells (NFs) were isolated and identified. MiR-206 was knocked in or down in CAFs and CCA cells, respectively, to explore the role of miR-206, and coculture of these treated CCAs and CAFs was conducted to explore the effects of miR-206 on their mutual promoting effects. Exosomes carrying miR-206 and an orthotopic mouse model were used to determine the inhibitory effects of miR-206 on iCCA deterioration in vivo. Results: We confirmed that miR-206 is a suppressor of iCCA. Overexpressing miR-206 in CCA cells inhibited cell proliferation, migration and invasion. When cocultured with CCA cells, NFs downregulated miR-206 expression, and NFs were susceptible to transforming into CAFs. Moreover, CAFs promoted CCA cell malignant behaviors and gemcitabine resistance. Overexpressing miR-206 in CAFs or CCA cells inhibited this mutual promoting effect. Additionally, when delivered by exosomes, miR-206 suppressed tumor deterioration. And combined with gemcitabine, this treatment resulted in a longer survival time. Conclusion: Our study explained that the interaction between CCA cells and CAFs promoted iCCA deterioration. As a suppressive factor, miR-206 inhibited aggressive characteristics and gemcitabine resistance by interfering with this mutual promoting effect. This research elucidated the molecular mechanism underlying the unfavorable chemotherapeutic response of patients with iCCA, which provided a promising target for iCCA treatment.
Collapse
Affiliation(s)
- Renjie Yang
- School of Medicine, Southeast University, Nanjing, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Dong Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Shen Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yichao Gu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Zhi Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Lei Deng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Aihong Yin
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yun Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Yue Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| | - Xuehao Wang
- School of Medicine, Southeast University, Nanjing, China
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, Jiangsu Province, China
| |
Collapse
|
14
|
Gruttadauria S, Barbera F, Pagano D, Liotta R, Miraglia R, Barbara M, Bavetta MG, Cammà C, Petridis I, Di Carlo D, Conaldi PG, Di Francesco F. Liver Transplantation for Unresectable Intrahepatic Cholangiocarcinoma: The Role of Sequencing Genetic Profiling. Cancers (Basel) 2021; 13:6049. [PMID: 34885159 PMCID: PMC8657183 DOI: 10.3390/cancers13236049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/20/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is a rare and aggressive primary liver tumor, characterized by a range of different clinical manifestations and by increasing incidence and mortality rates even after curative treatment with radical resection. In recent years, growing attention has been devoted to this disease and some evidence supports liver transplantation (LT) as an appropriate treatment for intrahepatic cholangiocarcinoma; evolving work has also provided a framework for better understanding the genetic basis of this cancer. The aim of this study was to provide a clinical description of our series of patients complemented with Next-Generation Sequencing genomic profiling. From 1999 to 2021, 12 patients who underwent LT with either iCCA or a combined hepatocellular and cholangiocellular carcinoma (HCC-iCCA) were included in this study. Mutations were observed in gene activating signaling pathways known to be involved with iCCA tumorigenesis (KRAS/MAPK, P53, PI3K-Akt/mTOR, cAMP, WNT, epigenetic regulation and chromatin remodeling). Among several others, a strong association was observed between the Notch pathway and tumor size (point-biserial rhopb = 0.93). Our results are suggestive of the benefit potentially derived from molecular analysis to improve our diagnostic capabilities and to devise new treatment protocols, and eventually ameliorate long-term survival of patients affected by iCCA or HCC-iCCA.
Collapse
Affiliation(s)
- Salvatore Gruttadauria
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (D.P.); (I.P.); (F.D.F.)
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Floriana Barbera
- Laboratorio di Patologia Clinica, Microbiologia e Virologia, Dipartimento di Medicina di Laboratorio e Biotecnologie Avanzate, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (F.B.); (D.D.C.)
| | - Duilio Pagano
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (D.P.); (I.P.); (F.D.F.)
| | - Rosa Liotta
- Pathology Unit, Department of Diagnostic and Therapeutic Services, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy;
| | - Roberto Miraglia
- Radiology Service, Department of Diagnostic and Therapeutic Services, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy;
| | - Marco Barbara
- Research Department, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (M.B.); (P.G.C.)
| | - Maria Grazia Bavetta
- Unit of Hepatic Oncology, Division of Internal Medicine 2, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, 90146 Palermo, Italy;
| | - Calogero Cammà
- Section of Gastroenterology & Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy;
| | - Ioannis Petridis
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (D.P.); (I.P.); (F.D.F.)
| | - Daniele Di Carlo
- Laboratorio di Patologia Clinica, Microbiologia e Virologia, Dipartimento di Medicina di Laboratorio e Biotecnologie Avanzate, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (F.B.); (D.D.C.)
| | - Pier Giulio Conaldi
- Research Department, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (M.B.); (P.G.C.)
| | - Fabrizio Di Francesco
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico-Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione (IRCCS-ISMETT), University of Pittsburgh Medical Center Italy (UPMC Italy), 90127 Palermo, Italy; (D.P.); (I.P.); (F.D.F.)
| |
Collapse
|
15
|
Tóth M, Wehling L, Thiess L, Rose F, Schmitt J, Weiler SME, Sticht C, De La Torre C, Rausch M, Albrecht T, Grabe N, Duwe L, Andersen JB, Köhler BC, Springfeld C, Mehrabi A, Kulu Y, Schirmacher P, Roessler S, Goeppert B, Breuhahn K. Co-expression of YAP and TAZ associates with chromosomal instability in human cholangiocarcinoma. BMC Cancer 2021; 21:1079. [PMID: 34615513 PMCID: PMC8496054 DOI: 10.1186/s12885-021-08794-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/16/2021] [Indexed: 12/25/2022] Open
Abstract
Background Activation of the oncogene yes-associated protein (YAP) is frequently detected in intrahepatic cholangiocarcinoma (iCCA); however, the expression pattern and the functional impact of its paralogue WW domain-containing transcription regulator 1 (WWTR1; synonym: TAZ) are not well described in different CCA subtypes. Methods Immunohistochemical analysis of YAP and TAZ in iCCA and extrahepatic CCA (eCCA) cohorts was performed. YAP/TAZ shuttling and their functional impact on CCA cell lines were investigated. Target genes expression after combined YAP/TAZ inhibition was analyzed. Results Immunohistochemical analysis of iCCA and eCCA revealed YAP or TAZ positivity in up to 49.2%; however, oncogene co-expression was less frequent (up to 23%). In contrast, both proteins were jointly detectable in most CCA cell lines and showed nuclear/cytoplasmic shuttling in a cell density-dependent manner. Next to the pro-proliferative function of YAP/TAZ, both transcriptional co-activators cooperated in the regulation of a gene signature that indicated the presence of chromosomal instability (CIN). A correlation between YAP and the CIN marker phospho-H2A histone family member X (pH2AX) was particularly observed in tissues from iCCA and distal CCA (dCCA). The presence of the CIN genes in about 25% of iCCA was statistically associated with worse prognosis. Conclusions YAP and TAZ activation is not uncoupled from cell density in CCA cells and both factors cooperatively contribute to proliferation and expression of CIN-associated genes. The corresponding group of CCA patients is characterized by CIN and may benefit from YAP/TAZ-directed therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08794-5.
Collapse
Affiliation(s)
- Marcell Tóth
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Lilija Wehling
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany.,Centre for Organismal Studies/BioQuant, Heidelberg University, Heidelberg, Germany
| | - Lena Thiess
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Fabian Rose
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Jennifer Schmitt
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Sofia M E Weiler
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Carolina De La Torre
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Melina Rausch
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Thomas Albrecht
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Niels Grabe
- Hamamatsu Tissue Imaging and Analysis Center (TIGA), BioQuant, Heidelberg University, Heidelberg, Germany
| | - Lea Duwe
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bruno C Köhler
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany;, Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Springfeld
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany;, Liver Cancer Center Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Arianeb Mehrabi
- Department of General Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Yakup Kulu
- Department of General Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Safarpour AR, Askari H, Ejtehadi F, Azarnezhad A, Raeis-Abdollahi E, Tajbakhsh A, Abazari MF, Tarkesh F, Shamsaeefar A, Niknam R, Sivandzadeh GR, Lankarani KB, Ejtehadi F. Cholangiocarcinoma and liver transplantation: What we know so far? World J Gastrointest Pathophysiol 2021; 12:84-105. [PMID: 34676129 PMCID: PMC8481789 DOI: 10.4291/wjgp.v12.i5.84] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/28/2021] [Accepted: 08/11/2021] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a type of cancer with increasing prevalence around the world that originates from cholangiocytes, the epithelial cells of the bile duct. The tumor begins insidiously and is distinguished by high grade neoplasm, poor outcome, and high risk for recurrence. Liver transplantation has become broadly accepted as a treatment option for CCA. Liver transplantation is expected to play a crucial role as palliative and curative therapy for unresectable hilar CCA and intrahepatic CCA. The purpose of this study was to determine which cases with CCA should be subjected to liver transplantation instead of resection, although reported post-transplant recurrence rate averages approximately 20%. This review also aims to highlight the molecular current frontiers of CCA and directions of liver transplantation for CCA.
Collapse
Affiliation(s)
- Ali Reza Safarpour
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Hassan Askari
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Farshid Ejtehadi
- The Princess Alexandra Hospital HNS Trust, Harlow, Essex CM20 1QX, United Kingdom
| | - Asaad Azarnezhad
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj 6617913446, Iran
| | - Ehsan Raeis-Abdollahi
- Department of Basic Medical Sciences, Qom Medical Branch, Islamic Azad University, Qom, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Mohammad Foad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran 1417653761, Iran
| | - Firoozeh Tarkesh
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Alireza Shamsaeefar
- Shiraz Organ Transplant Center, Shiraz University of Medical Sciences, Shiraz 7193711351, Iran
| | - Ramin Niknam
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | - Gholam Reza Sivandzadeh
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| | | | - Fardad Ejtehadi
- Department of Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran
| |
Collapse
|
17
|
Xu X. Diagnosis of hilar cholangiocarcinoma using intravoxel incoherent motion diffusion-weighted magnetic resonance imaging. Abdom Radiol (NY) 2021; 46:3159-3167. [PMID: 33660039 DOI: 10.1007/s00261-021-02997-x] [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: 11/30/2020] [Revised: 02/02/2021] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To investigate the value of intravoxel incoherent motion diffusion-weighed magnetic resonance imaging (IVIM-DWI) in discriminating the pathological grades of hilar cholangiocarcinoma (HC). PATIENTS AND METHODS Thirty-seven HC patients were enrolled and received routine and advanced DWI scanning with multiple b-values. IVIM-DWI images were obtained using echo-planar imaging sequence. RESULTS The consistency of the maximum cross-sectional area ROI measuring method was higher than that of the repeated sampling ROI measuring method. ADCslow values were closely correlated with the pathological grades of HC. The degrees of biliary dilatation and MELD scores had no influence on the negative correlation between ADCslow values and the pathological degrees of HC patients. CONCLUSIONS ADCslow values could be applied in indicating the pathological grades of HC, which was independent on the extent of biliary dilatation.
Collapse
|
18
|
Cadamuro M, Lasagni A, Lamarca A, Fouassier L, Guido M, Sarcognato S, Gringeri E, Cillo U, Strazzabosco M, Marin JJ, Banales JM, Fabris L. Targeted therapies for extrahepatic cholangiocarcinoma: preclinical and clinical development and prospects for the clinic. Expert Opin Investig Drugs 2021; 30:377-388. [PMID: 33622120 DOI: 10.1080/13543784.2021.1880564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Until recently, cholangiocarcinoma (CCA) was a largely overlooked disease, and among CCAs, extrahepatic CCA (eCCA) was even more neglected. Despite the growing impact of molecularly targeted therapies and immunotherapy, prognosis of eCCA is dismal. Therefore, unraveling the complex molecular landscape of eCCA has become an urgent need. Deep phenotyping studies have revealed that eCCA is a heterogeneous tumor, harboring specific alterations categorizable into four classes, 'Mesenchymal', 'Proliferation', 'Immune', 'Metabolic'. Molecular alterations convey the activation of several pro-oncogenic pathways, where either actionable drivers or outcome predictors can be identified.Areas covered: We offer insights on perturbed pathways, molecular profiling, and actionable targets in eCCA and present a perspective on the potential stepping-stones to future progress. A systematic literature search in PubMed/ClinicalTrials.gov websites was performed by authors from different disciplines according to their specific topic knowledge to identify the newest and most relevant advances in precision medicine of eCCA.Expert opinion: eCCA is a distinct entity with unique features in terms of molecular classes, oncogenic drivers, and tumor microenvironment. Since more prevalent mutations are currently undruggable, and immunotherapy can be offered only to a minority of patients, international collaborations are instrumental to improve the understanding of the molecular underpins of this disease.
Collapse
Affiliation(s)
- Massimiliano Cadamuro
- Department of Molecular Medicine (DMM), University of Padua, Padua. Italy.,International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy
| | - Alberto Lasagni
- Division of General Medicine, Padua University-Hospital, Padua, Italy
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation, Manchester, United Kingdom.,Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, Treviso, Italy.,Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Enrico Gringeri
- Hepatobiliary Surgery and Liver Transplantation, Padua University-Hospital, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Umberto Cillo
- Hepatobiliary Surgery and Liver Transplantation, Padua University-Hospital, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy
| | - Mario Strazzabosco
- International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy.,Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US
| | - Jose Jg Marin
- Experimental Hepatology and Drug Targeting (HEVEPHARM), IBSAL, CIBERehd, University of Salamanca, Salamanca, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute - Donostia University Hospital -, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Luca Fabris
- Department of Molecular Medicine (DMM), University of Padua, Padua. Italy.,International Center for Digestive Health (ICDH), University of Milan-Bicocca, Milan, Italy.,Division of General Medicine, Padua University-Hospital, Padua, Italy.,Digestive Disease Section, Liver Center, Yale University, New Haven, CT, US
| |
Collapse
|
19
|
Crispo F, Pietrafesa M, Condelli V, Maddalena F, Bruno G, Piscazzi A, Sgambato A, Esposito F, Landriscina M. IDH1 Targeting as a New Potential Option for Intrahepatic Cholangiocarcinoma Treatment-Current State and Future Perspectives. Molecules 2020; 25:molecules25163754. [PMID: 32824685 PMCID: PMC7464324 DOI: 10.3390/molecules25163754] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma is a primary malignancy of the biliary tract characterized by late and unspecific symptoms, unfavorable prognosis, and few treatment options. The advent of next-generation sequencing has revealed potential targetable or actionable molecular alterations in biliary tumors. Among several identified genetic alterations, the IDH1 mutation is arousing interest due to its role in epigenetic and metabolic remodeling. Indeed, some IDH1 point mutations induce widespread epigenetic alterations by means of a gain-of-function of the enzyme, which becomes able to produce the oncometabolite 2-hydroxyglutarate, with inhibitory activity on α-ketoglutarate-dependent enzymes, such as DNA and histone demethylases. Thus, its accumulation produces changes in the expression of several key genes involved in cell differentiation and survival. At present, small-molecule inhibitors of IDH1 mutated enzyme are under investigation in preclinical and clinical phases as promising innovative treatments for IDH1-mutated intrahepatic cholangiocarcinomas. This review examines the molecular rationale and the results of preclinical and early-phase studies on novel pharmacological agents targeting mutant IDH1 in cholangiocarcinoma patients. Contextually, it will offer a starting point for discussion on combined therapies with metabolic and epigenetic drugs, to provide molecular support to target the interplay between metabolism and epigenetics, two hallmarks of cancer onset and progression.
Collapse
Affiliation(s)
- Fabiana Crispo
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
| | - Michele Pietrafesa
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
| | - Valentina Condelli
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
| | - Francesca Maddalena
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
| | - Giuseppina Bruno
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (G.B.); (A.P.)
| | - Annamaria Piscazzi
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (G.B.); (A.P.)
| | - Alessandro Sgambato
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
| | - Franca Esposito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: (F.E.); (M.L.); Tel.: +39-081-746-3145 (F.E.); +39-088-173-6426 (M.L.)
| | - Matteo Landriscina
- Laboratory of Pre-Clinical and Translational Research, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (PZ), Italy; (F.C.); (M.P.); (V.C.); (F.M.); (A.S.)
- Medical Oncology Unit, Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy; (G.B.); (A.P.)
- Correspondence: (F.E.); (M.L.); Tel.: +39-081-746-3145 (F.E.); +39-088-173-6426 (M.L.)
| |
Collapse
|
20
|
Passeri MJ, Baimas-George MR, Sulzer JK, Iannitti DA, Martinie JB, Baker EH, Ocuin LM, Vrochides D. Prognostic impact of the Bismuth-Corlette classification: Higher rates of local unresectability in stage IIIb hilar cholangiocarcinoma. Hepatobiliary Pancreat Dis Int 2020; 19:157-162. [PMID: 32088126 DOI: 10.1016/j.hbpd.2020.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/01/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The Bismuth-Corlette (BC) classification is used to categorize hilar cholangiocarcinoma by proximal extension along the biliary tree. As the right hepatic artery crosses just behind the left bile duct, we hypothesized that BC IIIb tumors would have a higher likelihood of local unresectability due to involvement of the contralateral artery. METHODS A retrospective review of a prospectively maintained database identified patients with hilar cholangiocarcinoma taken to the operating room for intended curative resection between April 2008 and September 2016. Cases were assigned BC stages based on preoperative imaging. RESULTS Sixty-eight patients were included in the study. All underwent staging laparoscopy after which 16 cases were aborted for metastatic disease. Of the remaining 52 cases, 14 cases were explored and aborted for locally advanced disease. Thirty-eight underwent attempt at curative resection. After excluding cases aborted for metastatic disease, the chance of proceeding with resection was 55.6% for BC IIIb staged lesions compared to 80.0% of BC IIIa lesions and to 82.4% for BC I-IIIa staged lesions (P < 0.05). About 44.4% of BC IIIb lesions were aborted for locally advanced disease versus 17.6% of remaining BC stages. CONCLUSIONS When hilar cholangiocarcinoma is preoperatively staged as BC IIIb, surgeons should anticipate higher rates of locally unresectable disease, likely involving the right hepatic artery.
Collapse
Affiliation(s)
- Michael J Passeri
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - Maria R Baimas-George
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - Jesse K Sulzer
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - David A Iannitti
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - John B Martinie
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - Erin H Baker
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - Lee M Ocuin
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA
| | - Dionisios Vrochides
- Division of HPB Surgery, Department of Surgery, Carolinas Medical Center, 1025 Morehead Medical Dr., Suite 600, Charlotte, NC 28204, USA.
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Cholangiocarcinoma (CCA) are heterogeneous tumors that arise from the malignant transformation of cholangiocytes along the biliary tree. CCA heterogeneity occurs at multiple levels and results in resistance to therapy and poor prognosis. Here, we review the molecular classification of CCA by focusing on the latest progresses based on genetic, epigenetic, transcriptomic and proteomic profiles. In addition, we introduce the emerging field of radiogenomics. RECENT FINDINGS Genome-wide integrative omics approaches have been widely reported by using large cohorts of CCA patients. Morphomolecular correlations have been established, including enrichment of FGFR2 gene fusions and IDH1/2 mutations in iCCA. A specific IDH mutant iCCA subtype displays high mitochondrial and low chromatin modifier expression linked to ARID1A promoter hypermethylation. Examples of translation of these classifications for the management of CCA have also been reported, with prediction of drug efficacy based on genetic alterations. SUMMARY Although there is currently no international consensus on CCA morphomolecular classification, the recent initiatives developed under the umbrella of The European Network for the Study of Cholangiocarcinoma (ENSCCA) should favor new collaborative research. Identifying distinct molecular subgroups and developing appropriate targeted therapies will improve the clinical outcome of patients with CCA.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW To give a state-of-art knowledge regarding cancer-associated fibroblasts (CAF) in cholangiocarcinoma (CCA) based both on direct evidence and studies on other desmoplastic cancers. High contingency of CAF characterizes CCA, a tumor with a biliary epithelial phenotype that can emerge anywhere in the biliary tree. Current treatments are very limited, the surgical resection being the only effective treatment but restricted to a minority of patients, whereas the remaining patients undergo palliative chemotherapy regimens. In cancer, CAF shape the tumor microenvironment, drive cancer growth and progression, and contribute to drug resistance. All these functions are accomplished through an interplay network between CAF and surrounding cells including tumor and other stromal cells, i.e. immune and endothelial cells. RECENT FINDINGS Several studies have pointed out the existence of CAF sub-populations carrying out several and opposite functions, cancer-promoting or cancer-restraining as shown in pancreatic cancer, another prototypic desmoplastic tumor in which heterogeneity of CAF is well demonstrated. SUMMARY New CAF functions are now emerging in pancreatic and breast cancers like the modulation of immune responses or tumor metabolism, opening new area for treatments.
Collapse
|
23
|
Taghizadeh H, Müllauer L, Mader R, Prager GW. Applied precision cancer medicine in metastatic biliary tract cancer. Hepatol Int 2020; 14:288-295. [PMID: 32100259 PMCID: PMC7136181 DOI: 10.1007/s12072-020-10020-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/28/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Advanced therapy-refractory biliary tract cancer (BTC) has poor prognosis and constitutes a major challenge for adequate treatment strategies. By mapping the molecular profiles of advanced BTC patients, precision cancer medicine may provide targeted therapies for these patients. OBJECTIVE In this analysis, we aimed to show the potential of PCM in metastatic BTC. METHODS In this single-center, real-world retrospective analysis of our PCM platform, we describe the molecular profiling of 30 patients diagnosed with different types of metastatic BTC. Tumor samples of the patients were examined using a 161-gene next-generation sequencing panel, immunohistochemistry (IHC), and fluorescence in situ hybridization for chromosomal translocations. RESULTS In total, we identified 35 molecular aberrations in 30 patients. The predominant mutations were KRAS (n = 8), TP53 (n = 7), IDH2 (n = 4), and IDH1 (n = 3) that accounted for the majority of all molecular alterations (62.86%). BRAF mutations were observed in two patients. Less frequent alterations were noted in ARID1A, CTNNB1, ESR1, FBXW7, FGFR2, MET, NOTCH2, PIK3CA, PTCH1, SMAD4, and SRC1, each in one case. FGFR fusion gene was detected in one patient. No mutations were detected in eight patients. IHC revealed EGFR and p-mTOR expression in 28 patients. Applying these results to our patients, targeted therapy was recommended for 60% of the patients (n = 18). One patient achieved stable disease. CONCLUSIONS PCM is a feasible treatment approach and may provide molecular-guided therapy recommendations for metastatic BTC.
Collapse
Affiliation(s)
- H Taghizadeh
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.,Comprehensive Cancer Center Vienna, Vienna, Austria
| | - L Müllauer
- Clinical Institute of Pathology, Medical University Vienna, Vienna, Austria
| | - R Mader
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.,Comprehensive Cancer Center Vienna, Vienna, Austria
| | - G W Prager
- Department of Medicine I, Clinical Division of Oncology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria. .,Comprehensive Cancer Center Vienna, Vienna, Austria.
| |
Collapse
|
24
|
Pu XH, Yue S, Wu HY, Yang J, Fan XS, Fu Y, Ye Q, Chen J. C-MET in intrahepatic cholangiocarcinoma: High-Frequency amplification predicts protein expression and a unique molecular subtype. Pathol Res Pract 2020; 216:152857. [PMID: 32089411 DOI: 10.1016/j.prp.2020.152857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/10/2020] [Accepted: 02/10/2020] [Indexed: 12/16/2022]
Abstract
As an increasing number of gene alterations have been discovered in intrahepatic cholangiocarcinoma (ICC), molecular targets are promising for the diagnosis and treatment of distinct subpopulations carrying unique molecular signatures. C-MET amplification is associated with a variety of tumors, including ICC; however, the characteristics of this alteration have not been assessed in ICC. By determining the ratios of C-MET/chromosome enumeration probe (CEP) 7 double-colour probes, we evaluated the presence of C-MET amplification in a cohort of 133 ICC tumors by fluorescence in situ hybridization (FISH). We further determined the levels of MET protein expression by immunohistochemistry (IHC) and analyzed clinicopathologic records. Of the samples, 21 (15.8 %) had high-frequency and 41 (30.8 %) had low-frequency C-MET genetic amplification, and 71 (53.4 %) had a normal C-MET gene. There were significant differences in gross classification (p = 0.045), microscopic cholangitis (p = 0.030), mucus level in tumors (p = 0.012) and T stage (p = 0.007) between the three groups. When we combined high-frequency and low-frequency amplifications of C-MET into one group, only microscopic cholangitis (p = 0.010) and stage (p = 0.016) showed significant differences compared to normal C-MET gene expression. However, when we combined the low-frequency C-MET amplification group with the normal C-MET group and compared this combined group with the high-frequency C-MET amplification group, the high-frequency group had more younger patients (p = 0.047), had more non-mass-forming (MF)-type cases according to gross classification (p = 0.015), secreted more mucus (p = 0.002) and appeared to have a higher T stage (p = 0.031) than the combined group. For IHC results, although only cluster C-MET amplification predicted protein overexpression, high-frequency amplification was associated with more protein expression than the other genetic statuses (p = 0.000). As low-frequency C-MET amplification exhibited similar biology to that of the normal gene, we regarded high-frequency amplification of C-MET as a unique molecular subtype. It may play important roles in tumor progression and may be used as a prognostic marker for targeted therapy.
Collapse
Affiliation(s)
- Xiao-Hong Pu
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Shen Yue
- Department of Medical Genetics, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Hong-Yan Wu
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Jun Yang
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Xiang-Shan Fan
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Yao Fu
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Qing Ye
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China.
| | - Jun Chen
- Departments of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China.
| |
Collapse
|
25
|
Guo L, Zhang Y, Yin Z, Ji Y, Yang G, Qian B, Li S, Wang J, Liang T, Li C, Li X. Screening and identification of genes associated with cell proliferation in cholangiocarcinoma. Aging (Albany NY) 2020; 12:2626-2646. [PMID: 32040444 PMCID: PMC7041743 DOI: 10.18632/aging.102766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/12/2020] [Indexed: 11/25/2022]
Abstract
Cholangiocarcinoma (CCA), an aggressive tumor with poor prognosis, is a malignant cancer with increasing incidence and mortality rates. It is important to survey crucial genes in CCA to find and design potential drug targets, especially for those genes associated with cell proliferation that is a key biological process in tumorgenesis. Herein, we surveyed genes associated with cell proliferation via a comprehensive pan-cancer analysis. Candidate genes were further analyzed using multiple approaches, including cross-analysis from diverse molecular levels, examination of potential function and interactions, and additional experimental validation. We primarily screened 15 potential genes based on 11 validated genes, and these 26 genes were further examined to delineate their biological functions and potential roles in cancer treatment. Several of them were involved synthetically lethal genetic interactions, especially for RECQL4, TOP2A, MKI67 and ASPM, indicating their potential roles in drug design and cancer treatment. Further experimental validation indicated that some genes were significantly upregulated in several cancer cell lines, implying their important roles in tumorigenesis. Our study identifies some genes associated with cell proliferation, which may be potential future targets in molecular targeted therapy.
Collapse
Affiliation(s)
- Li Guo
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yaodong Zhang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zibo Yin
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yaya Ji
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guowei Yang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Bowen Qian
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Sunjing Li
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jun Wang
- Department of Bioinformatics, Smart Health Big Data Analysis and Location Services Engineering Lab of Jiangsu Province, School of Geographic and Biologic Information, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Changxian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiangcheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
26
|
Feng F, Cheng Q, Zhang D, Li B, Qin H, Xu C, Han M, Yu Y, Li Z, Li JY, Qiu Z, Xiong L, Liu C, Li F, Yi B, Jiang X. Targeted deep sequencing contributes to guiding personalized targeted therapy for advanced biliary tract cancer patients with non‑radical resection: A real‑world study. Oncol Rep 2020; 43:1089-1102. [PMID: 32323774 PMCID: PMC7057932 DOI: 10.3892/or.2020.7491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/10/2020] [Indexed: 12/30/2022] Open
Abstract
Targeted therapy based on specific genetic alterations has been proven to be an effective treatment for various types of cancer. In the present study, we aimed to explore the efficacy of personalized targeted therapy guided by targeted deep sequencing for patients with advanced biliary tract cancer (BTC) after non-radical resection. Targeted deep sequencing was performed on 49 patients with BTC, to whom biologic agents were recommended. Among 32 patients with stage IV and R2 resection (a non-radical resection), 21 patients underwent conventional chemotherapy (mGEMOX), while the remaining 11 patients received a personalized targeted agent. The genomic landscape of the 49 patients with BTC was determined and the results showed that genetic alterations were enriched in the ERBB family and cell cycle pathway. After a median follow-up of 12 months, the 11 BTC patients with personalized targeted therapy showed a median progression-free survival (PFS) of 4.5 months (2.5–20.5 months), a median overall survival (OS) of 12.9 months (4.7–24.8 months) and a disease control rate (DCR) of 63.6%. In the other 21 BTC patients, who were undergoing conventional chemotherapy, the BTC patients had a median PFS of 1.5 months (0.5–11.6 months), a median OS of 4.1 months (1.3–18.4 months), and a DCR of 33.3%. In addition, 36.4% of the patients in the personalized targeted therapy group experienced grade >2 treatment-related toxicity vs. 19.0% of patients in the conventional chemotherapy group. This real-world study suggests that targeted deep sequencing contributes to the guidance of personalized targeted therapy based on individual actionable mutations, which may benefit advanced BTC patients undergoing non-radical resection.
Collapse
Affiliation(s)
- Feiling Feng
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Qingbao Cheng
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Dadong Zhang
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Bin Li
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Hao Qin
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Chang Xu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Miao Han
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Yong Yu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Zhizhen Li
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Jing-Yu Li
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Zhiquan Qiu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Lei Xiong
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Chen Liu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Fugen Li
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Bin Yi
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Xiaoqing Jiang
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
27
|
Functions of FGFR2 corrupted by translocations in intrahepatic cholangiocarcinoma. Cytokine Growth Factor Rev 2019; 52:56-67. [PMID: 31899106 DOI: 10.1016/j.cytogfr.2019.12.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 12/18/2019] [Indexed: 12/23/2022]
Abstract
Cholangiocarcinoma, originating from the biliary duct, represents a subset of liver cancer. With about 8000 new cases of cholangiocarcinoma diagnosed annually in the U.S., these fall into three categories: intrahepatic, peri-hilar, and extrahepatic cholangiocarcinoma. Arising from the epithelium of the bile duct, intrahepatic cholangiocarcinoma (ICC) is a universally fatal malignancy with very few treatment options. The poor prognosis and lack of molecular targeted therapies highlights ICC as a critical unmet medical need. With advances in sequencing technology, numerous chromosomal translocations have been discovered as drivers in cancer initiation and progression. Particularly in ICC, chromosomal translocations involving Fibroblast Growth Factor Receptor 2 (FGFR2) have been frequently identified, resulting in the creation of oncogenic fusion proteins. At the N-terminus, these fusion proteins share a nearly-identical FGFR2 moiety retaining an intact kinase domain and, at the C-terminus, a dimerization/oligomerization domain provided by different partner genes, including: Periphilin 1 (PPHLN1), Bicaudal family RNA binding protein 1 (BICC1), Adenosylhomocysteinase Like 1 (AHCYL1), and Transforming Acidic Coiled-Coil Containing Protein 3 (TACC3). A number of pre-clinical and clinical trials have shown the effectiveness of FGFR inhibitors in treating FGFR2 fusion-positive ICC patients. However, the efficacy of these inhibitors may be short-lived due to acquired resistance. In this review, we provide an overview of FGFR2 fusions, comparing their structures and mechanism of dimerization, examining the importance of FGFR2 as a partner gene, as well as highlighting the significance of alternative splicing of FGFR2 in these fusion proteins. In addition, we discuss various therapeutic options and their associated potencies in targeting these translocation-induced ICCs.
Collapse
|
28
|
Balitzer D, Joseph NM, Ferrell L, Shafizadeh N, Jain D, Zhang X, Yeh M, di Tommaso L, Kakar S. Immunohistochemical and molecular features of cholangiolocellular carcinoma are similar to well-differentiated intrahepatic cholangiocarcinoma. Mod Pathol 2019; 32:1486-1494. [PMID: 31186529 DOI: 10.1038/s41379-019-0290-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 02/08/2023]
Abstract
Cholangiolocellular carcinoma is characterized by low grade cytologic atypia, and anastomosing cords and glands resembling cholangioles or canals of Hering. Cholangiolocellular carcinoma has been variously regarded as a subtype of intrahepatic cholangiocarcinoma (World Health Organization 2000), combined hepatocellular-cholangiocarcinoma of stem cell subtype (World Health Organization 2010) and a distinct type of primary liver carcinoma. Capture-based next generation sequencing targeting the coding regions of 479 cancer genes and select introns was performed on 17 cases (5 cholangiolocellular carcinomas, 7 intrahepatic cholangiocarcinomas, 5 mixed cholangiolocellular-intrahepatic cholangiocarcinomas) along with immunohistochemistry for CK19, SALL4, CD56, CD117, and EMA. For 5 mixed cholangiolocellular-intrahepatic cholangiocarcinoma, the individual areas were micro-dissected prior to sequencing. CK19 and EMA were positive in all cases; both luminal and cytoplasmic EMA was seen in 3/5 cholangiolocellular carcinoma and 3/6 intrahepatic cholangiocarcinomas. CD117 and SALL4 were negative in all cases. CD56 was positive in 2/5 cholangiolocellular carcinoma, 4/6 intrahepatic cholangiocarcinoma and 2/5 mixed cases. Mutations typical of intrahepatic cholangiocarcinoma (IDH1/2, PBRM1, FGFR2) were present in 90% of cases with cholangiolocellular carcinoma component. The genomic profile (IDH1/2 mutations, FGFR2 fusions, chromatin-remodeling gene mutations such as ARID1A, PBRM1) and copy number alterations were similar in cholangiolocellular carcinoma, intrahepatic cholangiocarcinoma and mixed cholangiolocellular-intrahepatic cholangiocarcinoma. In all mixed cases, the immunohistochemistry results, mutational profile and copy number alterations in both components were similar. Cholangiolocellular carcinoma should be categorized as a histologic subtype of well-differentiated intrahepatic cholangiocarcinoma, and should not be considered a distinct entity, or combined hepatocellular-cholangiocarcinoma unless a distinct hepatocellular component is also present.
Collapse
Affiliation(s)
- Dana Balitzer
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Nancy M Joseph
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Linda Ferrell
- Department of Pathology, University of California, San Francisco, CA, USA
| | - Nafis Shafizadeh
- Department of Pathology, Kaiser Permanente Woodland Hills Medical Center, Woodland Hills, CA, USA
| | - Dhanpat Jain
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale New Haven Hospital, New Haven, CT, USA
| | - Matthew Yeh
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Luca di Tommaso
- Department of Pathology, Humanitas Clinical and Research Center, Rozzano-IRCCS, and Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| | - Sanjay Kakar
- Department of Pathology, University of California, San Francisco, CA, USA.
| |
Collapse
|
29
|
Czauderna C, Castven D, Mahn FL, Marquardt JU. Context-Dependent Role of NF-κB Signaling in Primary Liver Cancer-from Tumor Development to Therapeutic Implications. Cancers (Basel) 2019; 11:cancers11081053. [PMID: 31349670 PMCID: PMC6721782 DOI: 10.3390/cancers11081053] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/19/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory cell death is a major risk factor for the development of diverse cancers including liver cancer. Herein, disruption of the hepatic microenvironment as well as the immune cell composition are major determinants of malignant transformation and progression in hepatocellular carcinomas (HCC). Considerable research efforts have focused on the identification of predisposing factors that promote induction of an oncogenic field effect within the inflammatory liver microenvironment. Among the most prominent factors involved in this so-called inflammation-fibrosis-cancer axis is the NF-κB pathway. The dominant role of this pathway for malignant transformation and progression in HCC is well documented. Pathway activation is significantly linked to poor prognostic traits as well as stemness characteristics, which places modulation of NF-κB signaling in the focus of therapeutic interventions. However, it is well recognized that the mechanistic importance of the pathway for HCC is highly context and cell type dependent. While constitutive pathway activation in an inflammatory etiological background can significantly promote HCC development and progression, absence of NF-κB signaling in differentiated liver cells also significantly enhances liver cancer development. Thus, therapeutic targeting of NF-κB as well as associated family members may not only exert beneficial effects but also negatively impact viability of healthy hepatocytes and/or cholangiocytes, respectively. The review presented here aims to decipher the complexity and paradoxical functions of NF-κB signaling in primary liver and non-parenchymal cells, as well as the induced molecular alterations that drive HCC development and progression with a particular focus on (immune-) therapeutic interventions.
Collapse
Affiliation(s)
- Carolin Czauderna
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Darko Castven
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Friederike L Mahn
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany
| | - Jens U Marquardt
- Department of Medicine I, Lichtenberg Research Group for Molecular Hepatocarcinogenesis, University Medical Center of the Johannes Gutenberg University of Mainz, 55131 Mainz, Germany.
| |
Collapse
|
30
|
Castven D, Becker D, Czauderna C, Wilhelm D, Andersen JB, Strand S, Hartmann M, Heilmann‐Heimbach S, Roth W, Hartmann N, Straub BK, Mahn FL, Franck S, Pereira S, Haupts A, Vogel A, Wörns MA, Weinmann A, Heinrich S, Lang H, Thorgeirsson SS, Galle PR, Marquardt JU. Application of patient‐derived liver cancer cells for phenotypic characterization and therapeutic target identification. Int J Cancer 2018; 144:2782-2794. [DOI: 10.1002/ijc.32026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/18/2018] [Accepted: 11/07/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Darko Castven
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Diana Becker
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Carolin Czauderna
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Diana Wilhelm
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical ScienceUniversity of Copenhagen Copenhagen Denmark
| | - Susanne Strand
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Monika Hartmann
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Stefanie Heilmann‐Heimbach
- Institute of Human Genetics, University of Bonn School of Medicine & University Hospital of Bonn Department of Genomics, Life & Brain CenterUniversity of Bonn Bonn Germany
- Department of Genomics, Life & Brain CenterUniversity of Bonn Bonn Germany
| | - Wilfried Roth
- Institute of PathologyJohannes Gutenberg University Mainz Germany
| | - Nils Hartmann
- Institute of PathologyJohannes Gutenberg University Mainz Germany
| | - Beate K. Straub
- Institute of PathologyJohannes Gutenberg University Mainz Germany
| | - Friederike L. Mahn
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Sophia Franck
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Sharon Pereira
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Anna Haupts
- Institute of PathologyJohannes Gutenberg University Mainz Germany
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology and EndocrinologyHannover Medical School Hannover Germany
| | - Marcus A. Wörns
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Arndt Weinmann
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Stefan Heinrich
- Department of SurgeryJohannes Gutenberg University Mainz Germany
| | - Hauke Lang
- Department of SurgeryJohannes Gutenberg University Mainz Germany
| | - Snorri S. Thorgeirsson
- Laboratory of Human Carcinogenesis (LHC), Center for Cancer ResearchNational Cancer Institute, NIH Bethesda MD USA
| | - Peter R. Galle
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| | - Jens U. Marquardt
- Department of Medicine I, Lichtenberg Research GroupJohannes Gutenberg University Mainz Germany
| |
Collapse
|
31
|
Thongchot S, Vidoni C, Ferraresi A, Loilome W, Yongvanit P, Namwat N, Isidoro C. Dihydroartemisinin induces apoptosis and autophagy-dependent cell death in cholangiocarcinoma through a DAPK1-BECLIN1 pathway. Mol Carcinog 2018; 57:1735-1750. [PMID: 30136419 DOI: 10.1002/mc.22893] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/15/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Cholangiocarcinoma (CCA) is a very aggressive cancer arising from the malignant transformation of cholangiocytes. Intrahepatic CCA is associated with reactive inflammation and intense fibrosis of the hepatobiliary tract. Dihydroartemisinin (DHA), the active compound found in Artemisia annua, has been shown to possess anti-tumor activity in a variety of human cancers, including hepatoma. Here, we tested the ability of DHA to specifically kill CCA cells and have investigated the underlying mechanisms. DHA induced both apoptosis and autophagy-dependent caspase-independent cell death in many CCA cell lines, while being slightly toxic to immortalized cholangiocytes. DHA induced the expression of many apoptosis- and autophagy-related genes in CCA cells. In particular, it greatly induced the expression of DAPK1, and reduced the interaction of BECLIN1 with BCL-2 while promoting its interaction with PI3KC3. Genetic silencing of DAPK1 prevented DHA-induced autophagy. Pharmacologic and genetic inhibition of BECLIN1 function prevented autophagy and cell death induced by DHA in CCA cells. These data unravel a novel pathway of DHA cancer toxicity and open the possibility to introduce DHA in the therapeutic regimen for the treatment of CCA.
Collapse
Affiliation(s)
- Suyanee Thongchot
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.,Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand
| | - Chiara Vidoni
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Watcharin Loilome
- Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Puangrat Yongvanit
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Faculty of Medicine, Department of Biochemistry, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| |
Collapse
|
32
|
Bonomini F, Borsani E, Favero G, Rodella LF, Rezzani R. Dietary Melatonin Supplementation Could Be a Promising Preventing/Therapeutic Approach for a Variety of Liver Diseases. Nutrients 2018; 10:nu10091135. [PMID: 30134592 PMCID: PMC6164189 DOI: 10.3390/nu10091135] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023] Open
Abstract
In the therapeutic strategies, the role of diet is a well-established factor that can also have an important role in liver diseases. Melatonin, identified in animals, has many antioxidant properties and it was after discovered also in plants, named phytomelatonin. These substances have a positive effect during aging and in pathological conditions too. In particular, it is important to underline that the amount of melatonin produced by pineal gland in human decreases during lifetime and its reduction in blood could be related to pathological conditions in which mitochondria and oxidative stress play a pivotal role. Moreover, it has been indicated that melatonin/phytomelatonin containing foods may provide dietary melatonin, so their ingestion through balanced diets could be sufficient to confer health benefits. In this review, the classification of liver diseases and an overview of the most important aspects of melatonin/phytomelatonin, concerning the differences among their synthesis, their presence in foods and their role in health and diseases, are summarized. The findings suggest that melatonin/phytomelatonin supplementation with diet should be considered important in preventing different disease settings, in particular in liver. Currently, more studies are needed to strengthen the potential beneficial effects of melatonin/phytomelatonin in liver diseases and to better clarify the molecular mechanisms of action.
Collapse
Affiliation(s)
- Francesca Bonomini
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Elisa Borsani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| | - Luigi F Rodella
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
- Interdipartimental University Center of Research "Adaption and Regeneration of Tissues and Organs-(ARTO)", University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
33
|
Sun C, Zhu J, Wu B, Chen J, Zhu Z, Cai P, Guo W, Gu Z, Wang J, Huang S. Diagnostic and prognostic value of microRNAs in cholangiocarcinoma: a systematic review and meta-analysis. Cancer Manag Res 2018; 10:2125-2139. [PMID: 30050323 PMCID: PMC6055881 DOI: 10.2147/cmar.s158155] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background and aim Several dysregulated microRNAs (miRNAs) have been implicated in the pathogenesis of cholangiocarcinoma (CCA); however, small sample sizes and invariable research designs are limitations, hindering a thorough analysis of miRNAs as diagnostic and prognostic tools for CCA. This study aimed to systematically summarize the clinical value of miRNAs in human CCA both for all available miRNAs and single miRNA with multiple researches. Methods Pooled parameters included the area under the curve (AUC), sensitivity, specificity, and hazard ratios (HRs) to separately determine overall diagnostic and prognostic performance. Subgroup and sensitivity analyses were performed only in the event of heterogeneity. Thirty-four studies including 12 diagnostic studies and 22 prognostic studies were eligible for inclusion in this meta-analysis. Results We observed that miR-21, miR-26, miR-483, miR-106a, miR-150, miR-192, and miR-194 were employed for distinguishing patients with CCA from healthy controls. Pooled sensitivity, specificity, and AUC were 0.82 (95% confidence interval [CI] 0.77–0.86), 0.83 (95% CI 0.75–0.89), and 0.88 (95% CI 0.85–0.91), respectively. Abnormal expression of miR-21, miR-26a, miR-192, miR-200c, miR-221, miR-29a, miR-191, miR-181c, miR-34a, miR-106a, miR-203, and miR-373 in patients was confirmed to associate with poor survival rate. Pooled HRs and 95% CIs were calculated using STATA, resulting in the pooled HR of 1.47 (95% CI 0.91–2.37) for overall survival (OS), 0.67 (95% CI 0.16–2.81) for disease-free survival (DFS), 2.31 (95% CI 1.59–3.36) for progression-free survival (PFS), and 2.68 (95% CI 0.88–8.15) for relapse-free survival (RFS). Thus, CCA patients with dysregulated miRNA expression were confirmed to have shorter OS, DFS, PFS, and RFS. Data regarding the diagnostic and prognostic roles of miR-21 suggested pooled diagnostic results of miR-21 for sensitivity, specificity, and AUC were 0.85 (95% CI 0.76–0.91), 0.92 (95% CI 0.81–0.97), and 0.93 (95% CI 0.91–0.95), respectively, suggesting better diagnostic performance of miR-21 compared with other miRNAs. Meanwhile, pooled prognostic result of miR-21 for HR was 1.88 (95% CI 1.41–2.51), indicating miR-21 could more appropriately predict shorter OS in patients with CCA. Conclusion miRNAs may provide a new approach for clinical application, and miR-21 may be a promising biomarker for diagnosis and prognosis of CCA.
Collapse
Affiliation(s)
- Chao Sun
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Jie Zhu
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Bin Wu
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Jianlei Chen
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Zhenwei Zhu
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Peng Cai
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Wanliang Guo
- Radiology Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China
| | - Zhicheng Gu
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Jian Wang
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| | - Shungen Huang
- General Surgery Department, Children's Hospital of Soochow University, Suzhou, 215003, People's Republic of China,
| |
Collapse
|
34
|
Roeksomtawin S, Navasumrit P, Waraprasit S, Parnlob V, Sricharunrat T, Bhudhisawasdi V, Savaraj N, Ruchirawat M. Decreased argininosuccinate synthetase expression in Thai patients with cholangiocarcinoma and the effects of ADI-PEG20 treatment in CCA cell lines. Oncol Lett 2018; 16:1529-1538. [PMID: 30008833 PMCID: PMC6036342 DOI: 10.3892/ol.2018.8807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a severe cancer with poor prognosis. The aim of the present study was to explore the expression of argininosuccinate synthetase (ASS), as well as the possibility of using pegylated arginine deiminase (ADI-PEG20) for the treatment of CCA. ASS expression was determined in CCA specimens from 40 patients in Thailand. Immunohistochemical detection of ASS and determination of the proliferative index, Ki-67, were carried out in paraffin-embedded sections of these specimens, as well as in two CCA cell lines, HuCCA and RmCCA-1, derived from CCA samples from patients in Thailand. In total, ~45% of the CCA specimens had low ASS expression, and the level of expression was significantly negatively associated with cell differentiation (P<0.05) and Ki-67 expression (P<0.05). The level of ASS expression in tumor cells was significantly lower than that in non-tumor cells (1.3-fold, P<0.05). The HuCCA cell line had significantly lower levels (P<0.05) of ASS expression at the mRNA and protein levels relative to those of normal human immortalized fibroblast cells (BJ-1). By contrast, the RmCCA-1 cell line showed no significant difference. In addition, the effects of ADI-PEG20 on growth inhibition, apoptosis and cell cycle arrest were determined in HuCCA and RmCCA-1 cells. ADI-PEG20 treatment reduced cell viability and cell proliferation in the two CCA cell lines, though it had no effect in immortalized BJ-1 cells. Furthermore, ADI-PEG20 treatment significantly increased G0/G1 cell cycle arrest in HuCCA, though not in RmCCA-1 cells. ASS silencing in the RmCCA-1 cell line significantly enhanced its sensitivity to ADI-PEG20 treatment. Results from the in vitro study demonstrated that ADI-PEG20 has antitumor activity against CCA with low ASS expression.
Collapse
Affiliation(s)
- Somphon Roeksomtawin
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand
| | - Panida Navasumrit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Bangkok 10300, Thailand
| | - Somchamai Waraprasit
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Varabhorn Parnlob
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | | | - Vajarabhongsa Bhudhisawasdi
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kean 40000, Thailand.,Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Niramol Savaraj
- Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL 33125, USA
| | - Mathuros Ruchirawat
- Laboratory of Environmental Toxicology, Chulabhorn Research Institute, Bangkok 10210, Thailand.,Chulabhorn Graduate Institute, Bangkok 10210, Thailand.,Center of Excellence on Environmental Health and Toxicology, CHE, Ministry of Education, Bangkok 10300, Thailand
| |
Collapse
|
35
|
Two classes of intrahepatic cholangiocarcinoma defined by relative abundance of mutations and copy number alterations. Oncotarget 2018; 7:23825-36. [PMID: 27009864 PMCID: PMC5029666 DOI: 10.18632/oncotarget.8183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 03/02/2016] [Indexed: 12/15/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a biliary tree-origin epithelial malignancy in liver with unfavorable clinical outcomes. Systematic genome analyses may advance our understanding of ICC pathogenesis also improving current diagnostic and therapeutic modalities. In this study, we analyzed 17 ICC tumor-vs-matched normal pairs using either whole-exome (n = 7), transcriptome sequencing (n = 7) or both platforms (n = 3). For somatic mutations, we identified recurrent mutations of previously reported genes such as KRAS, TP53, APC as well as epigenetic regulators and those of TGFβ signaling pathway. According to the abundance of somatic mutations and DNA copy number alterations (CNA), ten ICC exome cases were distinguished into two classes as those primarily driven by either somatic mutations (M class) or CNAs (C class). Compared to M class ICCs (92-147 somatic mutations; n = 5) with a relative deficit of CNAs, C class ICCs (54-84 mutations; n = 5) harbor recurrent focal CNAs including deletions involving CDKN2A, ROBO1, ROBO2, RUNX3, and SMAD4. We also show that transcriptome sequencing can be used for expression-based ICC categorization but the somatic mutation calling from the transcriptome can be heavily influenced by the gene expression level and potentially, by posttranscriptional modification such as nonsense mediated decay. Along with a substantial level of mutational heterogeneity of ICC genomes, our study reveals previously unrecognized two ICC classes defined by relative abundance of somatic mutations over CNAs or vice versa, which should be considered in the selection of genotyping platforms and sensitive screening of targets for ICC therapeutics.
Collapse
|
36
|
Nicotinamide suppresses cell growth by G1-phase arrest and induces apoptosis in intrahepatic cholangiocarcinoma. Mol Cell Toxicol 2018. [DOI: 10.1007/s13273-018-0006-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
37
|
Ferrell LD, Kakar S, Terracciano LM, Wee A. Tumours and Tumour-like Lesions of the Liver. MACSWEEN'S PATHOLOGY OF THE LIVER 2018:780-879. [DOI: 10.1016/b978-0-7020-6697-9.00013-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Abstract
Cholangiocarcinoma is a malignant neoplasm originating from biliary epithelial cells. The incidence and mortality of this cancer are rising in the world. Currently, cholangiocarcinoma is accepted as a stem cell disease with many risk factors. Diagnosis is relatively simple but therapy is extremely difficult. Surgery is the mainstay of treatment for early stage patients. Endobiliary approaches, chemotherapy and radiotherapy are other therapeutic approaches.
Collapse
Affiliation(s)
- Vedat Goral
- Department of Gastroenterology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
39
|
Oliveira DVNP, Zhang S, Chen X, Calvisi DF, Andersen JB. Molecular profiling of intrahepatic cholangiocarcinoma: the search for new therapeutic targets. Expert Rev Gastroenterol Hepatol 2017; 11:349-356. [PMID: 28162004 DOI: 10.1080/17474124.2017.1292127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is the second most frequent primary tumor of the liver and a highly lethal disease. Therapeutic options for advanced iCCA are limited and ineffective due to the largely incomplete understanding of the molecular pathogenesis of this deadly tumor. Areas covered: The present review article outlines the main studies and resulting discoveries on the molecular profiling of iCCA, with a special emphasis on the different techniques used for this purpose, the diagnostic and prognostic markers identified, as well as the genes and pathways that could be potentially targeted with innovative therapies. Expert commentary: Molecular profiling has led to the identification of distinct iCCA subtypes, characterized by peculiar genetic alterations and transcriptomic features. Targeted therapies against some of the identified genes are ongoing and hold great promise to improve the prognosis of iCCA patients.
Collapse
Affiliation(s)
- Douglas V N P Oliveira
- a Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences , University of Copenhagen , Copenhagen N , Denmark
| | - Shanshan Zhang
- b Department of Bioengineering and Therapeutic Sciences and Liver Center , University of California , San Francisco , CA , USA
| | - Xin Chen
- b Department of Bioengineering and Therapeutic Sciences and Liver Center , University of California , San Francisco , CA , USA
| | - Diego F Calvisi
- c Institute of Pathology, University Medicine of Greifswald , Greifswald , Germany
| | - Jesper B Andersen
- a Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences , University of Copenhagen , Copenhagen N , Denmark
| |
Collapse
|
40
|
Jain A, Javle M. Molecular profiling of biliary tract cancer: a target rich disease. J Gastrointest Oncol 2016; 7:797-803. [PMID: 27747093 DOI: 10.21037/jgo.2016.09.01] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Biliary tract cancers (BTCs) are relatively uncommon orphan tumors that have an aggressive disease course and a poor clinical outcome. Surgery is the only curative treatment, but most patients present with advanced disease and therefore have a limited survival. Gemcitabine and cisplatin based chemotherapy has been the only widely accepted standard systemic therapy regimen in these patients but these tumors can be chemoresistant, further complicating their management. In recent times, there has been considerable research in the genetics of BTC and with the advent of new, advanced technologies like next-generation sequencing (NGS) we are achieving a greater understanding of its disease biology. With the help of NGS, we have now been able to identify actionable mutations such as in the isocitrate dehydrogenase 1 (IDH1), FGFR2, BRAF and HER2/neu genes for targeted therapeutics and correlate the genetic variations with distinct clinical prognoses. This recent genetic information has the potential to make precision medicine a part of routine clinical practice for the management of BTC patients.
Collapse
Affiliation(s)
- Apurva Jain
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
41
|
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: 300] [Impact Index Per Article: 33.3] [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
|
42
|
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: 3.1] [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
|
43
|
Wei M, Lü L, Lin P, Chen Z, Quan Z, Tang Z. Multiple cellular origins and molecular evolution of intrahepatic cholangiocarcinoma. Cancer Lett 2016; 379:253-61. [PMID: 26940139 DOI: 10.1016/j.canlet.2016.02.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/12/2022]
Abstract
Intrahepatic cholangiocarcinoma (ICC) is an aggressive malignancy associated with unfavorable prognosis and for which no effective treatments are available. Its molecular pathogenesis is poorly understood. Genome-wide sequencing and high-throughput technologies have provided critical insights into the molecular basis of ICC while sparking a heated debate on the cellular origin. Cancer exhibits variabilities in origin, progression and cell biology. Recent evidence suggests that ICC has multiple cellular origins, including differentiated hepatocytes; intrahepatic biliary epithelial cells (IBECs)/cholangiocytes; pluripotent stem cells, such as hepatic stem/progenitor cells (HPCs) and biliary tree stem/progenitor cells (BTSCs); and peribiliary gland (PBG). However, both somatic mutagenesis and epigenomic features are highly cell type-specific. Multiple cellular origins may have profoundly different genomic landscapes and key signaling pathways, driving phenotypic variation and thereby posing significant challenges to personalized medicine in terms of achieving the optimal drug response and patient outcome. Considering this information, we have summarized the latest experimental evidence and relevant literature to provide an up-to-date view of the cellular origin of ICC, which will contribute to establishment of a hierarchical model of carcinogenesis and allow for improvement of the anatomical-based classification of ICC. These new insights have important implications for both the diagnosis and treatment of ICC patients.
Collapse
Affiliation(s)
- Miaoyan Wei
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Lisheng Lü
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Peiyi Lin
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zhisheng Chen
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zhiwei Quan
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zhaohui Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
44
|
Gillet JP, Andersen JB, Madigan JP, Varma S, Bagni RK, Powell K, Burgan WE, Wu CP, Calcagno AM, Ambudkar SV, Thorgeirsson SS, Gottesman MM. A Gene Expression Signature Associated with Overall Survival in Patients with Hepatocellular Carcinoma Suggests a New Treatment Strategy. Mol Pharmacol 2016; 89:263-72. [PMID: 26668215 PMCID: PMC4727122 DOI: 10.1124/mol.115.101360] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/11/2015] [Indexed: 12/11/2022] Open
Abstract
Despite improvements in the management of liver cancer, the survival rate for patients with hepatocellular carcinoma (HCC) remains dismal. The survival benefit of systemic chemotherapy for the treatment of liver cancer is only marginal. Although the reasons for treatment failure are multifactorial, intrinsic resistance to chemotherapy plays a primary role. Here, we analyzed the expression of 377 multidrug resistance (MDR)-associated genes in two independent cohorts of patients with advanced HCC, with the aim of finding ways to improve survival in this poor-prognosis cancer. Taqman-based quantitative polymerase chain reaction revealed a 45-gene signature that predicts overall survival (OS) in patients with HCC. Using the Connectivity Map Tool, we were able to identify drugs that converted the gene expression profiles of HCC cell lines from ones matching patients with poor OS to profiles associated with good OS. We found three compounds that convert the gene expression profiles of three HCC cell lines to gene expression profiles associated with good OS. These compounds increase histone acetylation, which correlates with the synergistic sensitization of those MDR tumor cells to conventional chemotherapeutic agents, including cisplatin, sorafenib, and 5-fluorouracil. Our results indicate that it is possible to modulate gene expression profiles in HCC cell lines to those associated with better outcome. This approach also increases sensitization of HCC cells toward conventional chemotherapeutic agents. This work suggests new treatment strategies for a disease for which few therapeutic options exist.
Collapse
Affiliation(s)
- Jean-Pierre Gillet
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Jesper B Andersen
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - James P Madigan
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Sudhir Varma
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Rachel K Bagni
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Katie Powell
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - William E Burgan
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Chung-Pu Wu
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Anna Maria Calcagno
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Suresh V Ambudkar
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Snorri S Thorgeirsson
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| | - Michael M Gottesman
- Laboratory of Cell Biology (J-P.G., J.P.M., C-P.W., A.M.C., S.V.A., M.M.G.) and Laboratory of Experimental Carcinogenesis (J.B.A., S.S.T.), Center for Cancer Research, National Cancer Institute, and Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, Office of Science Management and Operations, National Institute of Allergy and Infectious Diseases (S.V.), National Institutes of Health, Bethesda, Maryland; and the Viral Technologies Group and Molecular Detection Group, Protein Expression Laboratory, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Marylanld (R.K.B., K.P., W.E.B.)
| |
Collapse
|
45
|
Recurrent Amplification at 13q34 Targets at CUL4A, IRS2, and TFDP1 As an Independent Adverse Prognosticator in Intrahepatic Cholangiocarcinoma. PLoS One 2015; 10:e0145388. [PMID: 26684807 PMCID: PMC4686179 DOI: 10.1371/journal.pone.0145388] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023] Open
Abstract
Amplification of genes at 13q34 has been reported to be associated with tumor proliferation and progression in diverse types of cancers. However, its role in intrahepatic cholangiocarcinoma (iCCA) has yet to be explored. We examined two iCCA cell lines and 86 cases of intrahepatic cholangiocarcinoma to analyze copy number of three target genes, including cullin 4A (CUL4A), insulin receptor substrate 2 (IRS2), and transcription factor Dp-1 (TFDP1) at 13q34 by quantitative real-time polymerase chain reaction. The cell lines and all tumor samples were used to test the relationship between copy number (CN) alterations and protein expression by western blotting and immunohistochemical assays, respectively. IRS2 was introduced, and each target gene was silenced in cell lines. The mobility potential of cells was compared in the basal condition and after manipulation using cell migration and invasion assays. CN alterations correlated with protein expression levels. The SNU1079 cell line containing deletions of the target genes demonstrated decreased protein expression levels and significantly lower numbers of migratory and invasive cells, as opposed to the RBE cell line, which does not contain CN alterations. Overexpression of IRS2 by introducing IRS2 in SUN1079 cells increased the mobility potential. In contrast, silencing each target gene showed a trend or statistical significance toward inhibition of migratory and invasive capacities in RBE cells. In tumor samples, the amplification of each of these genes was associated with poor disease-free survival. Twelve cases (13.9%) demonstrated copy numbers > 4 for all three genes tested (CUL4A, IRS2, and TFDP1), and showed a significant difference in disease-free survival by both univariate and multivariate survival analyses (hazard ratio, 2.69; 95% confidence interval, 1.23 to 5.88; P = 0.013). Our data demonstrate that amplification of genes at 13q34 plays an oncogenic role in iCCA featuring adverse disease-free survival, which may provide new directions for targeted therapy.
Collapse
|
46
|
Tang ZH, Lv LS, Lin PY, Chen ZS, Quan ZW. Viewing cellular origin of intrahepatic cholangiocarcinoma from perspective of tumor heterogeneity. Shijie Huaren Xiaohua Zazhi 2015; 23:5255-5262. [DOI: 10.11569/wcjd.v23.i33.5255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is the second most frequent primary liver malignancy, representing approximately 5%-15% of all cases. ICC is a very heterogeneous tumor from any point of view, including epidemiology, risk factors, morphology, pathology, molecular pathology, growth and clinical features. For a long time, scholar believe that ICC is derived from the intrahepatic bile duct epithelium. Genome-wide, high-throughput technologies have resulted in major advances in understanding the molecular basis of ICC. Recent data have revealed that ICC may be derived from different cell types, including hepatocytes, dysplastic or immature bile duct epithelial cells, pluripotent stem cells (biliary tree stem/progenitor cells) and peribiliary glands cells. Therefore, we hypothesize that different cellular origins may be responsible for the heterogeneity of ICC.
Collapse
|
47
|
Wang M, Wen TF, He LH, Li C, Zhu WJ, Trishul NM. A six-microRNA set as prognostic indicators for bile duct cancer. Int J Clin Exp Med 2015; 8:17261-17270. [PMID: 26770318 PMCID: PMC4694218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 07/11/2015] [Indexed: 06/05/2023]
Abstract
MicroRNAs (miRNAs) play important roles in cancer progression by altering transcriptional control. The purpose of this study is to identify and explore specific miRNAs as prognostic and predictive biomarkers for bile duct cancer (BDC) by analyzing Next-generation data. miRNA expression profiles and corresponding clinical information of BDC samples were extracted from The Cancer Genome Atlas (TCGA). The differentially expressed miRNAs were determined by SAMR package in R software. Target genes of those miRNAs were predicted by Targetscan. Functional enrichment analysis and hypergeometric test analysis of target genes were performed. Then, diagnosis accuracy of miRNAs was judged by ROC Curves analysis. Total 120 differentially expressed miRNAs were obtained, of which six important miRNAs were selected and predicted as prognosis and predicting biomarkers in BDC. Besides, functional analysis showed that both enriched pathways were significantly related with ion binding, which might involve in the carcinogenesis of BDC. Moreover, top 3 important pathways sharing the most influence were noted. Our results demonstrated that hsa-miR-483-5p, hsa-miR-675, hsa-miR-139-3p, hsa-miR-598, hsa-miR-625 and hsa-miR-187 could serve as prognostic and predictive markers for survival of BDC patients and could potentially be provided as targets for future therapy.
Collapse
Affiliation(s)
- Ming Wang
- Department of Hepatic Surgery, West China Hospital of Sichuan UniversityChengdu 610041, China
| | - Tian-Fu Wen
- Department of Hepatic Surgery, West China Hospital of Sichuan UniversityChengdu 610041, China
| | - Lin-Hai He
- Department of General Surgery, Xishuangbanna Prefecture People’s HospitalXishuangbanna 666100, China
| | - Chuan Li
- Department of Hepatic Surgery, West China Hospital of Sichuan UniversityChengdu 610041, China
| | - Wen-Jiang Zhu
- Department of Hepatic Surgery, West China Hospital of Sichuan UniversityChengdu 610041, China
| | | |
Collapse
|
48
|
Loaiza-Bonilla A, Furth EE, Morrissette JJD. Next-generation sequencing and personalized genomic medicine in hepatobiliary malignancies. Hepat Oncol 2015; 2:359-370. [PMID: 30191018 PMCID: PMC6095428 DOI: 10.2217/hep.15.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Liver cancer is a heterogeneous group of tumors characterized by significant molecular and genomic heterogeneity. The advent of powerful genomic technologies has allowed detection of recurrent somatic alterations in liver cancer, including mutations, copy number alterations as well as changes in transcriptomes and epigenomes, with the potential to translate these data into clinically relevant predictive and prognostic factors. In this review, we discuss recent advances in the application of high-throughput genomic technologies in liver cancer and the integration of such cancer genome profiling data, highlighting specific relevant subgroups and explain how this knowledge can be used in translational clinical research, 'basket trials', molecular tumor boards, targeted therapy and for personalized genomic medicine applications.
Collapse
Affiliation(s)
- Arturo Loaiza-Bonilla
- Abramson Cancer Center, Perelman Center for Advanced Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Emma E Furth
- Department of Pathology & Laboratory Medicine, University of Pennsylvania School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Jennifer JD Morrissette
- Department of Pathology & Laboratory Medicine, University of Pennsylvania School of Medicine, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Arnold A, Bahra M, Lenze D, Bradtmöller M, Guse K, Gehlhaar C, Bläker H, Heppner FL, Koch A. Genome wide DNA copy number analysis in cholangiocarcinoma using high resolution molecular inversion probe single nucleotide polymorphism assay. Exp Mol Pathol 2015; 99:344-53. [DOI: 10.1016/j.yexmp.2015.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 08/05/2015] [Indexed: 12/23/2022]
|
50
|
Seeree P, Pearngam P, Kumkate S, Janvilisri T. 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: 1.4] [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
Affiliation(s)
- Pattaya Seeree
- Department of Biology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Phorutai Pearngam
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Supeecha Kumkate
- Department of Biology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|