1651
|
Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, Chakravarty D, Daian F, Gao Q, Bailey MH, Liang WW, Foltz SM, Shmulevich I, Ding L, Heins Z, Ochoa A, Gross B, Gao J, Zhang H, Kundra R, Kandoth C, Bahceci I, Dervishi L, Dogrusoz U, Zhou W, Shen H, Laird PW, Way GP, Greene CS, Liang H, Xiao Y, Wang C, Iavarone A, Berger AH, Bivona TG, Lazar AJ, Hammer GD, Giordano T, Kwong LN, McArthur G, Huang C, Tward AD, Frederick MJ, McCormick F, Meyerson M, Van Allen EM, Cherniack AD, Ciriello G, Sander C, Schultz N. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 2018; 173:321-337.e10. [PMID: 29625050 PMCID: PMC6070353 DOI: 10.1016/j.cell.2018.03.035] [Citation(s) in RCA: 2069] [Impact Index Per Article: 295.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Genetic alterations in signaling pathways that control cell-cycle progression, apoptosis, and cell growth are common hallmarks of cancer, but the extent, mechanisms, and co-occurrence of alterations in these pathways differ between individual tumors and tumor types. Using mutations, copy-number changes, mRNA expression, gene fusions and DNA methylation in 9,125 tumors profiled by The Cancer Genome Atlas (TCGA), we analyzed the mechanisms and patterns of somatic alterations in ten canonical pathways: cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGFβ signaling, p53 and β-catenin/Wnt. We charted the detailed landscape of pathway alterations in 33 cancer types, stratified into 64 subtypes, and identified patterns of co-occurrence and mutual exclusivity. Eighty-nine percent of tumors had at least one driver alteration in these pathways, and 57% percent of tumors had at least one alteration potentially targetable by currently available drugs. Thirty percent of tumors had multiple targetable alterations, indicating opportunities for combination therapy.
Collapse
Affiliation(s)
- Francisco Sanchez-Vega
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marco Mina
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland and Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Joshua Armenia
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Walid K Chatila
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Augustin Luna
- cBio Center, Dana-Farber Cancer Institute, Boston, MA; Department of Cell Biology, Harvard Medical School, Boston, MA
| | - Konnor C La
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - David L Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, US
| | | | - Sadegh Saghafinia
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland and Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Debyani Chakravarty
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Foysal Daian
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Qingsong Gao
- Department of Medicine and McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Matthew H Bailey
- Department of Medicine and McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Wen-Wei Liang
- Department of Medicine and McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | - Steven M Foltz
- Department of Medicine and McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, 63110, USA
| | | | - Li Ding
- Department of Medicine and McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri, 63110, USA; Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zachary Heins
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Angelica Ochoa
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Benjamin Gross
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jianjiong Gao
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hongxin Zhang
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ritika Kundra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cyriac Kandoth
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Istemi Bahceci
- Computer Engineering Department, Bilkent University, Ankara 06800, Turkey
| | - Leonard Dervishi
- Computer Engineering Department, Bilkent University, Ankara 06800, Turkey
| | - Ugur Dogrusoz
- Computer Engineering Department, Bilkent University, Ankara 06800, Turkey
| | - Wanding Zhou
- Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids Michigan, 49503, USA
| | - Hui Shen
- Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids Michigan, 49503, USA
| | - Peter W Laird
- Van Andel Research Institute, 333 Bostwick Ave NE, Grand Rapids Michigan, 49503, USA
| | - Gregory P Way
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | | | - Chen Wang
- Department of Health Sciences Research and Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN, 55905, USA
| | - Antonio Iavarone
- Institute for Cancer Genetics, Department of Neurology and Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Alice H Berger
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Trever G Bivona
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1450 3rd Street, San Francisco, California 94143, USA
| | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine & Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd-Unit 85, Houston, Texas 77030, USA
| | - Gary D Hammer
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Endocrine Oncology Program, University of Michigan, Ann Arbor, Michigan, MI 48105, USA
| | - Thomas Giordano
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI; Department of Internal Medicine, Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI; Comprehensive Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grant McArthur
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Chenfei Huang
- Dept. of Otolaryngology, Baylor College of Medicine, USA
| | - Aaron D Tward
- University of California, San Francisco Department of Otolaryngology-Head and Neck Surgery. 2233 Post Street, San Francisco, CA, 94143, USA
| | | | - Frank McCormick
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 1450 3rd Street, San Francisco, CA 94143, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, US
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, US
| | - Andrew D Cherniack
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, US
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne (UNIL), 1011 Lausanne, Vaud, Switzerland and Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland.
| | - Chris Sander
- cBio Center, Dana-Farber Cancer Institute, Boston, MA; Department of Cell Biology, Harvard Medical School, Boston, MA.
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Departments of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
1652
|
Shen Q, Eun JW, Lee K, Kim HS, Yang HD, Kim SY, Lee EK, Kim T, Kang K, Kim S, Min DH, Oh SN, Lee YJ, Moon H, Ro SW, Park WS, Lee JY, Nam SW. Barrier to autointegration factor 1, procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3, and splicing factor 3b subunit 4 as early-stage cancer decision markers and drivers of hepatocellular carcinoma. Hepatology 2018; 67:1360-1377. [PMID: 29059470 DOI: 10.1002/hep.29606] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/20/2017] [Accepted: 10/16/2017] [Indexed: 01/05/2023]
Abstract
UNLABELLED An accurate tool enabling early diagnosis of hepatocellular carcinoma (HCC) is clinically important, given that early detection of HCC markedly improves survival. We aimed to investigate the molecular markers underlying early progression of HCC that can be detected in precancerous lesions. We designed a gene selection strategy to identify potential driver genes by integrative analysis of transcriptome and clinicopathological data of human multistage HCC tissues, including precancerous lesions, low- and high-grade dysplastic nodules. The gene selection process was guided by detecting the selected molecules in both HCC and precancerous lesion. Using various computational approaches, we selected 10 gene elements as a candidate and, through immunohistochemical staining, showed that barrier to autointegration factor 1 (BANF1), procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3 (PLOD3), and splicing factor 3b subunit 4 (SF3B4) are HCC decision markers with superior capability to diagnose early-stage HCC in a large cohort of HCC patients, as compared to the currently popular trio of HCC diagnostic markers: glypican 3, glutamine synthetase, and heat-shock protein 70. Targeted inactivation of BANF1, PLOD3, and SF3B4 inhibits in vitro and in vivo liver tumorigenesis by selectively modulating epithelial-mesenchymal transition and cell-cycle proteins. Treatment of nanoparticles containing small-interfering RNAs of the three genes suppressed liver tumor incidence as well as tumor growth rates in a spontaneous mouse HCC model. We also demonstrated that SF3B4 overexpression triggers SF3b complex to splice tumor suppressor KLF4 transcript to nonfunctional skipped exon transcripts. This contributes to malignant transformation and growth of hepatocyte through transcriptional inactivation of p27Kip1 and simultaneously activation of Slug genes. CONCLUSION The findings suggest molecular markers of BANF1, PLOD3, and SF3B4 indicating early-stage HCC in precancerous lesion, and also suggest drivers for understanding the development of hepatocarcinogenesis. (Hepatology 2018;67:1360-1377).
Collapse
Affiliation(s)
- Qingyu Shen
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Woo Eun
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Kyungbun Lee
- Department of Pathology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyung Seok Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee Doo Yang
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Yean Kim
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Taemook Kim
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan, Republic of Korea
| | - Keunsoo Kang
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan, Republic of Korea
| | - Seongchan Kim
- Center for RNA Research, Institute for Basic Science (IBS), Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Dal-Hee Min
- Center for RNA Research, Institute for Basic Science (IBS), Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Soon-Nam Oh
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Young-Joon Lee
- Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyuk Moon
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Simon Weonsang Ro
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Young Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Functional RNomics Research Center, The Catholic University of Korea, Seoul, Republic of Korea
- Cancer Evolution Research Center, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
1653
|
Ng CKY, Di Costanzo GG, Terracciano LM, Piscuoglio S. Circulating Cell-Free DNA in Hepatocellular Carcinoma: Current Insights and Outlook. Front Med (Lausanne) 2018; 5:78. [PMID: 29632864 PMCID: PMC5880118 DOI: 10.3389/fmed.2018.00078] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 03/08/2018] [Indexed: 12/25/2022] Open
Abstract
Over the past decade, the advancements in massively parallel sequencing have provided a new paradigm in biomedical research to uncover the genetic basis of human diseases. Integration of ‘omics information has begun transforming clinical management of cancer patients in terms of diagnostics and treatment options, giving rise to the era of precision medicine. Currently, nucleic acids for molecular profiling for patients diagnosed with hepatocellular carcinoma (HCC) are typically obtained from resected tumor materials or transplanted neoplastic liver and occasionally from biopsies. Given the intrinsic risks associated with such invasive procedures, circulating cell-free DNA (cfDNA) has been proposed as an alternative source for tumor DNA. Circulating cfDNA is a type of cell-free nucleic acid that derives from apoptotic, necrotic, as well as living eukaryotic cells. Importantly, the detection of abnormal forms of circulating cfDNA that originate from cancer cells provides a new tool for cancer detection, disease monitoring, and molecular profiling. Currently, cfDNA is beginning to be adopted into clinical practice as a non-invasive tool to monitor disease by tracking the evolution of disease-specific genetic alterations in several major cancer types. Moreover, cfDNA is demonstrating potential clinical value as a surrogate to assess the molecular makeup of tumors and to overcome the sampling biases inherent to intra-tumor genetic heterogeneity, especially in the metastatic setting. With the improvements in ‘omics and molecular biology techniques, coupled with the increasing understanding in the molecular pathogenesis of cancer, it can be anticipated that the detection and analysis of cfDNA will become more specific and sensitive and thus enable cfDNA analysis to be used as a diagnostic aid in patients with early-stage disease and perhaps even in a screening setting. In this review, we provide an overview of the latest findings on the role and potential utility of cfDNA analysis in the diagnosis, management, and screening of HCC.
Collapse
Affiliation(s)
- Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Hepatology Laboratory, University of Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
1654
|
Inhibition of Enhancer of zeste homolog 2 (EZH2) induces natural killer cell-mediated eradication of hepatocellular carcinoma cells. Proc Natl Acad Sci U S A 2018; 115:E3509-E3518. [PMID: 29581297 DOI: 10.1073/pnas.1802691115] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cell-mediated tumor cell eradication could inhibit tumor initiation and progression. However, the factors that regulate NK cell-mediated cancer cell eradication remain unclear. We determined that hepatocellular carcinoma (HCC) cells exhibit transcriptional down-regulation of NK group 2D (NKG2D) ligands and are largely resistant to NK cell-mediated eradication. Because the down-regulation of NKG2D ligands occurred at the transcriptional level, we tested 32 chemical inhibitors of epigenetic regulators for their ability to re-express NKG2D ligands and enhance HCC cell eradication by NK cells and found that Enhancer of zeste homolog 2 (EZH2) was a transcriptional repressor of NKG2D ligands. The inhibition of EZH2 by small-molecule inhibitors or genetic means enhanced HCC cell eradication by NK cells in a NKG2D ligand-dependent manner. Collectively, these results demonstrate that EZH2 inhibition enhances HCC eradication by NK cells and that EZH2 functions, in part, as an oncogene by inhibiting immune response.
Collapse
|
1655
|
Krstic J, Galhuber M, Schulz TJ, Schupp M, Prokesch A. p53 as a Dichotomous Regulator of Liver Disease: The Dose Makes the Medicine. Int J Mol Sci 2018; 19:E921. [PMID: 29558460 PMCID: PMC5877782 DOI: 10.3390/ijms19030921] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 02/07/2023] Open
Abstract
Lifestyle-related disorders, such as the metabolic syndrome, have become a primary risk factor for the development of liver pathologies that can progress from hepatic steatosis, hepatic insulin resistance, steatohepatitis, fibrosis and cirrhosis, to the most severe condition of hepatocellular carcinoma (HCC). While the prevalence of liver pathologies is steadily increasing in modern societies, there are currently no approved drugs other than chemotherapeutic intervention in late stage HCC. Hence, there is a pressing need to identify and investigate causative molecular pathways that can yield new therapeutic avenues. The transcription factor p53 is well established as a tumor suppressor and has recently been described as a central metabolic player both in physiological and pathological settings. Given that liver is a dynamic tissue with direct exposition to ingested nutrients, hepatic p53, by integrating cellular stress response, metabolism and cell cycle regulation, has emerged as an important regulator of liver homeostasis and dysfunction. The underlying evidence is reviewed herein, with a focus on clinical data and animal studies that highlight a direct influence of p53 activity on different stages of liver diseases. Based on current literature showing that activation of p53 signaling can either attenuate or fuel liver disease, we herein discuss the hypothesis that, while hyper-activation or loss of function can cause disease, moderate induction of hepatic p53 within physiological margins could be beneficial in the prevention and treatment of liver pathologies. Hence, stimuli that lead to a moderate and temporary p53 activation could present new therapeutic approaches through several entry points in the cascade from hepatic steatosis to HCC.
Collapse
Affiliation(s)
- Jelena Krstic
- Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, 8010 Graz, Austria.
| | - Markus Galhuber
- Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, 8010 Graz, Austria.
| | - Tim J Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition, Potsdam-Rehhbrücke, 14558 Nuthetal, Germany.
- German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany.
- Institute of Nutritional Science, University of Potsdam, 14558 Nuthetal, Germany.
| | - Michael Schupp
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, 10117 Berlin, Germany.
| | - Andreas Prokesch
- Gottfried Schatz Research Center for Cell Signaling, Metabolism & Aging, Medical University of Graz, 8010 Graz, Austria.
- BioTechMed-Graz, 8010 Graz, Austria.
| |
Collapse
|
1656
|
Harris WP, Wong KM, Saha S, Dika IE, Abou-Alfa GK. Biomarker-Driven and Molecular Targeted Therapies for Hepatobiliary Cancers. Semin Oncol 2018; 45:116-123. [PMID: 30348531 DOI: 10.1053/j.seminoncol.2018.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/07/2018] [Indexed: 12/18/2022]
Abstract
The recent accumulation of molecular profiling data for primary hepatobiliary malignancies, including hepatocellular carcinoma and biliary tract cancers, has led to a proliferation of promising therapeutic investigations in recent years. Treatment with pathway-specific targeted inhibitors and immunotherapeutic agents have demonstrated promising early clinical results. Key molecular alterations in common hepatobiliary cancers and ongoing interventional clinical trials of molecularly targeted systemic agents focusing on hepatocellular carcinoma and biliary tract cancer are reviewed.
Collapse
Affiliation(s)
- William P Harris
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Kit Man Wong
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle WA; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Supriya Saha
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle WA; Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle WA
| | - Imane El Dika
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Medical College at Cornell University, New York, NY.
| |
Collapse
|
1657
|
Koenig AB, Barajas JM, Guerrero MJ, Ghoshal K. A Comprehensive Analysis of Argonaute-CLIP Data Identifies Novel, Conserved and Species-Specific Targets of miR-21 in Human Liver and Hepatocellular Carcinoma. Int J Mol Sci 2018; 19:E851. [PMID: 29538313 PMCID: PMC5877712 DOI: 10.3390/ijms19030851] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/08/2018] [Accepted: 03/13/2018] [Indexed: 12/16/2022] Open
Abstract
MicroRNAs are ~22 nucleotide RNAs that regulate gene expression at the post-transcriptional level by binding messenger RNA transcripts. miR-21 is described as an oncomiR whose steady-state levels are commonly increased in many malignancies, including hepatocellular carcinoma (HCC). Methods known as cross-linking and immunoprecipitation of RNA followed by sequencing (CLIP-seq) have enabled transcriptome-wide identification of miRNA interactomes. In our study, we use a publicly available Argonaute-CLIP dataset (GSE97061), which contains nine HCC cases with matched benign livers, to characterize the miR-21 interactome in HCC. Argonaute-CLIP identified 580 miR-21 bound target sites on coding transcripts, of which 332 were located in the coding sequences, 214 in the 3'-untranslated region, and 34 in the 5'-untranslated region, introns, or downstream sequences. We compared the expression of miR-21 targets in 377 patients with liver cancer from the data generated by The Cancer Genome Atlas (TCGA) and found that mRNA levels of 402 miR-21 targets are altered in HCC. Expression of three novel predicted miR-21 targets (CAMSAP1, DDX1 and MARCKSL1) correlated with HCC patient survival. Analysis of RNA-seq data from SK-Hep1 cells treated with a miR-21 antisense oligonucleotide (GSE65892) identified RMND5A, an E3 ubiquitin ligase, as a strong miR-21 candidate target. Collectively, our analysis identified novel miR-21 targets that are likely to play a causal role in hepatocarcinogenesis.
Collapse
Affiliation(s)
- Aaron Balasingam Koenig
- Department of Pathology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | - Juan Martín Barajas
- Department of Pathology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - María Jose Guerrero
- Department of Pathology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Kalpana Ghoshal
- Department of Pathology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
1658
|
Gingold JA, Zhu D, Lee DF, Kaseb A, Chen J. Genomic Profiling and Metabolic Homeostasis in Primary Liver Cancers. Trends Mol Med 2018. [PMID: 29530485 DOI: 10.1016/j.molmed.2018.02.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), the two most common primary liver cancers, represent the second most common cancer-related cause of death worldwide, with most cases being diagnosed at an advanced stage. Recent genome-wide studies have helped to elucidate the molecular pathogenesis and genetic heterogeneity of liver cancers. This review of the genetic landscape of HCC and iCCA discusses the most recent findings from genomic profiling and the current understanding of the pathways involved in the initiation and progression of liver cancer. We highlight recent insights gained from metabolic profiling of HCC and iCCA. This knowledge will be key to developing clinically useful diagnostic/prognostic profiles, building targeted molecular and immunologic therapies, and ultimately curing these complex and heterogeneous diseases.
Collapse
Affiliation(s)
- Julian A Gingold
- Women's Health Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Dandan Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ahmed Kaseb
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Chen
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
1659
|
Mani SKK, Andrisani O. Hepatitis B Virus-Associated Hepatocellular Carcinoma and Hepatic Cancer Stem Cells. Genes (Basel) 2018; 9:genes9030137. [PMID: 29498629 PMCID: PMC5867858 DOI: 10.3390/genes9030137] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/23/2018] [Accepted: 02/23/2018] [Indexed: 02/06/2023] Open
Abstract
Chronic Hepatitis B Virus (HBV) infection is linked to hepatocellular carcinoma (HCC) pathogenesis. Despite the availability of a HBV vaccine, current treatments for HCC are inadequate. Globally, 257 million people are chronic HBV carriers, and children born from HBV-infected mothers become chronic carriers, destined to develop liver cancer. Thus, new therapeutic approaches are needed to target essential pathways involved in HCC pathogenesis. Accumulating evidence supports existence of hepatic cancer stem cells (hCSCs), which contribute to chemotherapy resistance and cancer recurrence after treatment or surgery. Understanding how hCSCs form will enable development of therapeutic strategies to prevent their formation. Recent studies have identified an epigenetic mechanism involving the downregulation of the chromatin modifying Polycomb Repressive Complex 2 (PRC2) during HBV infection, which results in re-expression of hCSC marker genes in infected hepatocytes and HBV-associated liver tumors. However, the genesis of hCSCs requires, in addition to the expression of hCSC markers cellular changes, rewiring of metabolism, cell survival, escape from programmed cell death, and immune evasion. How these changes occur in chronically HBV-infected hepatocytes is not yet understood. In this review, we will present the basics about HBV infection and hepatocarcinogenesis. Next, we will discuss studies describing the mutational landscape of liver cancers and how epigenetic mechanisms likely orchestrate cellular reprograming of hepatocytes to enable formation of hCSCs.
Collapse
Affiliation(s)
- Saravana Kumar Kailasam Mani
- Department of Basic Medical Sciences and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| | - Ourania Andrisani
- Department of Basic Medical Sciences and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
1660
|
Li YL, Zhang NY, Hu X, Chen JL, Rao MJ, Wu LW, Li QY, Zhang B, Yan W, Zhang C. Evodiamine induces apoptosis and promotes hepatocellular carcinoma cell death induced by vorinostat via downregulating HIF-1α under hypoxia. Biochem Biophys Res Commun 2018; 498:481-486. [PMID: 29505792 DOI: 10.1016/j.bbrc.2018.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 03/01/2018] [Indexed: 12/16/2022]
Abstract
Hypoxia promotes HCC progression and therapy resistance, and there is no systemic treatment for HCC patients after sorafenib resistance. Thus, it is urgent to develop potential therapeutic regimens for HCC patients by targeting hypoxia signaling. In this study, we showed that evodiamine might be a potential therapeutic medicine for HCC by suppressing HIF-1α. In addition, evodiamine could sensitize the anti-HCC effect of vorinostat in HCC cells under hypoxia. Furthermore, evodiamine plus vorinostat accelerated the degradation of HIF-1α in HCC cells under hypoxia. In general, evodiamine might be a potential therapeutic candidate for HCC patients, and evodiamine combining with vorinostat might be an attractive chemotherapy strategy for HCC treatment.
Collapse
Affiliation(s)
- Yang-Ling Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang, 310006, China
| | - Ning-Yu Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Xiu Hu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jia-Ling Chen
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China
| | - Ming-Jun Rao
- Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311402, China
| | - Lin-Wen Wu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Qing-Yu Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang, 310006, China
| | - Bo Zhang
- Hangzhou Translational Medicine Research Center, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang, 310006, China
| | - Wei Yan
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou, Zhejiang, 310006, China.
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, 310015, China.
| |
Collapse
|
1661
|
Tanaka S. Precision medicine based on surgical oncology in the era of genome-scale analysis and genome editing technology. Ann Gastroenterol Surg 2018; 2:106-115. [PMID: 29863171 PMCID: PMC5881373 DOI: 10.1002/ags3.12059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022] Open
Abstract
Accumulated evidence suggests that multiple molecular and cellular interactions promote cancer evolution in vivo. Surgical oncology is of growing significance to a comprehensive understanding of the malignant diseases for therapeutic application. We have analyzed more than 1000 clinical samples from surgically resected tissue to identify molecular biomarkers and therapeutic targets for advanced malignancies. Cancer stemness and mitotic instability were then determined as the essential predictors of aggressive phenotype with poor prognosis. Recently, whole genome/exome sequencing showed a mutational landscape underlying phenotype heterogeneity in caners. In addition, integrated genomic, epigenomic, transcriptomic, metabolic, proteomic and phenomic analyses elucidated several molecular subtypes that cluster in liver, pancreatic, biliary, esophageal and gastroenterological cancers. Identification of each molecular subtype is expected to realize the precise medicine targeting subtype-specific molecules; however, there are obstacle limitations to determine matching druggable targets or synthetic lethal interactions. Current breakthroughs in genome editing technology can provide us with unprecedented opportunity to recapitulate subtype-specific pathophysiology in vitro and in vivo. Given a great potential, on-demand editing system can design actionable strategy and revolutionize precision cancer medicine based on surgical oncology.
Collapse
Affiliation(s)
- Shinji Tanaka
- Department of Molecular OncologyTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
1662
|
Fujiwara N, Friedman SL, Goossens N, Hoshida Y. Risk factors and prevention of hepatocellular carcinoma in the era of precision medicine. J Hepatol 2018; 68:526-549. [PMID: 28989095 PMCID: PMC5818315 DOI: 10.1016/j.jhep.2017.09.016] [Citation(s) in RCA: 520] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 01/27/2023]
Abstract
Patients who develop chronic fibrotic liver disease, caused by viral or metabolic aetiologies, are at a high risk of developing hepatocellular carcinoma (HCC). Even after complete HCC tumour resection or ablation, the carcinogenic tissue microenvironment in the remnant liver can give rise to recurrent de novo HCC tumours, which progress into incurable, advanced-stage disease in most patients. Thus, early detection and prevention of HCC development is, in principle, the most impactful strategy to improve patient prognosis. However, a "one-size-fits-all" approach to HCC screening for early tumour detection, as recommended by clinical practice guidelines, is utilised in less than 20% of the target population, and the performance of screening modalities, including ultrasound and alpha-fetoprotein, is suboptimal. Furthermore, optimal screening strategies for emerging at-risk patient populations, such as those with chronic hepatitis C after viral cure, or those with non-cirrhotic, non-alcoholic fatty liver disease remain controversial. New HCC biomarkers and imaging modalities may improve the sensitivity and specificity of HCC detection. Clinical and molecular HCC risk scores will enable precise HCC risk prediction followed by tailoured HCC screening of individual patients, maximising cost-effectiveness and optimising allocation of limited medical resources. Several aetiology-specific and generic HCC chemoprevention strategies are evolving. Epidemiological and experimental studies have identified candidate chemoprevention targets and therapies, including statins, anti-diabetic drugs, and selective molecular targeted agents, although their clinical testing has been limited by the lengthy process of cancer development that requires long-term, costly studies. Individual HCC risk prediction is expected to overcome the challenge by enabling personalised chemoprevention, targeting high-risk patients for precision HCC prevention and substantially improving the dismal prognosis of HCC.
Collapse
Affiliation(s)
- Naoto Fujiwara
- Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, USA; Department of Gastroenterology, Graduate School of Medicine, University of Tokyo, Japan
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, USA
| | - Nicolas Goossens
- Division of Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, USA.
| |
Collapse
|
1663
|
Riordan JD, Feddersen CR, Tschida BR, Jackson P, Keng VW, Linden MA, Amin K, Stipp CS, Largaespada DA, Dupuy AJ. Chronic liver injury alters driver mutation profiles in hepatocellular carcinoma in mice. Hepatology 2018; 67:924-939. [PMID: 28961327 PMCID: PMC5826818 DOI: 10.1002/hep.29565] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 07/31/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023]
Abstract
Most hepatocellular carcinomas (HCCs) develop in a chronically injured liver, yet the extent to which this microenvironment promotes neoplastic transformation or influences selective pressures for genetic drivers of HCC remains unclear. We sought to determine the impact of hepatic injury in an established mouse model of HCC induced by Sleeping Beauty transposon mutagenesis. Chemically induced chronic liver injury dramatically increased tumor penetrance and significantly altered driver mutation profiles, likely reflecting distinct selective pressures. In addition to established human HCC genes and pathways, we identified several injury-associated candidates that represent promising loci for further study. Among them, we found that FIGN is overexpressed in human HCC and promotes hepatocyte invasion. We also validated Gli2's oncogenic potential in vivo, providing direct evidence that Hedgehog signaling can drive liver tumorigenesis in the context of chronic injury. Finally, we show that a subset of injury-associated candidate genes identifies two distinct classes of human HCCs. Further analysis of these two subclasses revealed significant trends among common molecular classification schemes of HCC. The genes and mechanisms identified here provide functional insights into the origin of HCC in a chronic liver damage environment. CONCLUSION A chronically damaged liver microenvironment influences the genetic mechanisms that drive hepatocarcinogenesis. (Hepatology 2018;67:924-939).
Collapse
Affiliation(s)
- Jesse D. Riordan
- Department of Anatomy & Cell Biology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Charlotte R. Feddersen
- Department of Anatomy & Cell Biology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - Barbara R. Tschida
- Masonic Cancer Center, Department of Pediatrics & Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Pauline Jackson
- Masonic Cancer Center, Department of Pediatrics & Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Vincent W. Keng
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Michael A. Linden
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Khalid Amin
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher S. Stipp
- Department of Biology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| | - David A. Largaespada
- Masonic Cancer Center, Department of Pediatrics & Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Adam J. Dupuy
- Department of Anatomy & Cell Biology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
1664
|
Arid1a regulates response to anti-angiogenic therapy in advanced hepatocellular carcinoma. J Hepatol 2018; 68:465-475. [PMID: 29113912 DOI: 10.1016/j.jhep.2017.10.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/24/2017] [Accepted: 10/18/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS AT-rich interaction domain 1a (Arid1a), a component of the chromatin remodeling complex, has emerged as a tumor suppressor gene. It is frequently mutated in hepatocellular carcinoma (HCC). However, it remains unknown how Arid1a suppresses HCC development and whether Arid1a deficiency could be exploited for therapy, we aimed to explore these questions. METHODS The expression of Arid1a in human and mouse HCCs was determined by immunohistochemical (IHC) staining. Gene expression was determined by quantitative PCR, ELISA or western blotting. Arid1a knockdown HCC cell lines were established by lentiviral-based shRNA. Tumor angiogenesis was quantified based on vessel density. The regulation of angiopoietin (Ang2) expression by Arid1a was identified by chromatin immunoprecipitation (ChIP) assay. The tumor promoting function of Arid1a loss was studied with a xenograft model in nude mice and diethylnitrosamine (DEN)-induced HCC in Arid1a conditional knockout mice. The therapeutic values of Ang2 antibody and sorafenib treatment were evaluated both in vitro and in vivo. RESULTS We demonstrate that Arid1a deficiency, occurring in advanced human HCCs, is associated with increased vessel density. Mechanistically, loss of Arid1a causes aberrant histone H3K27ac deposition at the angiopoietin-2 (Ang2) enhancer and promoter, which eventually leads to ectopic expression of Ang2 and promotes HCC development. Ang2 blockade in Arid1a-deficient HCCs significantly reduces vessel density and tumor progression. Importantly, sorafenib treatment, which suppresses H3K27 acetylation and Ang2 expression, profoundly halts the progression of Arid1a-deficient HCCs. CONCLUSIONS Arid1a-deficiency activates Ang2-dependent angiogenesis and promotes HCC progression. Loss of Arid1a in HCCs confers sensitivity to Ang2 blockade and sorafenib treatment. LAY SUMMARY AT-rich interaction domain 1a (Arid1a), is a tumor suppressor gene. Arid1a-deficiency promotes Ang2-dependent angiogenesis leading to hepatocellular carcinoma progression. Arid1a-deficiency also sensitizes tumors to Ang2 blockade by sorafenib treatment.
Collapse
|
1665
|
Hu B, Ding GY, Fu PY, Zhu XD, Ji Y, Shi GM, Shen YH, Cai JB, Yang Z, Zhou J, Fan J, Sun HC, Kuang M, Huang C. NOD-like receptor X1 functions as a tumor suppressor by inhibiting epithelial-mesenchymal transition and inducing aging in hepatocellular carcinoma cells. J Hematol Oncol 2018; 11:28. [PMID: 29482578 PMCID: PMC5828065 DOI: 10.1186/s13045-018-0573-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/11/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This study was performed to investigate the role of nucleotide-binding oligomerization domain (NOD)-like receptor X1 (NLRX1) in regulating hepatocellular carcinoma (HCC) progression. METHODS Expression levels of NLRX1 in clinical specimens and cell lines were determined by reverse transcription-polymerase chain reaction (RT-PCR) and western blot (WB). Transwell assays were conducted to evaluate the effect of NLRX1 on cell invasion, and flow cytometry was used to assess apoptosis. Expression patterns of key molecules in the phosphoinositide 3-kinase (PI3K)-AKT pathways were determined via WB. The effect of NLRX1 on cell senescence was evaluated with β-galactosidase assays. Kaplan-Meier analyses and Cox regression models were used for prognostic evaluation. RESULTS NLRX1 was downregulated in tumor tissue compared with adjacent normal liver tissue. Low tumor NLRX1 expression was identified as an independent indicator for HCC prognosis (recurrence: hazard ratio [HR] 1.87, 95% confidence interval [CI] 1.26-2.76, overall survival [OS] 2.26, 95% CI 1.44-3.56). NLRX1 over-expression (OE) significantly inhibited invasiveness ability and induced apoptosis in HCC cells. In vivo experiments showed that NLRX1 knock-down (KD) significantly promoted HCC growth. Mechanistically, NLRX1 exhibited a suppressor function by decreasing phosphorylation of AKT and thus downregulating Snail1 expression, which inhibited epithelial-mesenchymal-transition (EMT) in HCC cells. Moreover, NLRX1 OE could induce cell senescence via an AKT-P21-dependent manner. CONCLUSIONS NLRX1 acted as a tumor suppressor in HCC by inducing apoptosis, promoting senescence, and decreasing invasiveness by repressing PI3K-AKT signaling pathway. Future investigations will focus on restoring expression of NLRX1 to provide new insights into HCC treatment.
Collapse
Affiliation(s)
- Bo Hu
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Guang-Yu Ding
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Pei-Yao Fu
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo-Ming Shi
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Jia-Bin Cai
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Zhen Yang
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai, 200031 China
| | - Jian Zhou
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| | - Ming Kuang
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhong Shan Rd 2, Guangzhou, 510080 China
| | - Cheng Huang
- Department of Liver Surgery and Transplant, Liver Cancer Institute and Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Rd, Shanghai, 200032 China
- Key Laboratory for Carcinogenesis and Cancer Invasion, Chinese Ministry of Education, Shanghai, China
| |
Collapse
|
1666
|
DeLeon TT, Ahn DH, Bogenberger JM, Anastasiadis PZ, Arora M, Ramanathan RK, Aqel BA, Vasmatzis G, Truty MJ, Oklu R, Bekaii-Saab TS, Borad MJ. Novel targeted therapy strategies for biliary tract cancers and hepatocellular carcinoma. Future Oncol 2018; 14:553-566. [PMID: 29460642 DOI: 10.2217/fon-2017-0451] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Worldwide hepatobiliary cancers are the second leading cause of cancer related death. Despite their relevance, hepatobiliary cancers have a paucity of approved systemic therapy options. However, there are a number of emerging therapeutic biomarkers and therapeutic concepts that show promise. In hepatocellular carcinoma, nivolumab appears particularly promising and recently received US FDA approval. In intrahepatic cholangiocarcinoma, therapies targeting FGFR2 and IDH1 and immune checkpoint inhibitors are the furthest along and generating the most excitement. There are additional biomarkers that merit further exploration in hepatobiliary cancers including FGF19, ERRFI1, TERT, BAP1, BRAF, CDKN2A, tumor mutational burden and ERBB2 (HER2/neu). Development of new and innovative therapies would help address the unmet need for effective systemic therapies in advanced and metastatic hepatobiliary cancers.
Collapse
Affiliation(s)
- Thomas T DeLeon
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Daniel H Ahn
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - James M Bogenberger
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | | | - Mansi Arora
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Ramesh K Ramanathan
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Bashar A Aqel
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - George Vasmatzis
- Department of Molecular Medicine, Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, MN 55902, USA
| | - Mark J Truty
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, Mayo Clinic, Rochester, MN 55902, USA
| | - Rahmi Oklu
- Division of Interventional Radiology, Department of Radiology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Tanios S Bekaii-Saab
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Mitesh J Borad
- Division of Hematology Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| |
Collapse
|
1667
|
Kancherla V, Abdullazade S, Matter MS, Lanzafame M, Quagliata L, Roma G, Hoshida Y, Terracciano LM, Ng CKY, Piscuoglio S. Genomic Analysis Revealed New Oncogenic Signatures in TP53-Mutant Hepatocellular Carcinoma. Front Genet 2018; 9:2. [PMID: 29456550 PMCID: PMC5801302 DOI: 10.3389/fgene.2018.00002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/03/2018] [Indexed: 01/04/2023] Open
Abstract
The TP53 gene is the most commonly mutated gene in human cancers and mutations in TP53 have been shown to have either gain-of-function or loss-of-function effects. Using the data generated by The Cancer Genome Atlas, we sought to define the spectrum of TP53 mutations in hepatocellular carcinomas (HCCs) and their association with clinicopathologic features, and to determine the oncogenic and mutational signatures in TP53-mutant HCCs. Compared to other cancer types, HCCs harbored distinctive mutation hotspots at V157 and R249, whereas common mutation hotspots in other cancer types, R175 and R273, were extremely rare in HCCs. In terms of clinicopathologic features, in addition to the associations with chronic viral infection and high Edmondson grade, we found that TP53 somatic mutations were less frequent in HCCs with cholestasis or tumor infiltrating lymphocytes, but were more frequent in HCCs displaying necrotic areas. An analysis of the oncogenic signatures based on the genetic alterations found in genes recurrently altered in HCCs identified four distinct TP53-mutant subsets, three of which were defined by CTNNB1 mutations, 1q amplifications or 8q24 amplifications, respectively, that co-occurred with TP53 mutations. We also found that mutational signature 12, a liver cancer-specific signature characterized by T>C substitutions, was prevalent in HCCs with wild-type TP53 or with missense TP53 mutations, but not in HCCs with deleterious TP53 mutations. Finally, whereas patients with HCCs harboring deleterious TP53 mutations had worse overall and disease-free survival than patients with TP53-wild-type HCCs, patients with HCCs harboring missense TP53 mutations did not have worse prognosis. In conclusion, our results highlight the importance to consider the genetic heterogeneity among TP53-mutant HCCs in studies of biomarkers and molecular characterization of HCCs.
Collapse
Affiliation(s)
| | - Samir Abdullazade
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Matthias S Matter
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Manuela Lanzafame
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Luca Quagliata
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Guglielmo Roma
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | | |
Collapse
|
1668
|
Chianchiano P, Pezhouh MK, Kim A, Luchini C, Cameron A, Weiss MJ, He J, Voltaggio L, Oshima K, Anders RA, Wood LD. Distinction of intrahepatic metastasis from multicentric carcinogenesis in multifocal hepatocellular carcinoma using molecular alterations. Hum Pathol 2018; 72:127-134. [PMID: 29180252 PMCID: PMC6435273 DOI: 10.1016/j.humpath.2017.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 11/10/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023]
Abstract
Patients with hepatocellular carcinoma (HCC) frequently have multiple anatomically distinct tumors. In these patients, multifocal HCC could represent intrahepatic metastases (IMs) of a single cancer or multicentric carcinogenesis (MC) with multiple independent neoplasms. To determine the frequency and clinical implications of these 2 possibilities, we performed histological and molecular analysis of 70 anatomically distinct HCCs from 24 patients. We assayed mutations in the TERT promoter region by Sanger sequencing and used next-generation sequencing to analyze the entire coding regions of 7 well-characterized HCC driver genes-based on shared or discordant mutations in these genes, we classified the HCCs in each patient as IM, MC, or indeterminate. Mutations in the TERT promoter were the most common alteration in our cohort, present in 71% of tumors analyzed. Mutations in the remaining genes occurred in less than 20% of analyzed tumors. We were able to determine the relatedness in 58% of the patients analyzed: MC occurred in 41% of patients, with 33% with exclusively MC and 8% with both MC and IM. IM occurred exclusively in 17% of patients, whereas the remainder were indeterminate. This study highlights the utility of molecular analyses to determine relatedness in multifocal HCC; however, targeted sequencing can only resolve this distinction in approximately 60% of patients with multifocal HCC.
Collapse
Affiliation(s)
- Peter Chianchiano
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Maryam Kherad Pezhouh
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Amy Kim
- Division of Gastroenterology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy
| | - Andrew Cameron
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Matthew J Weiss
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Jin He
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Lysandra Voltaggio
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Kiyoko Oshima
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Robert A Anders
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Laura D Wood
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA; Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
1669
|
Guerrero-Martínez JA, Reyes JC. High expression of SMARCA4 or SMARCA2 is frequently associated with an opposite prognosis in cancer. Sci Rep 2018; 8:2043. [PMID: 29391527 PMCID: PMC5794756 DOI: 10.1038/s41598-018-20217-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 01/16/2018] [Indexed: 12/21/2022] Open
Abstract
The gene encoding the ATPase of the chromatin remodeling SWI/SNF complexes SMARCA4 (BRG1) is often mutated or silenced in tumors, suggesting a role as tumor suppressor. Nonetheless, recent reports show requirement of SMARCA4 for tumor cells growth. Here, we performed a computational meta-analysis using gene expression, prognosis, and clinicopathological data to clarify the role of SMARCA4 and the alternative SWI/SNF ATPase SMARCA2 (BRM) in cancer. We show that while the SMARCA4 gene is mostly overexpressed in tumors, SMARCA2 is almost invariably downexpressed in tumors. High SMARCA4 expression was associated with poor prognosis in many types of tumors, including liver hepatocellular carcinoma (LIHC), and kidney renal clear cell carcinoma (KIRC). In contrast, high SMARCA2 expression was associated with good prognosis. We compared tumors with high versus low expression of SMARCA4 or SMARCA2 in LIHC and KIRC cohorts from The Cancer Genome Atlas. While a high expression of SMARCA4 is associated with aggressive tumors, a high expression of SMARCA2 is associated with benign differentiated tumors, suggesting that SMARCA4 and SMARCA2 play opposite roles in cancer. Our results demonstrate that expression of SMARCA4 and SMARCA2 have a high prognostic value and challenge the broadly accepted general role of SMARCA4 as a tumor suppressor.
Collapse
Affiliation(s)
- Jose A Guerrero-Martínez
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO). Av. Americo Vespucio 24, 41092, Seville, Spain
| | - Jose C Reyes
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla-Universidad Pablo de Olavide (CSIC-USE-UPO). Av. Americo Vespucio 24, 41092, Seville, Spain.
| |
Collapse
|
1670
|
Gerbes A, Zoulim F, Tilg H, Dufour J, Bruix J, Paradis V, Salem R, Peck–Radosavljevic M, Galle PR, Greten TF, Nault J, Avila MA. Gut roundtable meeting paper: selected recent advances in hepatocellular carcinoma. Gut 2018; 67:380-388. [PMID: 29150490 PMCID: PMC6309825 DOI: 10.1136/gutjnl-2017-315068] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/03/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) ranks number three among the most frequent causes of death from solid tumors worldwide. With obesity and fatty liver diseases as risk factors on the rise, HCC represents an ever increasing challenge. While there is still no curative treatment for most patients numerous novel drugs have been proposed, but most ultimately failed in phase III trials. This manuscript targets therapeutic advances and most burning issues. Expert key point summaries and urgent research agenda are provided regarding risk factors, including microbiota, need for prognostic and predictive biomarkers and the equivocal role of liver biopsy. Therapeutic topics highlighted are locoregional techniques, combination therapies and the potential of immunotherapy. Finally the manuscript provides a critical evaluation of novel targets and strategies for personalized treatment of HCC.
Collapse
Affiliation(s)
- Alexander Gerbes
- Department of Medicine 2, Liver Center Munich, University Hospital, LMU, Munich, Germany
| | - Fabien Zoulim
- Hepatology Department at the Hospices Civils de Lyon, Lyon University, Institut Universitaire de France, Lyon, France
- Viral Hepatitis Team, Cancer Research Center of Lyon (CRCL), INSERM, Lyon University, Lyon, France
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology, Innsbruck Medical University, Innsbruck, Austria
| | - Jean–François Dufour
- Hepatology, Department of Clinical Research, University of Bern, Bern, Switzerland
- University Clinic of Visceral Surgery and Medicine, Inselspital Bern, Bern, Switzerland
| | - Jordi Bruix
- BCLC Group, Liver Unit, Hospital Clínic, Universitat de Barcelona, IDIBAPS, CIBEREHD, Barcelona, Spain
| | - Valérie Paradis
- Pathology Department Beaujon Hospital & INSERM, INSERM 1149, University Paris–Diderot, Paris, France
| | - Riad Salem
- Department of Radiology, Section of Vascular and Interventional Radiology, Northwestern University, Chicago, Illinois, USA
| | - Markus Peck–Radosavljevic
- Department of Gastroenterology & Hepatology, Endocrinology and Nephrology, Klinikum Klagenfurt am Wörthersee, Klagenfurt, Austria
| | - Peter R Galle
- Department of Internal Medicine, University Medical Center I, Mainz, Germany
| | - Tim F Greten
- National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
- Gastrointestinal Malignancy Section, Thoracic and GI Oncology Branch, Center for Cancer Research, Bethesda, Maryland, USA
| | - Jean–Charles Nault
- Unité Mixte de Recherche 1162, Génomique fonctionnelle des tumeurs solides, Institut National de la Santé et de la Recherche Médicale, Paris, France
- Liver unit, Hôpital Jean Verdier, Hôpitaux Universitaires Paris–Seine–Saint–Denis, Assistance–Publique Hôpitaux de Paris, Paris, France
- Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d’Universités et Etablissements Sorbonne Paris Cité, Paris, France
| | - Matias A Avila
- Programme of Hepatology, CIMA, IdiSNA, CIBERehd, University of Navarra, Pamplona, Spain
| |
Collapse
|
1671
|
Affiliation(s)
- Jesper B Andersen
- Biotech Research & Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
1672
|
Zhang S, Wang J, Wang H, Fan L, Fan B, Zeng B, Tao J, Li X, Che L, Cigliano A, Ribback S, Dombrowski F, Chen B, Cong W, Wei L, Calvisi DF, Chen X. Hippo Cascade Controls Lineage Commitment of Liver Tumors in Mice and Humans. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:995-1006. [PMID: 29378174 DOI: 10.1016/j.ajpath.2017.12.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/11/2017] [Accepted: 12/28/2017] [Indexed: 02/05/2023]
Abstract
Primary liver cancer consists mainly of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). A subset of human HCCs expresses a ICC-like gene signature and is classified as ICC-like HCC. The Hippo pathway is a critical regulator of normal and malignant liver development. However, the precise function(s) of the Hippo cascade along liver carcinogenesis remain to be fully delineated. The role of the Hippo pathway in a murine mixed HCC/ICC model induced by activated forms of AKT and Ras oncogenes (AKT/Ras) was investigated. The authors demonstrated the inactivation of Hippo in AKT/Ras liver tumors leading to nuclear localization of Yap and TAZ. Coexpression of AKT/Ras with Lats2, which activates Hippo, or the dominant negative form of TEAD2 (dnTEAD2), which blocks Yap/TAZ activity, resulted in delayed hepatocarcinogenesis and elimination of ICC-like lesions in the liver. Mechanistically, Notch2 expression was found to be down-regulated by the Hippo pathway in liver tumors. Overexpression of Lats2 or dnTEAD2 in human HCC cell lines inhibited their growth and led to the decreased expression of ICC-like markers, as well as Notch2 expression. Altogether, this study supports the key role of the Hippo cascade in regulating the differentiation status of liver tumors.
Collapse
Affiliation(s)
- Shanshan Zhang
- Tumor Immunology and Gene Therapy Center, Second Military Medical University, Shanghai, China; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jingxiao Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Haichuan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Liver Transplantation Division, Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lingling Fan
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Biao Fan
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Department of Gastrointestinal Surgery, Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Billy Zeng
- Department of Pediatrics, University of California, San Francisco, California; Institute for Computational Health Sciences, University of California, San Francisco, California
| | - Junyan Tao
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Xiaolei Li
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Li Che
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Antonio Cigliano
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Frank Dombrowski
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Bin Chen
- Department of Pediatrics, University of California, San Francisco, California; Institute for Computational Health Sciences, University of California, San Francisco, California
| | - Wenming Cong
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Second Military Medical University, Shanghai, China
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California.
| |
Collapse
|
1673
|
Hepatic SMARCA4 predicts HCC recurrence and promotes tumour cell proliferation by regulating SMAD6 expression. Cell Death Dis 2018; 9:59. [PMID: 29352111 PMCID: PMC5833410 DOI: 10.1038/s41419-017-0090-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/29/2017] [Accepted: 10/23/2017] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer and is typically diagnosed at advanced stages. Identification and characterisation of genes within amplified and deleted chromosomal loci can provide new insights into the pathogenesis of cancer and lead to new approaches for diagnosis and therapy. In our previous study, we found a recurrent region of copy number amplification at 19p13.2 in hepatocellular carcinoma (HCC). In the present study, we performed integrated copy number analysis and expression profiling at this locus and a putative cancer gene, SMARCA4/BRG1, was uncovered in this region. BRG1 is a part of the large ATP-dependent chromatin remodelling complex SWI/SNF. The function of BRG1 in various cancers is unclear, including its role in HCC tumorigenesis. Here, we found that BRG1 is upregulated in HCC and that its level significantly correlates with cancer progression in HCC patients. Importantly, we also found that nuclear expression of BRG1 predicts early recurrence for HCC patients. Furthermore, we demonstrated that BRG1 promotes HCC cell proliferation in vitro and in vivo. BRG1 was observed not only to facilitate S-phase entry but also to attenuate cell apoptosis. Finally, we discovered that one of the mechanisms by which BRG1 promotes cell proliferation is the upregulation of SMAD6. These findings highlight the important role of BRG1 in the regulation of HCC proliferation and provide valuable information for cancer prognosis and treatment.
Collapse
|
1674
|
Joshi JJ, Coffey H, Corcoran E, Tsai J, Huang CL, Ichikawa K, Prajapati S, Hao MH, Bailey S, Wu J, Rimkunas V, Karr C, Subramanian V, Kumar P, MacKenzie C, Hurley R, Satoh T, Yu K, Park E, Rioux N, Kim A, Lai WG, Yu L, Zhu P, Buonamici S, Larsen N, Fekkes P, Wang J, Warmuth M, Reynolds DJ, Smith PG, Selvaraj A. H3B-6527 Is a Potent and Selective Inhibitor of FGFR4 in FGF19-Driven Hepatocellular Carcinoma. Cancer Res 2018; 77:6999-7013. [PMID: 29247039 DOI: 10.1158/0008-5472.can-17-1865] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/23/2017] [Accepted: 10/10/2017] [Indexed: 01/18/2023]
Abstract
Activation of the fibroblast growth factor receptor FGFR4 by FGF19 drives hepatocellular carcinoma (HCC), a disease with few, if any, effective treatment options. While a number of pan-FGFR inhibitors are being clinically evaluated, their application to FGF19-driven HCC may be limited by dose-limiting toxicities mediated by FGFR1-3 receptors. To evade the potential limitations of pan-FGFR inhibitors, we generated H3B-6527, a highly selective covalent FGFR4 inhibitor, through structure-guided drug design. Studies in a panel of 40 HCC cell lines and 30 HCC PDX models showed that FGF19 expression is a predictive biomarker for H3B-6527 response. Moreover, coadministration of the CDK4/6 inhibitor palbociclib in combination with H3B-6527 could effectively trigger tumor regression in a xenograft model of HCC. Overall, our results offer preclinical proof of concept for H3B-6527 as a candidate therapeutic agent for HCC cases that exhibit increased expression of FGF19. Cancer Res; 77(24); 6999-7013. ©2017 AACR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jeremy Wu
- H3 Biomedicine, Cambridge, Massachusetts
| | | | - Craig Karr
- H3 Biomedicine, Cambridge, Massachusetts
| | | | | | | | | | | | - Kun Yu
- H3 Biomedicine, Cambridge, Massachusetts
| | | | | | - Amy Kim
- H3 Biomedicine, Cambridge, Massachusetts
| | | | - Lihua Yu
- H3 Biomedicine, Cambridge, Massachusetts
| | - Ping Zhu
- H3 Biomedicine, Cambridge, Massachusetts
| | | | | | | | - John Wang
- H3 Biomedicine, Cambridge, Massachusetts
| | | | | | | | | |
Collapse
|
1675
|
Marquardt JU. The Role of Transforming Growth Factor-β in Human Hepatocarcinogenesis: Mechanistic and Therapeutic Implications From an Integrative Multiomics Approach. Gastroenterology 2018; 154:17-20. [PMID: 29174548 DOI: 10.1053/j.gastro.2017.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jens U Marquardt
- Department of Medicine I, Lichtenberg Research Group, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
1676
|
Fang F, Wang X, Song T. Five-CpG-based prognostic signature for predicting survival in hepatocellular carcinoma patients. Cancer Biol Med 2018; 15:425-433. [PMID: 30766752 PMCID: PMC6372912 DOI: 10.20892/j.issn.2095-3941.2018.0027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Hepatocellular carcinoma (HCC) is a common malignancy associated with high morbidity and mortality rates worldwide. Early diagnosis plays an important role in the improvement of HCC prognosis. Methods: In this study, we conducted a comprehensive analysis of HCC DNA methylation and gene expression datasets in The Cancer Genome Atlas (TCGA), to identify a prognostic signature for HCC diagnosis and survival prediction. First, we identified differential methylation CpG (dmCpG) sites in HCC samples and compared them with those in adjacent normal liver tissues; this was followed by univariate analysis and Sure Independence Screening (SIS) in the training set. The robustness of the identified prognostic signature was evaluated using the testing set. To explore the biological processes involved in HCC progression, we also performed functional enrichment analysis for overlapping genes between genes containing dmCpG sites (DMGs) and differential expression genes (DEGs) in HCC patients, using data from the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Results: As a result, we identified five CpG sites that were significantly associated with HCC survival through univariate analysis and SIS. Univariate analysis of clinical characteristics identified age and risk factors (including alcohol consumption and smoking) as independent factors that indicated HCC survival. Multivariate analysis indicated that the integrated prognostic signature (weighted combination of the five CpG sites) that took age and risk factors into consideration resulted in more accurate survival prediction. Conclusions: This study provides a novel signature for predicting HCC survival, and should be helpful for early HCC diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Feng Fang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiaoqing Wang
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
1677
|
Castrillo JI, Lista S, Hampel H, Ritchie CW. Systems Biology Methods for Alzheimer’s Disease Research Toward Molecular Signatures, Subtypes, and Stages and Precision Medicine: Application in Cohort Studies and Trials. Methods Mol Biol 2018; 1750:31-66. [PMID: 29512064 DOI: 10.1007/978-1-4939-7704-8_3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Juan I Castrillo
- Genetadi Biotech S.L. Parque Tecnológico de Bizkaia, Derio, Bizkaia, Spain.
| | - Simone Lista
- AXA Research Fund & UPMC Chair, F-75013, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | - Harald Hampel
- AXA Research Fund & UPMC Chair, F-75013, Paris, France
- Sorbonne Université, AP-HP, GRC n° 21, Alzheimer Precision Medicine (APM), Hôpital de la Pitié-Salpêtrière, Boulevard de l'hôpital, F-75013, Paris, France
- Institut du Cerveau et de la Moelle Épinière (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l'hôpital, F-75013, Paris, France
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Boulevard de l'hôpital, F-75013, Paris, France
| | - Craig W Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
1678
|
Biophotonic detection of high order chromatin alterations in field carcinogenesis predicts risk of future hepatocellular carcinoma: A pilot study. PLoS One 2018; 13:e0197427. [PMID: 29771950 PMCID: PMC5957523 DOI: 10.1371/journal.pone.0197427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/02/2018] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Hepatocellular carcinoma (HCC) results from chronic inflammation/cirrhosis. Unfortunately, despite use of radiological/serological screening techniques, HCC ranks as a leading cause of cancer deaths. Our group has used alterations in high order chromatin as a marker for field carcinogenesis and hence risk for a variety of cancers (including colon, lung, prostate, ovarian, esophageal). In this study we wanted to address whether these chromatin alterations occur in HCC and if it could be used for risk stratification. EXPERIMENTAL DESIGN A case control study was performed in patients with cirrhosis who went on to develop HCC and patients with cirrhosis who did not develop cancer. We performed partial wave spectroscopic microscopy (PWS) which measures nanoscale alterations on formalin fixed deparaffinized liver biopsy specimens, 17 progressors and 26 non-progressors. Follow up was 2089 and 2892 days, respectively. RESULTS PWS parameter disorder strength Ld were notably higher for the progressors (Ld = 1.47 ± 0.76) than the non-progressors (Ld = 1.00 ± 0.27) (p = 0.024). Overall, the Cohen's d effect size was 0.907 (90.7%). AUROC analysis yielded an area of 0.70. There was no evidence of confounding by gender, age, BMI, smoking status and race. CONCLUSIONS High order chromatin alterations, as detected by PWS, is altered in pre-malignant hepatocytes with cirrhosis and may predict future risk of HCC.
Collapse
|
1679
|
Chemical Modulation of WNT Signaling in Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:245-269. [DOI: 10.1016/bs.pmbts.2017.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
1680
|
Wnt Ligands as a Part of the Stem Cell Niche in the Intestine and the Liver. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 153:1-19. [DOI: 10.1016/bs.pmbts.2017.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
1681
|
Nguyen T, Le A. The Metabolism of Renal Cell Carcinomas and Liver Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1063:107-118. [DOI: 10.1007/978-3-319-77736-8_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
1682
|
Chen J, Zaidi S, Rao S, Chen JS, Phan L, Farci P, Su X, Shetty K, White J, Zamboni F, Wu X, Rashid A, Pattabiraman N, Mazumder R, Horvath A, Wu RC, Li S, Xiao C, Deng CX, Wheeler DA, Mishra B, Akbani R, Mishra L. Analysis of Genomes and Transcriptomes of Hepatocellular Carcinomas Identifies Mutations and Gene Expression Changes in the Transforming Growth Factor-β Pathway. Gastroenterology 2018; 154:195-210. [PMID: 28918914 PMCID: PMC6192529 DOI: 10.1053/j.gastro.2017.09.007] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/22/2017] [Accepted: 09/05/2017] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Development of hepatocellular carcinoma (HCC) is associated with alterations in the transforming growth factor-beta (TGF-β) signaling pathway, which regulates liver inflammation and can have tumor suppressor or promoter activities. Little is known about the roles of specific members of this pathway at specific of HCC development. We took an integrated approach to identify and validate the effects of changes in this pathway in HCC and identify therapeutic targets. METHODS We performed transcriptome analyses for a total of 488 HCCs that include data from The Cancer Genome Atlas. We also screened 301 HCCs reported in the Catalogue of Somatic Mutations in Cancer and 202 from Cancer Genome Atlas for mutations in genome sequences. We expressed mutant forms of spectrin beta, non-erythrocytic 1 (SPTBN1) in HepG2, SNU398, and SNU475 cells and measured phosphorylation, nuclear translocation, and transcriptional activity of SMAD family member 3 (SMAD3). RESULTS We found somatic mutations in at least 1 gene whose product is a member of TGF-β signaling pathway in 38% of HCC samples. SPTBN1 was mutated in the largest proportion of samples (12 of 202, 6%). Unsupervised clustering of transcriptome data identified a group of HCCs with activation of the TGF-β signaling pathway (increased transcription of genes in the pathway) and a group of HCCs with inactivation of TGF-β signaling (reduced expression of genes in this pathway). Patients with tumors with inactivation of TGF-β signaling had shorter survival times than patients with tumors with activation of TGF-β signaling (P = .0129). Patterns of TGF-β signaling correlated with activation of the DNA damage response and sirtuin signaling pathways. HepG2, SNU398, and SNU475 cells that expressed the D1089Y mutant or with knockdown of SPTBN1 had increased sensitivity to DNA crosslinking agents and reduced survival compared with cells that expressed normal SPTBN1 (controls). CONCLUSIONS In genome and transcriptome analyses of HCC samples, we found mutations in genes in the TGF-β signaling pathway in almost 40% of samples. These correlated with changes in expression of genes in the pathways; up-regulation of genes in this pathway would contribute to inflammation and fibrosis, whereas down-regulation would indicate loss of TGF-β tumor suppressor activity. Our findings indicate that therapeutic agents for HCCs can be effective, based on genetic features of the TGF-β pathway; agents that block TGF-β should be used only in patients with specific types of HCCs.
Collapse
Affiliation(s)
- Jian Chen
- Departments of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sobia Zaidi
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC
| | - Shuyun Rao
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC
| | - Jiun-Sheng Chen
- Departments of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Liem Phan
- Departments of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrizia Farci
- Hepatic Pathogenesis Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Xiaoping Su
- Departments of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirti Shetty
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jon White
- Institute of Clinical Research, Veterans Affairs Medical Center, Washington, DC
| | - Fausto Zamboni
- Department of General Surgery, Liver and Pancreas Transplantation, Brotzu Hospital, Cagliari, Italy
| | - Xifeng Wu
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Asif Rashid
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nagarajan Pattabiraman
- Department of Biochemistry and Molecular Medicine, McCormick Genomic and Proteomic Center, George Washington University, Washington, DC
| | - Raja Mazumder
- Department of Biochemistry and Molecular Medicine, McCormick Genomic and Proteomic Center, George Washington University, Washington, DC
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, McCormick Genomic and Proteomic Center, George Washington University, Washington, DC
| | - Ray-Chang Wu
- Department of Biochemistry and Molecular Biology, George Washington University, Washington, DC
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cuiying Xiao
- Genetics of Development and Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Chu-Xia Deng
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC; Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - Bibhuti Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC; Institute of Clinical Research, Veterans Affairs Medical Center, Washington, DC
| | - Rehan Akbani
- Departments of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lopa Mishra
- Center for Translational Medicine, Department of Surgery, George Washington University, Washington, DC; Institute of Clinical Research, Veterans Affairs Medical Center, Washington, DC.
| |
Collapse
|
1683
|
Ding Z, Shi C, Jiang L, Tolstykh T, Cao H, Bangari DS, Ryan S, Levit M, Jin T, Mamaat K, Yu Q, Qu H, Hopke J, Cindhuchao M, Hoffmann D, Sun F, Helms MW, Jahn-Hofmann K, Scheidler S, Schweizer L, Fang DD, Pollard J, Winter C, Wiederschain D. Oncogenic dependency on β-catenin in liver cancer cell lines correlates with pathway activation. Oncotarget 2017; 8:114526-114539. [PMID: 29383099 PMCID: PMC5777711 DOI: 10.18632/oncotarget.21298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/13/2017] [Indexed: 12/18/2022] Open
Abstract
Hepatocellular carcinoma (HCC) represents a serious public health challenge with few therapeutic options available to cancer patients.Wnt/β-catenin pathway is thought to play a significant role in HCC pathogenesis. In this study, we confirmed high frequency of CTNNB1 (β-catenin) mutations in two independent cohorts of HCC patients and demonstrated significant upregulation of β-catenin protein in the overwhelming majority of HCC patient samples, patient-derived xenografts (PDX) and established cell lines. Using genetic tools validated for target specificity through phenotypic rescue experiments, we went on to investigate oncogenic dependency on β-catenin in an extensive collection of human HCC cells lines. Our results demonstrate that dependency on β-catenin generally tracks with its activation status. HCC cell lines that harbored activating mutations in CTNNB1 or displayed elevated levels of non-phosphorylated (active) β-catenin were significantly more sensitive to β-catenin siRNA treatment than cell lines with wild-type CTNNB1 and lower active β-catenin. Finally, significant therapeutic benefit of β-catenin knock-down was demonstrated in established HCC tumor xenografts using doxycycline-inducible shRNA system. β-catenin downregulation and tumor growth inhibition was associated with reduction in AXIN2, direct transcriptional target of β-catenin, and decreased cancer cell proliferation as measured by Ki67 staining. Taken together, our data highlight fundamental importance of aberrant β-catenin signaling in the maintenance of oncogenic phenotype in HCC.
Collapse
Affiliation(s)
- Zhihu Ding
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | - Chaomei Shi
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | - Lan Jiang
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | | | - Hui Cao
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | | | - Susan Ryan
- Sanofi Translational In Vivo Models, Framingham, MA, USA
| | | | - Taiguang Jin
- Sanofi Asia Pacific R&D Hub, Shanghai, People’s Republic of China
| | - Karl Mamaat
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
- Current address: Leica Biosystems, Boston, MA
| | - Qunyan Yu
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | - Hui Qu
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | - Joern Hopke
- Sanofi Biologics Research/Molecular Screening Technology, Cambridge, MA, USA
| | - May Cindhuchao
- Sanofi Biologics Research/Molecular Screening Technology, Cambridge, MA, USA
| | - Dietmar Hoffmann
- Sanofi Biologics Research/Molecular Screening Technology, Cambridge, MA, USA
| | - Fangxian Sun
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | - Mike W. Helms
- Sanofi Biologics Research/Nucleic Acid Therapeutics, Frankfurt am Main, Germany
| | | | - Sabine Scheidler
- Sanofi Biologics Research/Nucleic Acid Therapeutics, Frankfurt am Main, Germany
| | - Liang Schweizer
- Sanofi Asia Pacific R&D Hub, Shanghai, People’s Republic of China
- Current address: Harbour BioMed, Shanghai, People’s Republic of China
| | - Douglas D. Fang
- Discovery Services, WuXi AppTec Co., Shanghai, China
- Current address: Ascentage Pharma Group, Ltd., Suzhou, People’s Republic of China
| | - Jack Pollard
- Sanofi Oncology Therapeutic Area, Cambridge, MA, USA
| | | | | |
Collapse
|
1684
|
Barnault R, Lahlali T, Plissonnier ML, Romero-López C, Laverdure N, Ducarouge B, Rivoire M, Mehlen P, Zoulim F, Parent R. Hepatocellular carcinoma-associated depletion of the netrin-1 receptor Uncoordinated Phenotype-5A (UNC5A) skews the hepatic unfolded protein response towards prosurvival outcomes. Biochem Biophys Res Commun 2017; 495:2425-2431. [PMID: 29277614 DOI: 10.1016/j.bbrc.2017.12.129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
Abstract
In the liver, HBV and HCV infections, exposure to toxics, genetic and metabolic disorders may induce endoplasmic reticulum (ER) stress and the unfolding protein response (UPR). The UPR allows cells to reach ER homeostasis after lumen overload, but also fosters survival of damaged cells and therefore HCC onset. Dependence receptors such as UNC5A trigger apoptosis when unbound to their ligands. We have previously shown that the main dependence receptor ligand, netrin-1, could protect cells against UPR-induced apoptosis through sustained translation. In this study, we show that UNC5A is cumulatively downregulated by the UPR at the transcriptional level in vitro and at the translational level both in vitro and in vivo. We have found that the 5'-untranslated region of the UNC5A mRNA shares a certain homology degree with that of netrin-1, suggesting linked translational regulatory mechanisms, at least during the initial stages of the UPR. RNAi and forced expression studies identified UNC5A as a modulator of cell death in the context of the UPR. UNC5A decrease of association with polysomes and expression oriented cells towards UPR-associated hepatocytic survival. Such data indicate that cooperation between the UPR and UNC5A depletion as previously observed by ourselves in HCC patients samples may foster liver cancer development and growth.
Collapse
Affiliation(s)
- Romain Barnault
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Thomas Lahlali
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Marie-Laure Plissonnier
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Cristina Romero-López
- Instituto de Parasitología y Biomedicina López-Neyra IPBLN-CSIC, CSIC, Granada, Spain.
| | - Noémie Laverdure
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Benjamin Ducarouge
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Michel Rivoire
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France.
| | - Patrick Mehlen
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| | - Fabien Zoulim
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France; Service d'Hépatogastroentérologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.
| | - Romain Parent
- INSERM U1052 & CNRS UMR5286, Cancer Research Centre of Lyon, Lyon, France; University of Lyon, Lyon, France; DevWeCan Laboratories of Excellence Network (Labex), Lyon, France.
| |
Collapse
|
1685
|
Lee B, Hutchinson R, Wong HL, Tie J, Putoczki T, Tran B, Gibbs P, Christie M. Emerging biomarkers for immunomodulatory cancer treatment of upper gastrointestinal, pancreatic and hepatic cancers. Semin Cancer Biol 2017; 52:241-252. [PMID: 29258858 DOI: 10.1016/j.semcancer.2017.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Carcinomas of the oesophagus, stomach, pancreas and liver are common and account for a disproportionately high number of cancer deaths. There is a need for new treatment options for patients with advanced disease. Immunomodulatory treatments including immune checkpoint blockade offer a promising new approach, with efficacy shown in other solid tumour types. However, only a small proportion of patients with carcinomas of the oesophagus, stomach, pancreas and liver have responded to single agent checkpoint inhibitors, and there is a need for markers that are predictive of response to guide treatment of individual patients. Predictive markers may include epidemiological factors such as ethnicity, the genomic status of the tumour, circulating markers, expression of immune checkpoint molecules, and other features of the stromal/immune response at the site of the tumour. This review will focus on predictive biomarkers for immune checkpoint blockade in oesophageal, gastric, pancreatic and hepatocellular carcinomas, including the genomic context and immune landscape in which they occur. Pancreatic carcinomas are largely resistant to immune checkpoint inhibition in trials to date, therefore emerging immunomodulatory treatments in this tumour type are also reviewed.
Collapse
Affiliation(s)
- Belinda Lee
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ryan Hutchinson
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Hui-Li Wong
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Jeanne Tie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tracy Putoczki
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ben Tran
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Peter Gibbs
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Oncology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Michael Christie
- Systems Biology and Personalised Medicine Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Pathology, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
1686
|
Acquisition of Cholangiocarcinoma Traits during Advanced Hepatocellular Carcinoma Development in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:656-671. [PMID: 29248454 DOI: 10.1016/j.ajpath.2017.11.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/01/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Past studies have identified hepatic tumors with mixed hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC) characteristics that have a more aggressive behavior and a poorer prognosis than classic HCC. Whether this pathologic heterogeneity is due to a cell of origin of bipotent liver progenitors or the plasticity of cellular constituents comprising these tumors remains debated. In this study, we investigated the potential acquisition of CC-like traits during advanced development of HCC in mice. Primary and rare high-grade HCC developed in a genetic mouse model. A mouse model of highly efficient HCC invasion and metastasis by orthotopic transplantation of liver cancer organoids propagated from primary tumors in the genetic model was further developed. Invasive/metastatic tumors developed in both models closely recapitulated advanced human HCC and displayed a striking acquisition of CC-related pathologic and molecular features, which was absent in the primary HCC tumors. Our study directly demonstrates the pathologic evolution of HCC during advanced tumor development, providing the first evidence that tumors with mixed HCC and CC features, or at least a subset of these tumors, represent a more advanced developmental stage of HCC. Finally, liver cancer organoid-generated high-grade tumors exhibited significantly increased extracellular vesicle secretion, suggesting that identifying tumor-specific extracellular vesicle proteins in plasma may be a promising tool for liver cancer detection.
Collapse
|
1687
|
Yoo S, Wang W, Wang Q, Fiel MI, Lee E, Hiotis SP, Zhu J. A pilot systematic genomic comparison of recurrence risks of hepatitis B virus-associated hepatocellular carcinoma with low- and high-degree liver fibrosis. BMC Med 2017; 15:214. [PMID: 29212479 PMCID: PMC5719570 DOI: 10.1186/s12916-017-0973-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/08/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chronic hepatitis B virus (HBV) infection leads to liver fibrosis, which is a major risk factor in hepatocellular carcinoma (HCC) and an independent risk factor of recurrence after HCC tumor resection. The HBV genome can be inserted into the human genome, and chronic inflammation may trigger somatic mutations. However, how HBV integration and other genomic changes contribute to the risk of tumor recurrence with regards to the different degree of liver fibrosis is not clearly understood. METHODS We sequenced mRNAs of 21 pairs of tumor and distant non-neoplastic liver tissues of HBV-HCC patients and performed comprehensive genomic analyses of our RNAseq data and public available HBV-HCC sequencing data. RESULTS We developed a robust pipeline for sensitively identifying HBV integration sites based on sequencing data. Simulations showed that our method outperformed existing methods. Applying it to our data, 374 and 106 HBV host genes were identified in non-neoplastic liver and tumor tissues, respectively. When applying it to other RNA sequencing datasets, consistently more HBV integrations were identified in non-neoplastic liver than in tumor tissues. HBV host genes identified in non-neoplastic liver samples significantly overlapped with known tumor suppressor genes. More significant enrichment of tumor suppressor genes was observed among HBV host genes identified from patients with tumor recurrence, indicating the potential risk of tumor recurrence driven by HBV integration in non-neoplastic liver tissues. We also compared SNPs of each sample with SNPs in a cancer census database and inferred samples' pathogenic SNP loads. Pathogenic SNP loads in non-neoplastic liver tissues were consistently higher than those in normal liver tissues. Additionally, HBV host genes identified in non-neoplastic liver tissues significantly overlapped with pathogenic somatic mutations, suggesting that HBV integration and somatic mutations targeting the same set of genes are important to tumorigenesis. HBV integrations and pathogenic mutations showed distinct patterns between low and high liver fibrosis patients with regards to tumor recurrence. CONCLUSIONS The results suggest that HBV integrations and pathogenic SNPs in non-neoplastic tissues are important for tumorigenesis and different recurrence risk models are needed for patients with low and high degrees of liver fibrosis.
Collapse
Affiliation(s)
- Seungyeul Yoo
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenhui Wang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Wang
- Department of Surgery, Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eunjee Lee
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Sema4, a Mount Sinai venture, Stamford, CT, USA
| | - Spiros P Hiotis
- Department of Surgery, Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Sema4, a Mount Sinai venture, Stamford, CT, USA.
| |
Collapse
|
1688
|
Zhang Z, Xu L, Sun C. Comprehensive characterization of cancer genes in hepatocellular carcinoma genomes. Oncol Lett 2017; 15:1503-1510. [PMID: 29434842 PMCID: PMC5777097 DOI: 10.3892/ol.2017.7521] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022] Open
Abstract
The present study was performed to detect moderate or low-frequency mutated cancer driver genes in hepatocellular carcinoma (HCC), using OncodriveFM and Dendrix. Following this, integrated analyses were conducted on these novel cancer driver genes. A total of 112,980 somatic mutations were retrieved from TCGA and classified into 11 categories based on their function. Driver genes and pathways were predicted by OncodriveFM and Dendrix, followed by differential expression, DNA-methylation, copy number variations and survival analyses. Overall, non-synonymous mutations accounted for >60% (72,149/112, 980) of total variants, 108 and 3 driver genes were determined by OncodriveFM and Dendrix, respectively. Tumor protein p53, SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4, smad family member 3, RB transcriptional corepressor 1, catenin β 1, smad family member 4, mitogen-activated protein kinase 1 and TSC complex subunit 2 are at the core of the driver gene interaction network. Two genes, transportin 1 (TNPO1) and chaperonin containing TCP1 subunit 3 (CCT3), were hypomethylated and overexpressed, and high expression of TNPO1 and CCT3 indicated a poor prognosis in patients with HCC. β-carotene oxygenase 2 was hypermethylated, under-expressed and associated with favorable prognosis in HCC. The present study has identified a set of novel cancer genes and pathways, offering potential therapeutic targets and prognostic biomarkers for the treatment of HCC.
Collapse
Affiliation(s)
- Zhihao Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Liping Xu
- Department of Liver, Gallbladder and Pancreas Surgery, Ningbo First Hospital, Ningbo, Zhejiang 315010, P.R. China
| | - Changyu Sun
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
1689
|
Wang N, Tan HY, Chan YT, Guo W, Li S, Feng Y. Identification of WT1 as determinant of heptatocellular carcinoma and its inhibition by Chinese herbal medicine Salvia chinensis Benth and its active ingredient protocatechualdehyde. Oncotarget 2017; 8:105848-105859. [PMID: 29285297 PMCID: PMC5739684 DOI: 10.18632/oncotarget.22406] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 09/22/2017] [Indexed: 01/02/2023] Open
Abstract
Candidates from Chinese herbal Medicine might be preferable in drug discovery as the abundant experiences of traditional use usually hint the clinical efficacy. In this study, we screened the anti-tumour effect of several commonly used Chinese herbal Medicines on human hepatocellular carcinoma cells (HCC). We identified that Salvia chinensia Benth. (Shijianchuan in Chinese, SJC) exhibited prominent in vitro inhibition of HCC cells and suppressed the orthotopic growth of HCC in the liver of mice and repressed the lung metastasis of tumour cells. Using a pathway-specific PCR array and Gene Ontology analysis, we identified that Wnt/β-catenin pathway was associated with the suppressive effect of SJC on HCC cell proliferation and cell cycle progression. SJC repressed transcription activity of Wnt/β-catenin pathway and reduced expression of β-catenin in GSK-3β-independent but promoter-specific transcription inhibition mechanism. The suppressive effect of SJC on β-catenin expression and its transcription activity was associated with Wilms' tumor 1 (WT1) protein. WT1 was overexpressed in HCC tissues, and was negatively correlated to the overall survival of HCC patients. WT1 promoted proliferation and invasion of HCC cells, as well as β-catenin-dependent transcription activation of Wnt products, while knockdown of WT1 had the opposite effect. Docking experiment revealed that protocatechualdehyde (PCA) might be the active component of the herb. PCA suppressed transcription activity of Wnt/β-catenin pathway in WT1-dependent manner. Our study sheds light on the potential of PCA from commonly used anti-cancer Chinese herbal Medicine SJC as a lead compound targeting WT1 in the discovery of anti-HCC drugs.
Collapse
Affiliation(s)
- Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Hor-Yue Tan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Wei Guo
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Sha Li
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong S.A.R., China
| |
Collapse
|
1690
|
Bévant K, Coulouarn C. Landscape of genomic alterations in hepatocellular carcinoma: current knowledge and perspectives for targeted therapies. Hepatobiliary Surg Nutr 2017; 6:404-407. [PMID: 29312976 DOI: 10.21037/hbsn.2017.10.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Kévin Bévant
- Inserm, Inra, Univ Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Rennes, France
| | - Cédric Coulouarn
- Inserm, Inra, Univ Rennes, UMR 1241, Nutrition Metabolisms and Cancer, Rennes, France
| |
Collapse
|
1691
|
Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T, Chipashvili O, Hagan T, Walker M, Ramachandran A, Diosdado B, Serna G, Mulet N, Landolfi S, Ramon Y Cajal S, Fasani R, Aguirre AJ, Ng K, Élez E, Ogino S, Tabernero J, Fuchs CS, Hahn WC, Nuciforo P, Meyerson M. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017; 358:1443-1448. [PMID: 29170280 DOI: 10.1126/science.aal5240] [Citation(s) in RCA: 1029] [Impact Index Per Article: 128.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 08/17/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022]
Abstract
Colorectal cancers comprise a complex mixture of malignant cells, nontransformed cells, and microorganisms. Fusobacterium nucleatum is among the most prevalent bacterial species in colorectal cancer tissues. Here we show that colonization of human colorectal cancers with Fusobacterium and its associated microbiome-including Bacteroides, Selenomonas, and Prevotella species-is maintained in distal metastases, demonstrating microbiome stability between paired primary and metastatic tumors. In situ hybridization analysis revealed that Fusobacterium is predominantly associated with cancer cells in the metastatic lesions. Mouse xenografts of human primary colorectal adenocarcinomas were found to retain viable Fusobacterium and its associated microbiome through successive passages. Treatment of mice bearing a colon cancer xenograft with the antibiotic metronidazole reduced Fusobacterium load, cancer cell proliferation, and overall tumor growth. These observations argue for further investigation of antimicrobial interventions as a potential treatment for patients with Fusobacterium-associated colorectal cancer.
Collapse
Affiliation(s)
- Susan Bullman
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chandra S Pedamallu
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ewa Sicinska
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas E Clancy
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xiaoyang Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Diana Cai
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Donna Neuberg
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine Huang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Fatima Guevara
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy Nelson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Otari Chipashvili
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy Hagan
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Mark Walker
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aruna Ramachandran
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Begoña Diosdado
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Garazi Serna
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Nuria Mulet
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Stefania Landolfi
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Santiago Ramon Y Cajal
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Roberta Fasani
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Andrew J Aguirre
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kimmie Ng
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Élez
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Shuji Ogino
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Josep Tabernero
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Charles S Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - William C Hahn
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Nuciforo
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, CIBERONC, Universitat Autònoma de Barcelona, Spain
| | - Matthew Meyerson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
1692
|
Kielkopf CL. Insights from structures of cancer-relevant pre-mRNA splicing factors. Curr Opin Genet Dev 2017; 48:57-66. [PMID: 29128695 DOI: 10.1016/j.gde.2017.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022]
Abstract
Pre-mRNA splicing factors recognize consensus signals within preliminary transcripts, and as cogs of the spliceosome machine, orchestrate the excision and rejoining of pre-mRNA regions for gene expression. Large-scale sequencing has demonstrated that mutations in key genes encoding pre-mRNA splicing factors are common among myeloid neoplasms and also occur in a variety of other cancers. This revelation offers new therapeutic opportunities to target pre-mRNA splicing vulnerabilities in hematologic and other malignancies. The mutated residues typically alter 3' splice site choice for a subset of transcripts. In this review, we highlight mechanistic insights from recent 3D structures that reveal the affected residues poised for pre-mRNA recognition.
Collapse
Affiliation(s)
- Clara L Kielkopf
- Center for RNA Biology and Department of Biochemistry and Biophysics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
1693
|
Mutational signatures reveal the dynamic interplay of risk factors and cellular processes during liver tumorigenesis. Nat Commun 2017; 8:1315. [PMID: 29101368 PMCID: PMC5670220 DOI: 10.1038/s41467-017-01358-x] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/12/2017] [Indexed: 12/11/2022] Open
Abstract
Genomic alterations driving tumorigenesis result from the interaction of environmental exposures and endogenous cellular processes. With a diversity of risk factors, liver cancer is an ideal model to study these interactions. Here, we analyze the whole genomes of 44 new and 264 published liver cancers and we identify 10 mutational and 6 structural rearrangement signatures showing distinct relationships with environmental exposures, replication, transcription, and driver genes. The liver cancer-specific signature 16, associated with alcohol, displays a unique feature of transcription-coupled damage and is the main source of CTNNB1 mutations. Flood of insertions/deletions (indels) are identified in very highly expressed hepato-specific genes, likely resulting from replication-transcription collisions. Reconstruction of sub-clonal architecture reveals mutational signature evolution during tumor development exemplified by the vanishing of aflatoxin B1 signature in African migrants. Finally, chromosome duplications occur late and may represent rate-limiting events in tumorigenesis. These findings shed new light on the natural history of liver cancers. Tumorigenesis is a complex process driven by numerous risk factors. Here, genomic analysis of liver cancer reveals the evolution of mutational signatures during tumor development, highlighting mutational and structural signatures linked to environmental exposures and endogenous cellular processes.
Collapse
|
1694
|
Progress towards molecular patient stratification of hepatocellular carcinoma: Lost in translation? J Hepatol 2017; 67:893-895. [PMID: 28837834 DOI: 10.1016/j.jhep.2017.07.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 07/26/2017] [Accepted: 07/30/2017] [Indexed: 12/25/2022]
|
1695
|
Shu J, Silva BVRE, Gao T, Xu Z, Cui J. Dynamic and Modularized MicroRNA Regulation and Its Implication in Human Cancers. Sci Rep 2017; 7:13356. [PMID: 29042600 PMCID: PMC5645395 DOI: 10.1038/s41598-017-13470-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 09/26/2017] [Indexed: 12/19/2022] Open
Abstract
MicroRNA is responsible for the fine-tuning of fundamental cellular activities and human disease development. The altered availability of microRNAs, target mRNAs, and other types of endogenous RNAs competing for microRNA interactions reflects the dynamic and conditional property of microRNA-mediated gene regulation that remains under-investigated. Here we propose a new integrative method to study this dynamic process by considering both competing and cooperative mechanisms and identifying functional modules where different microRNAs co-regulate the same functional process. Specifically, a new pipeline was built based on a meta-Lasso regression model and the proof-of-concept study was performed using a large-scale genomic dataset from ~4,200 patients with 9 cancer types. In the analysis, 10,726 microRNA-mRNA interactions were identified to be associated with a specific stage and/or type of cancer, which demonstrated the dynamic and conditional miRNA regulation during cancer progression. On the other hands, we detected 4,134 regulatory modules that exhibit high fidelity of microRNA function through selective microRNA-mRNA binding and modulation. For example, miR-18a-3p, -320a, -193b-3p, and -92b-3p co-regulate the glycolysis/gluconeogenesis and focal adhesion in cancers of kidney, liver, lung, and uterus. Furthermore, several new insights into dynamic microRNA regulation in cancers have been discovered in this study.
Collapse
Affiliation(s)
- Jiang Shu
- Systems Biology and Biomedical Informatics (SBBI) Laboratory, Department of Computer Science and Engineering, Lincoln, NE, 68588, USA
| | - Bruno Vieira Resende E Silva
- Systems Biology and Biomedical Informatics (SBBI) Laboratory, Department of Computer Science and Engineering, Lincoln, NE, 68588, USA
| | - Tian Gao
- Systems Biology and Biomedical Informatics (SBBI) Laboratory, Department of Computer Science and Engineering, Lincoln, NE, 68588, USA
| | - Zheng Xu
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Quantitative Life Sciences Initiative, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Juan Cui
- Systems Biology and Biomedical Informatics (SBBI) Laboratory, Department of Computer Science and Engineering, Lincoln, NE, 68588, USA.
| |
Collapse
|
1696
|
Sourbier C. Plasma HSP90α and liver cancer: a potential biomarker? EBioMedicine 2017; 25:7-8. [PMID: 29037606 PMCID: PMC5704057 DOI: 10.1016/j.ebiom.2017.10.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
- Carole Sourbier
- Urologic Oncology Branch, National Cancer Institute, NIH, Bldg 10, CRC 1W-3961, 9000 Rockville Pike, Bethesda, MD 20892, United States.
| |
Collapse
|
1697
|
Sia D, Llovet JM. Liver cancer: Translating '-omics' results into precision medicine for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2017; 14:571-572. [PMID: 28765583 DOI: 10.1038/nrgastro.2017.103] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Daniela Sia
- Mount Sinai Liver Cancer Program, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York 10029, USA
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York 10029, USA; at the Liver Cancer Translational Research Laboratory, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic, Univesitat de Barcelona, Rosselló 153, Barcelona 08039, Spain; and at the Institució Catalana de Recerca i Estudis Avançats, Passeig de Lluís Companys 23, Barcelona 08010, Spain
| |
Collapse
|
1698
|
Castelli G, Pelosi E, Testa U. Liver Cancer: Molecular Characterization, Clonal Evolution and Cancer Stem Cells. Cancers (Basel) 2017; 9:cancers9090127. [PMID: 28930164 PMCID: PMC5615342 DOI: 10.3390/cancers9090127] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/15/2022] Open
Abstract
Liver cancer is the second most common cause of cancer-related death. The major forms of primary liver cancer are hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA). Both these tumors develop against a background of cirrhotic liver, non-alcoholic fatty liver disease, chronic liver damage and fibrosis. HCC is a heterogeneous disease which usually develops within liver cirrhosis related to various etiologies: hepatitis B virus (HBV) infection (frequent in Asia and Africa), hepatitis C virus (HCV), chronic alcohol abuse, or metabolic syndrome (frequent in Western countries). In cirrhosis, hepatocarcinogenesis is a multi-step process where pre-cancerous dysplastic macronodules transform progressively into HCC. The patterns of genomic alterations observed in these tumors were recently identified and were instrumental for the identification of potential targeted therapies that could improve patient care. Liver cancer stem cells are a small subset of undifferentiated liver tumor cells, responsible for cancer initiation, metastasis, relapse and chemoresistance, enriched and isolated according to immunophenotypic and functional properties: cell surface proteins (CD133, CD90, CD44, EpCAM, OV-6, CD13, CD24, DLK1, α2δ1, ICAM-1 and CD47); the functional markers corresponding to side population, high aldehyde dehydrogenase (ALDH) activity and autofluorescence. The identification and definition of liver cancer stem cells requires both immunophenotypic and functional properties.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00141, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00141, Italy.
| | - Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00141, Italy.
| |
Collapse
|
1699
|
Xue R, Li J, Bai F, Wang X, Ji J, Lu Y. A race to uncover a panoramic view of primary liver cancer. Cancer Biol Med 2017; 14:335-340. [PMID: 29372099 PMCID: PMC5785169 DOI: 10.20892/j.issn.2095-3941.2017.0112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Ruidong Xue
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China.,Translational Cancer Research Center, Peking University First Hospital, Beijing 100034, China.,Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Jing Li
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China.,Translational Cancer Research Center, Peking University First Hospital, Beijing 100034, China
| | - Fan Bai
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China
| | - Xinwei Wang
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China
| | - Junfang Ji
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China
| | - Yinying Lu
- Comprehensive Liver Cancer Center, Beijing 302 Hospital of PLA, Beijing 100039, China
| |
Collapse
|