151
|
Liu Y, Huan W, Wu J, Zou S, Qu L. IGFBP6 Is Downregulated in Unstable Carotid Atherosclerotic Plaques According to an Integrated Bioinformatics Analysis and Experimental Verification. J Atheroscler Thromb 2020; 27:1068-1085. [PMID: 32037372 PMCID: PMC7585910 DOI: 10.5551/jat.52993] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aims: To investigate the differentially expressed genes (DEGs) and molecular interaction in unstable atherosclerotic carotid plaques. Methods: Gene expression datasets GSE41571, GSE118481, and E-MTAB-2055 were analyzed. Co-regulated DEGs in at least two datasets were analyzed with the enrichment of Gene Ontology Biological Process (GO-BP), Kyoto Encyclopedia of Genes and Genomes (KEGG), protein-protein interaction (PPI) networks, interrelationships between miRNAs/transcriptional factors, and their target genes and drug-gene interactions. The expression of notable DEGs in human carotid artery plaques and plasma was further identified. Results: The GO-BP enrichment analysis revealed that genes associated with inflammatory response, and extracellular matrix organization were altered. The KEGG enrichment analysis revealed that upregulated DEGs were enriched in the tuberculous, lysosomal, and chemokine signaling pathways, whereas downregulated genes were enriched in the focal adhesion and PI3K/Akt signaling pathway. Collagen type I alpha 2 chain (COL1A2), adenylate cyclase 3 (ADCY3), C-X-C motif chemokine receptor 4 (CXCR4), and TYRO protein tyrosine kinase binding protein (TYROBP) might play crucial roles in the PPI networks. In drug–gene interactions, colony-stimulating factor-1 receptor had the most drug interactions. Insulin-like growth factor binding protein 6 (IGFBP6) was markedly downregulated in unstable human carotid plaques and plasma. Under a receiver operating characteristic curve analysis, plasma IGFBP6 had a significant discriminatory power (AUC, 0.894; 95% CI, 0.810–0.977), with a cutoff value of 142.08 ng/mL. Conclusions: The genes COL1A2, ADCY3, CXCR4, and TYROBP are promising targets for the prevention of unstable carotid plaque formation. IGFBP6 may be an important biomarker for predicting vulnerable plaques.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Second Military Medical University
| | - Wei Huan
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Second Military Medical University
| | - Jianjin Wu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Second Military Medical University
| | - Sili Zou
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Second Military Medical University
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital Affiliated to the Second Military Medical University
| |
Collapse
|
152
|
Bagherian M, Sabeti E, Wang K, Sartor MA, Nikolovska-Coleska Z, Najarian K. Machine learning approaches and databases for prediction of drug-target interaction: a survey paper. Brief Bioinform 2020; 22:247-269. [PMID: 31950972 PMCID: PMC7820849 DOI: 10.1093/bib/bbz157] [Citation(s) in RCA: 201] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/01/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
The task of predicting the interactions between drugs and targets plays a key role in the process of drug discovery. There is a need to develop novel and efficient prediction approaches in order to avoid costly and laborious yet not-always-deterministic experiments to determine drug–target interactions (DTIs) by experiments alone. These approaches should be capable of identifying the potential DTIs in a timely manner. In this article, we describe the data required for the task of DTI prediction followed by a comprehensive catalog consisting of machine learning methods and databases, which have been proposed and utilized to predict DTIs. The advantages and disadvantages of each set of methods are also briefly discussed. Lastly, the challenges one may face in prediction of DTI using machine learning approaches are highlighted and we conclude by shedding some lights on important future research directions.
Collapse
Affiliation(s)
- Maryam Bagherian
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Elyas Sabeti
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kai Wang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maureen A Sartor
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Kayvan Najarian
- Department of Electrical Engineering and Computer Science, College of Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
153
|
Kancherla J, Rao S, Bhuvaneshwar K, Riggins RB, Beckman RA, Madhavan S, Corrada Bravo H, Boca SM. Evidence-Based Network Approach to Recommending Targeted Cancer Therapies. JCO Clin Cancer Inform 2020; 4:71-88. [PMID: 31990579 PMCID: PMC6995264 DOI: 10.1200/cci.19.00097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2019] [Indexed: 12/30/2022] Open
Abstract
PURPOSE In this work, we introduce CDGnet (Cancer-Drug-Gene Network), an evidence-based network approach for recommending targeted cancer therapies. CDGnet represents a user-friendly informatics tool that expands the range of targeted therapy options for patients with cancer who undergo molecular profiling by including the biologic context via pathway information. METHODS CDGnet considers biologic pathway information specifically by looking at targets or biomarkers downstream of oncogenes and is personalized for individual patients via user-inputted molecular alterations and cancer type. It integrates a number of different sources of knowledge: patient-specific inputs (molecular alterations and cancer type), US Food and Drug Administration-approved therapies and biomarkers (curated from DailyMed), pathways for specific cancer types (from Kyoto Encyclopedia of Genes and Genomes [KEGG]), gene-drug connections (from DrugBank), and oncogene information (from KEGG). We consider 4 different evidence-based categories for therapy recommendations. Our tool is delivered via an R/Shiny Web application. For the 2 categories that use pathway information, we include an interactive Sankey visualization built on top of d3.js that also provides links to PubChem. RESULTS We present a scenario for a patient who has estrogen receptor (ER)-positive breast cancer with FGFR1 amplification. Although many therapies exist for patients with ER-positive breast cancer, FGFR1 amplifications may confer resistance to such treatments. CDGnet provides therapy recommendations, including PIK3CA, MAPK, and RAF inhibitors, by considering targets or biomarkers downstream of FGFR1. CONCLUSION CDGnet provides results in a number of easily accessible and usable forms, separating targeted cancer therapies into categories in an evidence-based manner that incorporates biologic pathway information.
Collapse
|
154
|
Cong R, Yang J, Zhou J, Shi J, Zhu Y, Zhu J, Xiao J, Wang P, He Y, He B. The Potential Role of Protein Tyrosine Phosphatase, Receptor Type C (CD45) in the Intestinal Ischemia-Reperfusion Injury. J Comput Biol 2019; 27:1303-1312. [PMID: 31855448 DOI: 10.1089/cmb.2019.0244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study was designed to identify several key genes and their functions in preventing or ameliorating intestinal ischemia-reperfusion (IR) injury, which could provide rationale for further exploring the regulatory mechanisms or clinical treatment for intestinal IR injury. The microarray GSE37013 of human intestinal IR injury was downloaded from Gene Expression Omnibus database. The differentially expressed genes (DEGs) with changes of reperfusion time were screened using Short Time-series Expression Miner, followed by function enrichment analysis, protein-protein interaction (PPI) network, and module construction. Subsequently, the key DEGs were identified with VEEN analysis based on the significant results of function enrichment analysis and PPI module. Finally, the gene-drug interactions were predicted using DGIdb 2.0. The DEGs of intestinal IR injury were significantly divided into three clusters with changes of reperfusion time. The genes in the three clusters were mainly enriched in transmembrane transport, defense responses, and cellular component assembly related pathways, respectively. There were 121 nodes and 281 interactions in PPI network, including one significant submodule. Protein tyrosine phosphatase, receptor type C (PTPRC) was a hub code both in PPI network and in submodule. A total of eight key DEGs were identified but only PTPRC was predicted to be interacted with eight drugs, such as infliximab. Totally, eight key genes associated with intestinal IR were identified; PTPRC especially was the most prominent potential drug target. These findings provided several potential therapeutic targets or potential breakthrough area in the study of intestinal IR injury.
Collapse
Affiliation(s)
- Ruochen Cong
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jushun Yang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jie Zhou
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jianhua Shi
- Department of Biochemistry, Nantong University Medical School, Nantong, Jiangsu, China
| | - Yihua Zhu
- Department of Clinical Laboratory, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jianfeng Zhu
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Jing Xiao
- Department of Epidemiology and Medical Statistics, School of Public Health, Nantong University, Nantong, Jiangsu, China
| | - Ping Wang
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| | - Ying He
- Department of Ultrasound, Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Bosheng He
- Department of Radiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China.,Clinical Medicine Research Center, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
155
|
Zhang Y, Shen B, Zhuge L, Xie Y. Identification of differentially expressed genes between the colon and ileum of patients with inflammatory bowel disease by gene co-expression analysis. J Int Med Res 2019; 48:300060519887268. [PMID: 31822145 PMCID: PMC7251957 DOI: 10.1177/0300060519887268] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE We aimed to identify differentially expressed genes (DEG) in patients with inflammatory bowel disease (IBD). METHODS RNA-seq data were obtained from the Array Express database. DEG were identified using the edgeR package. A co-expression network was constructed and key modules with the highest correlation with IBD inflammatory sites were identified for analysis. The Cytoscape MCODE plugin was used to identify key sub-modules of the protein-protein interaction (PPI) network. The genes in the sub-modules were considered hub genes, and functional enrichment analysis was performed. Furthermore, we constructed a drug-gene interaction network. Finally, we visualized the hub gene expression pattern between the colon and ileum of IBD using the ggpubr package and analyzed it using the Wilcoxon test. RESULTS DEG were identified between the colon and ileum of IBD patients. Based on the co-expression network, the green module had the highest correlation with IBD inflammatory sites. In total, 379 DEG in the green module were identified for the PPI network. Nineteen hub genes were differentially expressed between the colon and ileum. The drug-gene network identified these hub genes as potential drug targets. CONCLUSION Nineteen DEG were identified between the colon and ileum of IBD patients.
Collapse
Affiliation(s)
- Yuting Zhang
- Institute of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P. R. China.,Department of Liver Diseases, People's Hospital of Yichun City, Yichun, Jiangxi Province, P. R. China
| | - Bo Shen
- Department of Hepatobiliary Surgery, People's Hospital of Yichun City, Yichun, Jiangxi Province, P R China
| | - Liya Zhuge
- Institute of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P. R. China
| | - Yong Xie
- Institute of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P. R. China.,Department of Gastroenterology, the First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P R China
| |
Collapse
|
156
|
Danos AM, Krysiak K, Barnell EK, Coffman AC, McMichael JF, Kiwala S, Spies NC, Sheta LM, Pema SP, Kujan L, Clark KA, Wollam AZ, Rao S, Ritter DI, Sonkin D, Raca G, Lin WH, Grisdale CJ, Kim RH, Wagner AH, Madhavan S, Griffith M, Griffith OL. Standard operating procedure for curation and clinical interpretation of variants in cancer. Genome Med 2019; 11:76. [PMID: 31779674 PMCID: PMC6883603 DOI: 10.1186/s13073-019-0687-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 11/07/2019] [Indexed: 02/04/2023] Open
Abstract
Manually curated variant knowledgebases and their associated knowledge models are serving an increasingly important role in distributing and interpreting variants in cancer. These knowledgebases vary in their level of public accessibility, and the complexity of the models used to capture clinical knowledge. CIViC (Clinical Interpretation of Variants in Cancer - www.civicdb.org) is a fully open, free-to-use cancer variant interpretation knowledgebase that incorporates highly detailed curation of evidence obtained from peer-reviewed publications and meeting abstracts, and currently holds over 6300 Evidence Items for over 2300 variants derived from over 400 genes. CIViC has seen increased adoption by, and also undertaken collaboration with, a wide range of users and organizations involved in research. To enhance CIViC’s clinical value, regular submission to the ClinVar database and pursuit of other regulatory approvals is necessary. For this reason, a formal peer reviewed curation guideline and discussion of the underlying principles of curation is needed. We present here the CIViC knowledge model, standard operating procedures (SOP) for variant curation, and detailed examples to support community-driven curation of cancer variants.
Collapse
Affiliation(s)
- Arpad M Danos
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Kilannin Krysiak
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica K Barnell
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam C Coffman
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua F McMichael
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Susanna Kiwala
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas C Spies
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Lana M Sheta
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Shahil P Pema
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Lynzey Kujan
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Kaitlin A Clark
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Amber Z Wollam
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Shruti Rao
- Innovation Center for Biomedical Informatics, Georgetown University, Washington DC, USA
| | - Deborah I Ritter
- Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Dmitriy Sonkin
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - Gordana Raca
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Wan-Hsin Lin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida, USA
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Raymond H Kim
- Fred A. Litwin Family Center in Genetic Medicine, University Health Network, Toronto, ON, Canada
| | - Alex H Wagner
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Subha Madhavan
- Innovation Center for Biomedical Informatics, Georgetown University, Washington DC, USA.,Georgetown Lombardi Comprehensive Cancer Center, Washington DC, USA
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA. .,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Obi L Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA. .,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
157
|
Li L, Ng SR, Colón CI, Drapkin BJ, Hsu PP, Li Z, Nabel CS, Lewis CA, Romero R, Mercer KL, Bhutkar A, Phat S, Myers DT, Muzumdar MD, Westcott PMK, Beytagh MC, Farago AF, Vander Heiden MG, Dyson NJ, Jacks T. Identification of DHODH as a therapeutic target in small cell lung cancer. Sci Transl Med 2019; 11:eaaw7852. [PMID: 31694929 PMCID: PMC7401885 DOI: 10.1126/scitranslmed.aaw7852] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/18/2019] [Accepted: 09/27/2019] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive lung cancer subtype with extremely poor prognosis. No targetable genetic driver events have been identified, and the treatment landscape for this disease has remained nearly unchanged for over 30 years. Here, we have taken a CRISPR-based screening approach to identify genetic vulnerabilities in SCLC that may serve as potential therapeutic targets. We used a single-guide RNA (sgRNA) library targeting ~5000 genes deemed to encode "druggable" proteins to perform loss-of-function genetic screens in a panel of cell lines derived from autochthonous genetically engineered mouse models (GEMMs) of SCLC, lung adenocarcinoma (LUAD), and pancreatic ductal adenocarcinoma (PDAC). Cross-cancer analyses allowed us to identify SCLC-selective vulnerabilities. In particular, we observed enhanced sensitivity of SCLC cells toward disruption of the pyrimidine biosynthesis pathway. Pharmacological inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in this pathway, reduced the viability of SCLC cells in vitro and strongly suppressed SCLC tumor growth in human patient-derived xenograft (PDX) models and in an autochthonous mouse model. These results indicate that DHODH inhibition may be an approach to treat SCLC.
Collapse
Affiliation(s)
- Leanne Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sheng Rong Ng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Caterina I Colón
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Peggy P Hsu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Zhaoqi Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Nabel
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Rodrigo Romero
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kim L Mercer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjun Bhutkar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sarah Phat
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - David T Myers
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Mandar Deepak Muzumdar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter M K Westcott
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mary Clare Beytagh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anna F Farago
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Matthew G Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Nicholas J Dyson
- Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
158
|
Kreuzaler P, Clarke MA, Brown EJ, Wilson CH, Kortlever RM, Piterman N, Littlewood T, Evan GI, Fisher J. Heterogeneity of Myc expression in breast cancer exposes pharmacological vulnerabilities revealed through executable mechanistic modeling. Proc Natl Acad Sci U S A 2019; 116:22399-22408. [PMID: 31611367 PMCID: PMC6825310 DOI: 10.1073/pnas.1903485116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cells with higher levels of Myc proliferate more rapidly and supercompetitively eliminate neighboring cells. Nonetheless, tumor cells in aggressive breast cancers typically exhibit significant and stable heterogeneity in their Myc levels, which correlates with refractoriness to therapy and poor prognosis. This suggests that Myc heterogeneity confers some selective advantage on breast tumor growth and progression. To investigate this, we created a traceable MMTV-Wnt1-driven in vivo chimeric mammary tumor model comprising an admixture of low-Myc- and reversibly switchable high-Myc-expressing clones. We show that such tumors exhibit interclonal mutualism wherein cells with high-Myc expression facilitate tumor growth by promoting protumorigenic stroma yet concomitantly suppress Wnt expression, which renders them dependent for survival on paracrine Wnt provided by low-Myc-expressing clones. To identify any therapeutic vulnerabilities arising from such interdependency, we modeled Myc/Ras/p53/Wnt signaling cross talk as an executable network for low-Myc, for high-Myc clones, and for the 2 together. This executable mechanistic model replicated the observed interdependence of high-Myc and low-Myc clones and predicted a pharmacological vulnerability to coinhibition of COX2 and MEK. This was confirmed experimentally. Our study illustrates the power of executable models in elucidating mechanisms driving tumor heterogeneity and offers an innovative strategy for identifying combination therapies tailored to the oligoclonal landscape of heterogenous tumors.
Collapse
Affiliation(s)
- Peter Kreuzaler
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
- Oncogenes and Tumour Metabolism Lab, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Matthew A Clarke
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Elizabeth J Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Catherine H Wilson
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Roderik M Kortlever
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Nir Piterman
- Department of Computer Science and Engineering, University of Gothenburg, SE-41296 Gothenburg, Sweden
| | - Trevor Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom
| | - Gerard I Evan
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom;
| | - Jasmin Fisher
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, United Kingdom;
- UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| |
Collapse
|
159
|
Tolios A, De Las Rivas J, Hovig E, Trouillas P, Scorilas A, Mohr T. Computational approaches in cancer multidrug resistance research: Identification of potential biomarkers, drug targets and drug-target interactions. Drug Resist Updat 2019; 48:100662. [PMID: 31927437 DOI: 10.1016/j.drup.2019.100662] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 02/07/2023]
Abstract
Like physics in the 19th century, biology and molecular biology in particular, has been fertilized and enhanced like few other scientific fields, by the incorporation of mathematical methods. In the last decades, a whole new scientific field, bioinformatics, has developed with an output of over 30,000 papers a year (Pubmed search using the keyword "bioinformatics"). Huge databases of mass throughput data have been established, with ArrayExpress alone containing more than 2.7 million assays (October 2019). Computational methods have become indispensable tools in molecular biology, particularly in one of the most challenging areas of cancer research, multidrug resistance (MDR). However, confronted with a plethora of different algorithms, approaches, and methods, the average researcher faces key questions: Which methods do exist? Which methods can be used to tackle the aims of a given study? Or, more generally, how do I use computational biology/bioinformatics to bolster my research? The current review is aimed at providing guidance to existing methods with relevance to MDR research. In particular, we provide an overview on: a) the identification of potential biomarkers using expression data; b) the prediction of treatment response by machine learning methods; c) the employment of network approaches to identify gene/protein regulatory networks and potential key players; d) the identification of drug-target interactions; e) the use of bipartite networks to identify multidrug targets; f) the identification of cellular subpopulations with the MDR phenotype; and, finally, g) the use of molecular modeling methods to guide and enhance drug discovery. This review shall serve as a guide through some of the basic concepts useful in MDR research. It shall give the reader some ideas about the possibilities in MDR research by using computational tools, and, finally, it shall provide a short overview of relevant literature.
Collapse
Affiliation(s)
- A Tolios
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Institute of Clinical Chemistry and Laboratory Medicine, Heinrich Heine University, Duesseldorf, Germany.
| | - J De Las Rivas
- Bioinformatics and Functional Genomics Group, Cancer Research Center (CiC-IMBCC, CSIC/USAL/IBSAL), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Campus Miguel de Unamuno s/n, Salamanca, Spain.
| | - E Hovig
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital and Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway.
| | - P Trouillas
- UMR 1248 INSERM, Univ. Limoges, 2 rue du Dr Marland, 87052, Limoges, France; RCPTM, University Palacký of Olomouc, tr. 17. listopadu 12, 771 46, Olomouc, Czech Republic.
| | - A Scorilas
- Department of Biochemistry & Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| | - T Mohr
- Institute of Cancer Research, Department of Medicine I, Medical University of Vienna, Vienna, Austria; ScienceConsult - DI Thomas Mohr KG, Guntramsdorf, Austria.
| |
Collapse
|
160
|
Vaske OM, Bjork I, Salama SR, Beale H, Tayi Shah A, Sanders L, Pfeil J, Lam DL, Learned K, Durbin A, Kephart ET, Currie R, Newton Y, Swatloski T, McColl D, Vivian J, Zhu J, Lee AG, Leung SG, Spillinger A, Liu HY, Liang WS, Byron SA, Berens ME, Resnick AC, Lacayo N, Spunt SL, Rangaswami A, Huynh V, Torno L, Plant A, Kirov I, Zabokrtsky KB, Rassekh SR, Deyell RJ, Laskin J, Marra MA, Sender LS, Mueller S, Sweet-Cordero EA, Goldstein TC, Haussler D. Comparative Tumor RNA Sequencing Analysis for Difficult-to-Treat Pediatric and Young Adult Patients With Cancer. JAMA Netw Open 2019; 2:e1913968. [PMID: 31651965 PMCID: PMC6822083 DOI: 10.1001/jamanetworkopen.2019.13968] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
IMPORTANCE Pediatric cancers are epigenetic diseases; therefore, considering tumor gene expression information is necessary for a complete understanding of the tumorigenic processes. OBJECTIVE To evaluate the feasibility and utility of incorporating comparative gene expression information into the precision medicine framework for difficult-to-treat pediatric and young adult patients with cancer. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted as a consortium between the University of California, Santa Cruz (UCSC) Treehouse Childhood Cancer Initiative and clinical genomic trials. RNA sequencing (RNA-Seq) data were obtained from the following 4 clinical sites and analyzed at UCSC: British Columbia Children's Hospital (n = 31), Lucile Packard Children's Hospital at Stanford University (n = 80), CHOC Children's Hospital and Hyundai Cancer Institute (n = 46), and the Pacific Pediatric Neuro-Oncology Consortium (n = 24). The study dates were January 1, 2016, to March 22, 2017. EXPOSURES Participants underwent tumor RNA-Seq profiling as part of 4 separate clinical trials at partner hospitals. The UCSC either downloaded RNA-Seq data from a partner institution for analysis in the cloud or provided a Docker pipeline that performed the same analysis at a partner institution. The UCSC then compared each participant's tumor RNA-Seq profile with more than 11 000 uniformly analyzed tumor profiles from pediatric and young adult patients with cancer, downloaded from public data repositories. These comparisons were used to identify genes and pathways that are significantly overexpressed in each patient's tumor. Results of the UCSC analysis were presented to clinical partners. MAIN OUTCOMES AND MEASURES Feasibility of a third-party institution (UCSC Treehouse Childhood Cancer Initiative) to obtain tumor RNA-Seq data from patients, conduct comparative analysis, and present analysis results to clinicians; and proportion of patients for whom comparative tumor gene expression analysis provided useful clinical and biological information. RESULTS Among 144 samples from children and young adults (median age at diagnosis, 9 years; range, 0-26 years; 72 of 118 [61.0%] male [26 patients sex unknown]) with a relapsed, refractory, or rare cancer treated on precision medicine protocols, RNA-Seq-derived gene expression was potentially useful for 99 of 144 samples (68.8%) compared with DNA mutation information that was potentially useful for only 34 of 74 samples (45.9%). CONCLUSIONS AND RELEVANCE This study's findings suggest that tumor RNA-Seq comparisons may be feasible and highlight the potential clinical utility of incorporating such comparisons into the clinical genomic interpretation framework for difficult-to-treat pediatric and young adult patients with cancer. The study also highlights for the first time to date the potential clinical utility of harmonized publicly available genomic data sets.
Collapse
Affiliation(s)
- Olena M. Vaske
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Isabel Bjork
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Sofie R. Salama
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Howard Hughes Medical Institute, University of California, Santa Cruz
| | - Holly Beale
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Avanthi Tayi Shah
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Lauren Sanders
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Jacob Pfeil
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Du L. Lam
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Katrina Learned
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Ann Durbin
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Ellen T. Kephart
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Rob Currie
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Yulia Newton
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Teresa Swatloski
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Duncan McColl
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - John Vivian
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Jingchun Zhu
- University of California, Santa Cruz Genomics Institute, Santa Cruz
| | - Alex G. Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Stanley G. Leung
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Aviv Spillinger
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Heng-Yi Liu
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco
| | - Winnie S. Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - Sara A. Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | | | - Adam C. Resnick
- Center for Data Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Norman Lacayo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Sheri L. Spunt
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Arun Rangaswami
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Van Huynh
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Lilibeth Torno
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Ashley Plant
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Ivan Kirov
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | | | - S. Rod Rassekh
- British Columbia Children’s Hospital Research Institute, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Rebecca J. Deyell
- British Columbia Children’s Hospital Research Institute, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | | | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard S. Sender
- CHOC Children’s Hospital, Hyundai Cancer Institute, Orange, California
| | - Sabine Mueller
- Department of Neurology, University of California, San Francisco
- Department of Neurosurgery, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| | | | - Theodore C. Goldstein
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Now with Anthem, Inc, Palo Alto, California
| | - David Haussler
- University of California, Santa Cruz Genomics Institute, Santa Cruz
- Howard Hughes Medical Institute, University of California, Santa Cruz
| |
Collapse
|
161
|
Depuydt P, Boeva V, Hocking TD, Cannoodt R, Ambros IM, Ambros PF, Asgharzadeh S, Attiyeh EF, Combaret V, Defferrari R, Fischer M, Hero B, Hogarty MD, Irwin MS, Koster J, Kreissman S, Ladenstein R, Lapouble E, Laureys G, London WB, Mazzocco K, Nakagawara A, Noguera R, Ohira M, Park JR, Pötschger U, Theissen J, Tonini GP, Valteau-Couanet D, Varesio L, Versteeg R, Speleman F, Maris JM, Schleiermacher G, De Preter K. Genomic Amplifications and Distal 6q Loss: Novel Markers for Poor Survival in High-risk Neuroblastoma Patients. J Natl Cancer Inst 2019. [PMID: 29514301 PMCID: PMC6186524 DOI: 10.1093/jnci/djy022] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Neuroblastoma is characterized by substantial clinical heterogeneity. Despite intensive treatment, the survival rates of high-risk neuroblastoma patients are still disappointingly low. Somatic chromosomal copy number aberrations have been shown to be associated with patient outcome, particularly in low- and intermediate-risk neuroblastoma patients. To improve outcome prediction in high-risk neuroblastoma, we aimed to design a prognostic classification method based on copy number aberrations. Methods In an international collaboration, normalized high-resolution DNA copy number data (arrayCGH and SNP arrays) from 556 high-risk neuroblastomas obtained at diagnosis were collected from nine collaborative groups and segmented using the same method. We applied logistic and Cox proportional hazard regression to identify genomic aberrations associated with poor outcome. Results In this study, we identified two types of copy number aberrations that are associated with extremely poor outcome. Distal 6q losses were detected in 5.9% of patients and were associated with a 10-year survival probability of only 3.4% (95% confidence interval [CI] = 0.5% to 23.3%, two-sided P = .002). Amplifications of regions not encompassing the MYCN locus were detected in 18.1% of patients and were associated with a 10-year survival probability of only 5.8% (95% CI = 1.5% to 22.2%, two-sided P < .001). Conclusions Using a unique large copy number data set of high-risk neuroblastoma cases, we identified a small subset of high-risk neuroblastoma patients with extremely low survival probability that might be eligible for inclusion in clinical trials of new therapeutics. The amplicons may also nominate alternative treatments that target the amplified genes.
Collapse
Affiliation(s)
- Pauline Depuydt
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Valentina Boeva
- Institut Cochin, Inserm U1016, CNRS UMR 8104, Université Paris Descartes UMR-S1016, Paris, France.,Institut Curie, Inserm U900, Mines ParisTech, PSL Research University, Paris, France
| | - Toby D Hocking
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Robrecht Cannoodt
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Data Mining and Modelling for Biomedicine Group, VIB Center for Inflammation Research, Ghent, Belgium
| | - Inge M Ambros
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Peter F Ambros
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Shahab Asgharzadeh
- Division of Hematology/Oncology, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Edward F Attiyeh
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Childhood Cancer Research, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA
| | - Valérie Combaret
- Centre Léon-Bérard, Laboratoire de Recherche Translationnelle, Lyon, France
| | | | - Matthias Fischer
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany.,University Children's Hospital Cologne, Medical Faculty, and Center for Molecular Medicine Cologne
| | - Barbara Hero
- Department of Pediatric Oncology and Hematology, University of Cologne, Cologne, Germany
| | - Michael D Hogarty
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Perelman School of Medicine (MDH), University of Pennsylvania, Philadelphia, PA
| | - Meredith S Irwin
- Division of Hematology-Oncology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Susan Kreissman
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | - Ruth Ladenstein
- Children's Cancer Research Institute, Austria.,Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Eve Lapouble
- Genetic Somatic Unit.,Institut Curie, Paris, France
| | - Geneviève Laureys
- Department of Pediatric Hematology and Oncology, Ghent University Hospital, De Pintelaan, Ghent, Belgium
| | - Wendy B London
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Katia Mazzocco
- Department of Pathology, Istituto Giannina Gaslini, Genova, Italy
| | | | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia, Valencia, Spain.,Medical Research Foundation INCLIVA, Valencia, Spain.,CIBERONC, Madrid, Spain
| | - Miki Ohira
- Research Institute for Clinical Oncology Saitama Cancer Center, Saitama, Japan
| | - Julie R Park
- Seattle Children's Hospital and University of Washington, Seattle, WA
| | | | - Jessica Theissen
- Department of Experimental Pediatric Oncology, University of Cologne, Cologne, Germany
| | - Gian Paolo Tonini
- Laboratory of Neuroblastoma, Onco/Haematology Laboratory, University of Padua, Pediatric Research Institute (IRP)-Città della Speranza, Padova, Italy
| | | | - Luigi Varesio
- Laboratory of Molecular Biology (LV), Istituto Giannina Gaslini, Genova, Italy
| | - Rogier Versteeg
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Frank Speleman
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - John M Maris
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA.,Center for Childhood Cancer Research, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, University of Pennsylvania, Philadelphia, PA.,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA.,Abramson Family Cancer Research Institute, Philadelphia, PA
| | - Gudrun Schleiermacher
- U830 INSERM, Recherche Translationelle en Oncologie Pédiatrique (RTOP) and Department of Pediatric Oncology
| | - Katleen De Preter
- Center for Medical Genetics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
162
|
Talwar P, Gupta R, Kushwaha S, Agarwal R, Saso L, Kukreti S, Kukreti R. Viral Induced Oxidative and Inflammatory Response in Alzheimer's Disease Pathogenesis with Identification of Potential Drug Candidates: A Systematic Review using Systems Biology Approach. Curr Neuropharmacol 2019; 17:352-365. [PMID: 29676229 PMCID: PMC6482477 DOI: 10.2174/1570159x16666180419124508] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/19/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is genetically complex with multifactorial etiology. Here, we aim to identify the potential viral pathogens leading to aberrant inflammatory and oxidative stress response in AD along with potential drug candidates using systems biology approach. We retrieved protein interactions of amyloid precursor protein (APP) and tau protein (MAPT) from NCBI and genes for oxidative stress from NetAge, for inflammation from NetAge and InnateDB databases. Genes implicated in aging were retrieved from GenAge database and two GEO expression datasets. These genes were individually used to create protein-protein interaction network using STRING database (score≥0.7). The interactions of candidate genes with known viruses were mapped using virhostnet v2.0 database. Drug molecules targeting candidate genes were retrieved using the Drug- Gene Interaction Database (DGIdb). Data mining resulted in 2095 APP, 116 MAPT, 214 oxidative stress, 1269 inflammatory genes. After STRING PPIN analysis, 404 APP, 109 MAPT, 204 oxidative stress and 1014 inflammation related high confidence proteins were identified. The overlap among all datasets yielded eight common markers (AKT1, GSK3B, APP, APOE, EGFR, PIN1, CASP8 and SNCA). These genes showed association with hepatitis C virus (HCV), Epstein- Barr virus (EBV), human herpes virus 8 and Human papillomavirus (HPV). Further, screening of drugs targeting candidate genes, and possessing anti-inflammatory property, antiviral activity along with a suggested role in AD pathophysiology yielded 12 potential drug candidates. Our study demonstrated the role of viral etiology in AD pathogenesis by elucidating interaction of oxidative stress and inflammation causing candidate genes with common viruses along with the identification of potential AD drug candidates.
Collapse
Affiliation(s)
- Puneet Talwar
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi, India
| | - Renu Gupta
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Suman Kushwaha
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Rachna Agarwal
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | | | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi, India
| |
Collapse
|
163
|
Tacutu R, Thornton D, Johnson E, Budovsky A, Barardo D, Craig T, Diana E, Lehmann G, Toren D, Wang J, Fraifeld VE, de Magalhães JP. Human Ageing Genomic Resources: new and updated databases. Nucleic Acids Res 2019; 46:D1083-D1090. [PMID: 29121237 PMCID: PMC5753192 DOI: 10.1093/nar/gkx1042] [Citation(s) in RCA: 467] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 10/18/2017] [Indexed: 12/17/2022] Open
Abstract
In spite of a growing body of research and data, human ageing remains a poorly understood process. Over 10 years ago we developed the Human Ageing Genomic Resources (HAGR), a collection of databases and tools for studying the biology and genetics of ageing. Here, we present HAGR’s main functionalities, highlighting new additions and improvements. HAGR consists of six core databases: (i) the GenAge database of ageing-related genes, in turn composed of a dataset of >300 human ageing-related genes and a dataset with >2000 genes associated with ageing or longevity in model organisms; (ii) the AnAge database of animal ageing and longevity, featuring >4000 species; (iii) the GenDR database with >200 genes associated with the life-extending effects of dietary restriction; (iv) the LongevityMap database of human genetic association studies of longevity with >500 entries; (v) the DrugAge database with >400 ageing or longevity-associated drugs or compounds; (vi) the CellAge database with >200 genes associated with cell senescence. All our databases are manually curated by experts and regularly updated to ensure a high quality data. Cross-links across our databases and to external resources help researchers locate and integrate relevant information. HAGR is freely available online (http://genomics.senescence.info/).
Collapse
Affiliation(s)
- Robi Tacutu
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK.,Computational Biology of Aging Group, Institute of Biochemistry, Romanian Academy, Bucharest 060031, Romania
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Emily Johnson
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Arie Budovsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.,Judea Regional Research & Development Center, Carmel 90404, Israel
| | - Diogo Barardo
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City 117597, Singapore.,Science Division, Yale-NUS College, Singapore City 138527, Singapore
| | - Thomas Craig
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Eugene Diana
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Gilad Lehmann
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Dmitri Toren
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Jingwei Wang
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - João P de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
164
|
Nakken S, Fournous G, Vodák D, Aasheim LB, Myklebost O, Hovig E. Personal Cancer Genome Reporter: variant interpretation report for precision oncology. Bioinformatics 2019; 34:1778-1780. [PMID: 29272339 PMCID: PMC5946881 DOI: 10.1093/bioinformatics/btx817] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 12/19/2017] [Indexed: 11/30/2022] Open
Abstract
Summary Individual tumor genomes pose a major challenge for clinical interpretation due to their unique sets of acquired mutations. There is a general scarcity of tools that can (i) systematically interrogate cancer genomes in the context of diagnostic, prognostic, and therapeutic biomarkers, (ii) prioritize and highlight the most important findings and (iii) present the results in a format accessible to clinical experts. We have developed a stand-alone, open-source software package for somatic variant annotation that integrates a comprehensive set of knowledge resources related to tumor biology and therapeutic biomarkers, both at the gene and variant level. Our application generates a tiered report that will aid the interpretation of individual cancer genomes in a clinical setting. Availability and implementation The software is implemented in Python/R, and is freely available through Docker technology. Documentation, example reports, and installation instructions are accessible via the project GitHub page: https://github.com/sigven/pcgr. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Sigve Nakken
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Ghislain Fournous
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Daniel Vodák
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Lars Birger Aasheim
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway.,Bioinformatics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Ola Myklebost
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway.,Department of Clinical Science, University of Bergen, Norway
| | - Eivind Hovig
- Norwegian Cancer Genomics Consortium, Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Norway.,Department of Informatics, University of Oslo, Norway.,Institute for Cancer Genetics and Informatics, Oslo University Hospital, Norway
| |
Collapse
|
165
|
Li MJ, Yao H, Huang D, Liu H, Liu Z, Xu H, Qin Y, Prinz J, Xia W, Wang P, Yan B, Tran NL, Kocher JP, Sham PC, Wang J. mTCTScan: a comprehensive platform for annotation and prioritization of mutations affecting drug sensitivity in cancers. Nucleic Acids Res 2019; 45:W215-W221. [PMID: 28482068 PMCID: PMC5793836 DOI: 10.1093/nar/gkx400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/27/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer therapies have experienced rapid progress in recent years, with a number of novel small-molecule kinase inhibitors and monoclonal antibodies now being widely used to treat various types of human cancers. During cancer treatments, mutations can have important effects on drug sensitivity. However, the relationship between tumor genomic profiles and the effectiveness of cancer drugs remains elusive. We introduce Mutation To Cancer Therapy Scan (mTCTScan) web server (http://jjwanglab.org/mTCTScan) that can systematically analyze mutations affecting cancer drug sensitivity based on individual genomic profiles. The platform was developed by leveraging the latest knowledge on mutation-cancer drug sensitivity associations and the results from large-scale chemical screening using human cancer cell lines. Using an evidence-based scoring scheme based on current integrative evidences, mTCTScan is able to prioritize mutations according to their associations with cancer drugs and preclinical compounds. It can also show related drugs/compounds with sensitivity classification by considering the context of the entire genomic profile. In addition, mTCTScan incorporates comprehensive filtering functions and cancer-related annotations to better interpret mutation effects and their association with cancer drugs. This platform will greatly benefit both researchers and clinicians for interrogating mechanisms of mutation-dependent drug response, which will have a significant impact on cancer precision medicine.
Collapse
Affiliation(s)
- Mulin Jun Li
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.,Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Hongcheng Yao
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Dandan Huang
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Huanhuan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zipeng Liu
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Hang Xu
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Yiming Qin
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Jeanette Prinz
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Weiyi Xia
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Panwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Bin Yan
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.,School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Nhan L Tran
- Department of Cancer Biology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Jean-Pierre Kocher
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Pak C Sham
- Center for Genomic Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China.,Departments of Psychiatry, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Junwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA.,Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, USA
| |
Collapse
|
166
|
Canver MC, Bauer DE, Maeda T, Pinello L. DrugThatGene: integrative analysis to streamline the identification of druggable genes, pathways and protein complexes from CRISPR screens. Bioinformatics 2019; 35:1981-1984. [PMID: 30395160 PMCID: PMC6546128 DOI: 10.1093/bioinformatics/bty913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/23/2018] [Accepted: 10/31/2018] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) nuclease system has allowed for high-throughput, large scale pooled screens for functional genomic studies. To aid in the translation of functional genomics to therapeutics, we developed DrugThatGene (DTG) as a web-based application that streamlines analysis of potential therapeutic targets identified from functional genetic screens. RESULTS Starting from a gene list as input, DTG offers automated identification of small molecules along with supporting information from human genetic and other relevant databases. Furthermore, DTG aids in the identification of common biological pathways and protein complexes in conjunction with associated small molecule inhibitors. Taken together, DTG aims to expedite the identification of small molecules from the abundance of functional genetic data generated from CRISPR screens. AVAILABILITY AND IMPLEMENTATION DTG is an open-source and free software available as a website at http://drugthatgene.pinellolab.org. Source code is available at: https://github.com/pinellolab/DrugThatGene, which can be downloaded in order to run DTG locally.
Collapse
Affiliation(s)
- Matthew C Canver
- Molecular Pathology Unit, Center for Computational and Integrative Biology, Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Boston, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Takahiro Maeda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Luca Pinello
- Molecular Pathology Unit, Center for Computational and Integrative Biology, Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
167
|
Quan Y, Luo ZH, Yang QY, Li J, Zhu Q, Liu YM, Lv BM, Cui ZJ, Qin X, Xu YH, Zhu LD, Zhang HY. Systems Chemical Genetics-Based Drug Discovery: Prioritizing Agents Targeting Multiple/Reliable Disease-Associated Genes as Drug Candidates. Front Genet 2019; 10:474. [PMID: 31191604 PMCID: PMC6549477 DOI: 10.3389/fgene.2019.00474] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/01/2019] [Indexed: 01/10/2023] Open
Abstract
Genetic disease genes are considered a promising source of drug targets. Most diseases are caused by more than one pathogenic factor; thus, it is reasonable to consider that chemical agents targeting multiple disease genes are more likely to have desired activities. This is supported by a comprehensive analysis on the relationships between agent activity/druggability and target genetic characteristics. The therapeutic potential of agents increases steadily with increasing number of targeted disease genes, and can be further enhanced by strengthened genetic links between targets and diseases. By using the multi-label classification models for genetics-based drug activity prediction, we provide universal tools for prioritizing drug candidates. All of the documented data and the machine-learning prediction service are available at SCG-Drug (http://zhanglab.hzau.edu.cn/scgdrug).
Collapse
Affiliation(s)
- Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Zhi-Hui Luo
- College of Life Sciences and Technology, Huazhong Agricultural University, Wuhan, China
| | - Qing-Yong Yang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Jiang Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Qiang Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Ye-Mao Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Bo-Min Lv
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Ze-Jia Cui
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Xuan Qin
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Yan-Hua Xu
- Sci-meds Biopharmaceutical Co., Ltd., Wuhan, China
| | - Li-Da Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
168
|
Singer J, Irmisch A, Ruscheweyh HJ, Singer F, Toussaint NC, Levesque MP, Stekhoven DJ, Beerenwinkel N. Bioinformatics for precision oncology. Brief Bioinform 2019; 20:778-788. [PMID: 29272324 PMCID: PMC6585151 DOI: 10.1093/bib/bbx143] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Molecular profiling of tumor biopsies plays an increasingly important role not only in cancer research, but also in the clinical management of cancer patients. Multi-omics approaches hold the promise of improving diagnostics, prognostics and personalized treatment. To deliver on this promise of precision oncology, appropriate bioinformatics methods for managing, integrating and analyzing large and complex data are necessary. Here, we discuss the specific requirements of bioinformatics methods and software that arise in the setting of clinical oncology, owing to a stricter regulatory environment and the need for rapid, highly reproducible and robust procedures. We describe the workflow of a molecular tumor board and the specific bioinformatics support that it requires, from the primary analysis of raw molecular profiling data to the automatic generation of a clinical report and its delivery to decision-making clinical oncologists. Such workflows have to various degrees been implemented in many clinical trials, as well as in molecular tumor boards at specialized cancer centers and university hospitals worldwide. We review these and more recent efforts to include other high-dimensional multi-omics patient profiles into the tumor board, as well as the state of clinical decision support software to translate molecular findings into treatment recommendations.
Collapse
Affiliation(s)
- Jochen Singer
- Department of Biosystems Science and Engineering of ETH Zurich in Basel, Switzerland
| | - Anja Irmisch
- Department of Dermatology at the University of Zurich Hospital in Zurich, Switzerland
| | | | | | | | | | | | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering of ETH Zurich in Basel, Switzerland
| |
Collapse
|
169
|
Gao YC, Zhou XH, Zhang W. An Ensemble Strategy to Predict Prognosis in Ovarian Cancer Based on Gene Modules. Front Genet 2019; 10:366. [PMID: 31068972 PMCID: PMC6491874 DOI: 10.3389/fgene.2019.00366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Due to the high heterogeneity and complexity of cancer, it is still a challenge to predict the prognosis of cancer patients. In this work, we used a clustering algorithm to divide patients into different subtypes in order to reduce the heterogeneity of the cancer patients in each subtype. Based on the hypothesis that the gene co-expression network may reveal relationships among genes, some communities in the network could influence the prognosis of cancer patients and all the prognosis-related communities could fully reveal the prognosis of cancer patients. To predict the prognosis for cancer patients in each subtype, we adopted an ensemble classifier based on the gene co-expression network of the corresponding subtype. Using the gene expression data of ovarian cancer patients in TCGA (The Cancer Genome Atlas), three subtypes were identified. Survival analysis showed that patients in different subtypes had different survival risks. Three ensemble classifiers were constructed for each subtype. Leave-one-out and independent validation showed that our method outperformed control and literature methods. Furthermore, the function annotation of the communities in each subtype showed that some communities were cancer-related. Finally, we found that the current drug targets can partially support our method.
Collapse
Affiliation(s)
| | - Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Wen Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
170
|
Freire PP, Fernandez GJ, Cury SS, de Moraes D, Oliveira JS, de Oliveira G, Dal-Pai-Silva M, Dos Reis PP, Carvalho RF. The Pathway to Cancer Cachexia: MicroRNA-Regulated Networks in Muscle Wasting Based on Integrative Meta-Analysis. Int J Mol Sci 2019; 20:E1962. [PMID: 31013615 PMCID: PMC6515458 DOI: 10.3390/ijms20081962] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/05/2019] [Accepted: 04/11/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer cachexia is a multifactorial syndrome that leads to significant weight loss. Cachexia affects 50%-80% of cancer patients, depending on the tumor type, and is associated with 20%-40% of cancer patient deaths. Besides the efforts to identify molecular mechanisms of skeletal muscle atrophy-a key feature in cancer cachexia-no effective therapy for the syndrome is currently available. MicroRNAs are regulators of gene expression, with therapeutic potential in several muscle wasting disorders. We performed a meta-analysis of previously published gene expression data to reveal new potential microRNA-mRNA networks associated with muscle atrophy in cancer cachexia. We retrieved 52 differentially expressed genes in nine studies of muscle tissue from patients and rodent models of cancer cachexia. Next, we predicted microRNAs targeting these differentially expressed genes. We also include global microRNA expression data surveyed in atrophying skeletal muscles from previous studies as background information. We identified deregulated genes involved in the regulation of apoptosis, muscle hypertrophy, catabolism, and acute phase response. We further predicted new microRNA-mRNA interactions, such as miR-27a/Foxo1, miR-27a/Mef2c, miR-27b/Cxcl12, miR-27b/Mef2c, miR-140/Cxcl12, miR-199a/Cav1, and miR-199a/Junb, which may contribute to muscle wasting in cancer cachexia. Finally, we found drugs targeting MSTN, CXCL12, and CAMK2B, which may be considered for the development of novel therapeutic strategies for cancer cachexia. Our study has broadened the knowledge of microRNA-regulated networks that are likely associated with muscle atrophy in cancer cachexia, pointing to their involvement as potential targets for novel therapeutic strategies.
Collapse
Affiliation(s)
- Paula Paccielli Freire
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Geysson Javier Fernandez
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Sarah Santiloni Cury
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Diogo de Moraes
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Jakeline Santos Oliveira
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Grasieli de Oliveira
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Maeli Dal-Pai-Silva
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| | - Patrícia Pintor Dos Reis
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-687, Brazil.
- Experimental Research Unity, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-687, Brazil.
| | - Robson Francisco Carvalho
- Department of Morphology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo 18.618-619, Brazil.
| |
Collapse
|
171
|
Lodovichi S, Mercatanti A, Cervelli T, Galli A. Computational analysis of data from a genome-wide screening identifies new PARP1 functional interactors as potential therapeutic targets. Oncotarget 2019; 10:2722-2737. [PMID: 31105872 PMCID: PMC6505629 DOI: 10.18632/oncotarget.26812] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Knowledge of interaction network between different proteins can be a useful tool in cancer therapy. To develop new therapeutic treatments, understanding how these proteins contribute to dysregulated cellular pathways is an important task. PARP1 inhibitors are drugs used in cancer therapy, in particular where DNA repair is defective. It is crucial to find new candidate interactors of PARP1 as new therapeutic targets in order to increase efficacy of PARP1 inhibitors and expand their clinical utility. By a yeast-based genome wide screening, we previously discovered 90 candidate deletion genes that suppress growth-inhibition phenotype conferred by PARP1 in yeast. Here, we performed an integrated and computational analysis to deeply study these genes. First, we identified which pathways these genes are involved in and putative relations with PARP1 through g:Profiler. Then, we studied mutation pattern and their relation to cancer by interrogating COSMIC and DisGeNET database; finally, we evaluated expression and alteration in several cancers with cBioPortal, and the interaction network with GeneMANIA. We identified 12 genes belonging to PARP1-related pathways. We decided to further validate RIT1, INCENP and PSTA1 in MCF7 breast cancer cells. We found that RIT1 and INCENP affected PARylation and PARP1 protein level more significantly in PARP1 inhibited cells. Furthermore, downregulation of RIT1, INCENP and PSAT1 affected olaparib sensitivity of MCF7 cells. Our study identified candidate genes that could have an effect on PARP inhibition therapy. Moreover, we also confirm that yeast-based screenings could be very helpful to identify novel potential therapy factors.
Collapse
Affiliation(s)
- Samuele Lodovichi
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Pisa, Italy.,PhD Student in Clinical and Translational Science Program, University of Pisa, Pisa, Italy
| | - Alberto Mercatanti
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Pisa, Italy
| | - Tiziana Cervelli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Pisa, Italy
| | - Alvaro Galli
- Yeast Genetics and Genomics Group, Laboratory of Functional Genetics and Genomics, Institute of Clinical Physiology CNR, Pisa, Italy
| |
Collapse
|
172
|
Zhang G, Zhou S, Zhong W, Hong L, Wang Y, Lu S, Pan J, Huang Y, Su M, Crawford R, Zhou Y, Mai R. Whole-Exome Sequencing Reveals Frequent Mutations in Chromatin Remodeling Genes in Mammary and Extramammary Paget's Diseases. J Invest Dermatol 2019; 139:789-795. [PMID: 30905357 DOI: 10.1016/j.jid.2018.08.030] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 02/05/2023]
Abstract
Paget's disease (PD) is an intraepidermal adenocarcinoma of the skin at the breast (mammary PD) or urogenital locations (extramammary PD [EMPD]). At present, there is lack of clarity on PD's pathogenesis, the relationship between its subtypes, and its lineage link with the underlying invasive carcinomas. Here we describe that mammary PD and EMPD have similar mutational profiles, with the most frequent recurrent mutations occurring in the chromatin remodeling genes, such as KMT2C (MLL3, 39%) and ARID2 (22%), with additional recurrent somatic mutations detected in genes previously not known to be mutated in cancers, such as CDCC168 (34%), FSIP2 (29%), CASP8AP2 (29%), and BIRC6 (24%). In paired mammary PD and underlying breast carcinoma samples, distinct gene mutations were detected, indicating that they represent independent oncogenic events. Finally, multistage EMPD tissue sequencing revealed KMT2C gene occurring early in EMPD oncogenesis, and that multifocal EMPD samples share the same early gene mutations, suggesting clonal origin of multifocal EMPD. Our results reveal similar genomic landscapes between mammary PD and EMPD, including early aberrations in chromatin remodeling genes. In addition, mammary PD and underlying breast ductal carcinomas represent independent oncogenic events. These findings provide approaches for developing diagnostic tools and therapeutic interventions for PD.
Collapse
Affiliation(s)
- Guohong Zhang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Songxia Zhou
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Weixiang Zhong
- Department of Pathology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liangli Hong
- Department of Pathology, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuanyuan Wang
- Department of Pathology, Shantou Central Hospital of and the Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, Guangdong, China
| | - Shanming Lu
- Department of Pathology, Meizhou Central Hospital, Meizhou, Guangdong, China
| | - Jiankai Pan
- Department of Pathology, Shantou Hospital of Dermatology, Shantou, Guangdong, China
| | - Yuansheng Huang
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mingwan Su
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard Crawford
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Youwen Zhou
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Ruiqin Mai
- Department of Laboratory Medicine, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
173
|
Gaspar HA, Gerring Z, Hübel C, Middeldorp CM, Derks EM, Breen G. Using genetic drug-target networks to develop new drug hypotheses for major depressive disorder. Transl Psychiatry 2019; 9:117. [PMID: 30877270 PMCID: PMC6420656 DOI: 10.1038/s41398-019-0451-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/28/2019] [Accepted: 02/12/2019] [Indexed: 12/25/2022] Open
Abstract
The major depressive disorder (MDD) working group of the Psychiatric Genomics Consortium (PGC) has published a genome-wide association study (GWAS) for MDD in 130,664 cases, identifying 44 risk variants. We used these results to investigate potential drug targets and repurposing opportunities. We built easily interpretable bipartite drug-target networks integrating interactions between drugs and their targets, genome-wide association statistics, and genetically predicted expression levels in different tissues, using the online tool Drug Targetor ( drugtargetor.com ). We also investigated drug-target relationships that could be impacting MDD. MAGMA was used to perform pathway analyses and S-PrediXcan to investigate the directionality of tissue-specific expression levels in patients vs. controls. Outside the major histocompatibility complex (MHC) region, 153 protein-coding genes are significantly associated with MDD in MAGMA after multiple testing correction; among these, five are predicted to be down or upregulated in brain regions and 24 are known druggable genes. Several drug classes were significantly enriched, including monoamine reuptake inhibitors, sex hormones, antipsychotics, and antihistamines, indicating an effect on MDD and potential repurposing opportunities. These findings not only require validation in model systems and clinical examination, but also show that GWAS may become a rich source of new therapeutic hypotheses for MDD and other psychiatric disorders that need new-and better-treatment options.
Collapse
Affiliation(s)
- Héléna A Gaspar
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK.
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK.
| | - Zachary Gerring
- Translational Neurogenomics Laboratory, QIMR Berghofer Institute of Medical Research, Brisbane City, QLD 4006, Australia
| | - Christopher Hübel
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christel M Middeldorp
- Child Health Research Centre, University of Queensland, South Brisbane, QLD 4072, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, South Brisbane, QLD 4101, Australia
- Biological Psychology, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, Netherlands
| | - Eske M Derks
- Translational Neurogenomics Laboratory, QIMR Berghofer Institute of Medical Research, Brisbane City, QLD 4006, Australia
| | - Gerome Breen
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK
| |
Collapse
|
174
|
Sakharov D, Maltseva D, Knyazev E, Nikulin S, Poloznikov A, Shilin S, Baranova A, Tsypina I, Tonevitsky A. Towards embedding Caco-2 model of gut interface in a microfluidic device to enable multi-organ models for systems biology. BMC SYSTEMS BIOLOGY 2019; 13:19. [PMID: 30836980 PMCID: PMC6399809 DOI: 10.1186/s12918-019-0686-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background A cancer cell line originating from human epithelial colorectal adenocarcinoma (Caco-2 cells) serves as a high capacity model for a preclinical screening of drugs. Recent need for incorporating barrier tissue into multi-organ chips calls for inclusion of Caco-2 cells into microperfused environment. Results This article describes a series of systems biology insights obtained from comparing Caco-2 models cells grown as conventional 2D layer and in a microfluidic chip. When basic electrical parameters of Caco-2 monolayers were evaluated using impedance spectrometry and MTT assays, no differences were noted. On the other hand, the microarray profiling of mRNAs and miRNAs revealed that grows on a microfluidic chip leads to the change in the production of specific miRNA, which regulate a set of genes for cell adhesion molecules (CAMs), and provide for more complete differentiation of Caco-2 monolayer. Moreover, the sets of miRNAs secreted at the apical surface of Caco-2 monolayers grown in conventional 2D culture and in microfluidic device differ. Conclusions When integrated into a multi-tissue platform, Caco-2 cells may aid in generating insights into complex pathophysiological processes, not possible to dissect in conventional cultures.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax VA, USA.,Research Center of Medical Genetics, Moscow, Russia
| | - Irina Tsypina
- SRC BioClinicum, Moscow, Russia.,Department of Cell Biology, Higher School of Economics, Moscow, Russia
| | - Alexander Tonevitsky
- SRC BioClinicum, Moscow, Russia.,Department of Cell Biology, Higher School of Economics, Moscow, Russia.,Art photonics GmbH, Berlin, Germany
| |
Collapse
|
175
|
CSgator: an integrated web platform for compound set analysis. J Cheminform 2019; 11:17. [PMID: 30830479 PMCID: PMC6419788 DOI: 10.1186/s13321-019-0339-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/26/2019] [Indexed: 12/13/2022] Open
Abstract
Drug discovery typically involves investigation of a set of compounds (e.g. drug screening hits) in terms of target, disease, and bioactivity. CSgator is a comprehensive analytic tool for set-wise interpretation of compounds. It has two unique analytic features of Compound Set Enrichment Analysis (CSEA) and Compound Cluster Analysis (CCA), which allows batch analysis of compound set in terms of (i) target, (ii) bioactivity, (iii) disease, and (iv) structure. CSEA and CCA present enriched profiles of targets and bioactivities in a compound set, which leads to novel insights on underlying drug mode-of-action, and potential targets. Notably, we propose a novel concept of 'Hit Enriched Assays", i.e. bioassays of which hits are enriched among a given set of compounds. As an example, we show its utility in revealing drug mode-of-action or identifying hidden targets for anti-lymphangiogenesis screening hits. CSgator is available at http://csgator.ewha.ac.kr , and most analytic results are downloadable.
Collapse
|
176
|
Cava C, Castiglioni I. In silico perturbation of drug targets in pan-cancer analysis combining multiple networks and pathways. Gene 2019; 698:100-106. [PMID: 30840853 DOI: 10.1016/j.gene.2019.02.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/13/2019] [Accepted: 02/23/2019] [Indexed: 12/13/2022]
Abstract
The knowledge of cancer cell response to conventional therapies is crucial in order to choose the correct therapy of patients affected by cancer. The major problem is generally attributed to the lack of specific biological processes able to predict the therapy efficacy. Here, we optimized a computational method for the analysis of gene networks able to detect and quantify the effects of a drug in a pan-cancer study. Overall, our method, using several network topological measures has identified a cancer gene network with a key role in biological processes. The gene network, able to classify with a good performance cancer vs normal samples, was modulated in silico to evaluate the effects of new or approved drugs. This computational model could offer an interesting hint to decipher molecular mechanisms contributing to resistance or inefficacy of drugs.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Segrate, Milan, Italy.
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Segrate, Milan, Italy.
| |
Collapse
|
177
|
Ray P, Torck A, Quigley L, Wangzhou A, Neiman M, Rao C, Lam T, Kim JY, Kim TH, Zhang MQ, Dussor G, Price TJ. Comparative transcriptome profiling of the human and mouse dorsal root ganglia: an RNA-seq-based resource for pain and sensory neuroscience research. Pain 2019; 159:1325-1345. [PMID: 29561359 DOI: 10.1097/j.pain.0000000000001217] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Molecular neurobiological insight into human nervous tissues is needed to generate next-generation therapeutics for neurological disorders such as chronic pain. We obtained human dorsal root ganglia (hDRG) samples from organ donors and performed RNA-sequencing (RNA-seq) to study the hDRG transcriptional landscape, systematically comparing it with publicly available data from a variety of human and orthologous mouse tissues, including mouse DRG (mDRG). We characterized the hDRG transcriptional profile in terms of tissue-restricted gene coexpression patterns and putative transcriptional regulators, and formulated an information-theoretic framework to quantify DRG enrichment. Relevant gene families and pathways were also analyzed, including transcription factors, G-protein-coupled receptors, and ion channels. Our analyses reveal an hDRG-enriched protein-coding gene set (∼140), some of which have not been described in the context of DRG or pain signaling. Most of these show conserved enrichment in mDRG and were mined for known drug-gene product interactions. Conserved enrichment of the vast majority of transcription factors suggests that the mDRG is a faithful model system for studying hDRG, because of evolutionarily conserved regulatory programs. Comparison of hDRG and tibial nerve transcriptomes suggests trafficking of neuronal mRNA to axons in adult hDRG, and are consistent with studies of axonal transport in rodent sensory neurons. We present our work as an online, searchable repository (https://www.utdallas.edu/bbs/painneurosciencelab/sensoryomics/drgtxome), creating a valuable resource for the community. Our analyses provide insight into DRG biology for guiding development of novel therapeutics and a blueprint for cross-species transcriptomic analyses.
Collapse
Affiliation(s)
- Pradipta Ray
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA.,Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Andrew Torck
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Lilyana Quigley
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Andi Wangzhou
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Matthew Neiman
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Chandranshu Rao
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Tiffany Lam
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Ji-Young Kim
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Tae Hoon Kim
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Michael Q Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
178
|
Yu LH, Huang QW, Zhou XH. Identification of Cancer Hallmarks Based on the Gene Co-expression Networks of Seven Cancers. Front Genet 2019; 10:99. [PMID: 30838028 PMCID: PMC6389798 DOI: 10.3389/fgene.2019.00099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/29/2019] [Indexed: 12/20/2022] Open
Abstract
Identifying the hallmarks of cancer is essential for cancer research, and the genes involved in cancer hallmarks are likely to be cancer drivers. However, there is no appropriate method in the current literature for identifying genetic cancer hallmarks, especially considering the interrelationships among the genes. Here, we hypothesized that "dense clusters" (or "communities") in the gene co-expression networks of cancer patients may represent functional units regarding cancer formation and progression, and the communities present in the co-expression networks of multiple types of cancer may be cancer hallmarks. Consequently, we mined the conserved communities in the gene co-expression networks of seven cancers in order to identify candidate hallmarks. Functional annotation of the communities showed that they were mainly related to immune response, the cell cycle and the biological processes that maintain basic cellular functions. Survival analysis using the genes involved in the conserved communities verified that two of these hallmarks could predict the survival risks of cancer patients in multiple types of cancer. Furthermore, the genes involved in these hallmarks, one of which was related to the cell cycle, could be useful in screening for cancer drugs.
Collapse
Affiliation(s)
- Ling-Hao Yu
- College of Science, Huazhong Agricultural University, Wuhan, China
| | - Qin-Wei Huang
- College of Science, Huazhong Agricultural University, Wuhan, China
| | - Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
179
|
Zhou XH, Chu XY, Xue G, Xiong JH, Zhang HY. Identifying cancer prognostic modules by module network analysis. BMC Bioinformatics 2019; 20:85. [PMID: 30777030 PMCID: PMC6380061 DOI: 10.1186/s12859-019-2674-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/08/2019] [Indexed: 02/08/2023] Open
Abstract
Background The identification of prognostic genes that can distinguish the prognostic risks of cancer patients remains a significant challenge. Previous works have proven that functional gene sets were more reliable for this task than the gene signature. However, few works have considered the cross-talk among functional gene sets, which may result in neglecting important prognostic gene sets for cancer. Results Here, we proposed a new method that considers both the interactions among modules and the prognostic correlation of the modules to identify prognostic modules in cancers. First, dense sub-networks in the gene co-expression network of cancer patients were detected. Second, cross-talk between every two modules was identified by a permutation test, thus generating the module network. Third, the prognostic correlation of each module was evaluated by the resampling method. Then, the GeneRank algorithm, which takes the module network and the prognostic correlations of all the modules as input, was applied to prioritize the prognostic modules. Finally, the selected modules were validated by survival analysis in various data sets. Our method was applied in three kinds of cancers, and the results show that our method succeeded in identifying prognostic modules in all the three cancers. In addition, our method outperformed state-of-the-art methods. Furthermore, the selected modules were significantly enriched with known cancer-related genes and drug targets of cancer, which may indicate that the genes involved in the modules may be drug targets for therapy. Conclusions We proposed a useful method to identify key modules in cancer prognosis and our prognostic genes may be good candidates for drug targets. Electronic supplementary material The online version of this article (10.1186/s12859-019-2674-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xin-Yi Chu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gang Xue
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiang-Hui Xiong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, People's Republic of China.,Lab of Epigenetics and Health Tracking Technology, Space Institute of Southern China, Shenzhen, People's Republic of China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
180
|
PheWAS-Based Systems Genetics Methods for Anti-Breast Cancer Drug Discovery. Genes (Basel) 2019; 10:genes10020154. [PMID: 30781719 PMCID: PMC6409623 DOI: 10.3390/genes10020154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/16/2019] [Accepted: 02/04/2019] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is a high-risk disease worldwide. For such complex diseases that are induced by multiple pathogenic genes, determining how to establish an effective drug discovery strategy is a challenge. In recent years, a large amount of genetic data has accumulated, particularly in the genome-wide identification of disorder genes. However, understanding how to use these data efficiently for pathogenesis elucidation and drug discovery is still a problem because the gene–disease links that are identified by high-throughput techniques such as phenome-wide association studies (PheWASs) are usually too weak to have biological significance. Systems genetics is a thriving area of study that aims to understand genetic interactions on a genome-wide scale. In this study, we aimed to establish two effective strategies for identifying breast cancer genes based on the systems genetics algorithm. As a result, we found that the GeneRank-based strategy, which combines the prognostic phenotype-based gene-dependent network with the phenotypic-related PheWAS data, can promote the identification of breast cancer genes and the discovery of anti-breast cancer drugs.
Collapse
|
181
|
Smith NG, Gyanchandani R, Shah OS, Gurda GT, Lucas PC, Hartmaier RJ, Brufsky AM, Puhalla S, Bahreini A, Kota K, Wald AI, Nikiforov YE, Nikiforova MN, Oesterreich S, Lee AV. Targeted mutation detection in breast cancer using MammaSeq™. Breast Cancer Res 2019; 21:22. [PMID: 30736836 PMCID: PMC6368740 DOI: 10.1186/s13058-019-1102-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 01/16/2019] [Indexed: 01/14/2023] Open
Abstract
Background Breast cancer is the most common invasive cancer among women worldwide. Next-generation sequencing (NGS) has revolutionized the study of cancer across research labs around the globe; however, genomic testing in clinical settings remains limited. Advances in sequencing reliability, pipeline analysis, accumulation of relevant data, and the reduction of costs are rapidly increasing the feasibility of NGS-based clinical decision making. Methods We report the development of MammaSeq, a breast cancer-specific NGS panel, targeting 79 genes and 1369 mutations, optimized for use in primary and metastatic breast cancer. To validate the panel, 46 solid tumors and 14 plasma circulating tumor DNA (ctDNA) samples were sequenced to a mean depth of 2311× and 1820×, respectively. Variants were called using Ion Torrent Suite 4.0 and annotated with cravat CHASM. CNVKit was used to call copy number variants in the solid tumor cohort. The oncoKB Precision Oncology Database was used to identify clinically actionable variants. Droplet digital PCR was used to validate select ctDNA mutations. Results In cohorts of 46 solid tumors and 14 ctDNA samples from patients with advanced breast cancer, we identified 592 and 43 protein-coding mutations. Mutations per sample in the solid tumor cohort ranged from 1 to 128 (median 3), and the ctDNA cohort ranged from 0 to 26 (median 2.5). Copy number analysis in the solid tumor cohort identified 46 amplifications and 35 deletions. We identified 26 clinically actionable variants (levels 1–3) annotated by OncoKB, distributed across 20 out of 46 cases (40%), in the solid tumor cohort. Allele frequencies of ESR1 and FOXA1 mutations correlated with CA.27.29 levels in patient-matched blood draws. Conclusions In solid tumor biopsies and ctDNA, MammaSeq detects clinically actionable mutations (OncoKB levels 1–3) in 22/46 (48%) solid tumors and in 4/14 (29%) of ctDNA samples. MammaSeq is a targeted panel suitable for clinically actionable mutation detection in breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1102-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nicholas G Smith
- Department of Pharmacology and Chemical Biology, and Human Genetics, UPMC Hillman Cancer Center, Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Rekha Gyanchandani
- Department of Pharmacology and Chemical Biology, and Human Genetics, UPMC Hillman Cancer Center, Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Osama S Shah
- Graduate Program in Integrated Systems Biology, University of Pittsburgh, Pittsburgh, USA
| | - Grzegorz T Gurda
- Department of Pathology, Gundersen Health System, La Crosse, WI, USA
| | - Peter C Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan J Hartmaier
- Department of Pharmacology and Chemical Biology, and Human Genetics, UPMC Hillman Cancer Center, Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Adam M Brufsky
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shannon Puhalla
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Amir Bahreini
- Department of Genetics and Molecular Biology, School of Medicine, University of Medical Sciences, Isfahan, Iran
| | - Karthik Kota
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abigail I Wald
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuri E Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, and Human Genetics, UPMC Hillman Cancer Center, Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA
| | - Adrian V Lee
- Department of Pharmacology and Chemical Biology, and Human Genetics, UPMC Hillman Cancer Center, Magee-Womens Research Institute, University of Pittsburgh, 204 Craft Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
182
|
Mahlokozera T, Vellimana AK, Li T, Mao DD, Zohny ZS, Kim DH, Tran DD, Marcus DS, Fouke SJ, Campian JL, Dunn GP, Miller CA, Kim AH. Biological and therapeutic implications of multisector sequencing in newly diagnosed glioblastoma. Neuro Oncol 2019; 20:472-483. [PMID: 29244145 DOI: 10.1093/neuonc/nox232] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Diagnostic workflows for glioblastoma (GBM) patients increasingly include DNA sequencing-based analysis of a single tumor site following biopsy or resection. We hypothesized that sequencing of multiple sectors within a given tumor would provide a more comprehensive representation of the molecular landscape and potentially inform therapeutic strategies. Methods Ten newly diagnosed, isocitrate dehydrogenase 1 (IDH1) wildtype GBM tumor samples were obtained from 2 (n = 9) or 4 (n = 1) spatially distinct tumor regions. Tumor and matched blood DNA samples underwent whole-exome sequencing. Results Across all 10 tumors, 51% of mutations were clonal and 3% were subclonal and shared in different sectors, whereas 46% of mutations were subclonal and private. Two of the 10 tumors exhibited a regional hypermutator state despite being treatment naïve, and remarkably, the high mutational load was predominantly limited to one sector in each tumor. Among the canonical cancer-associated genes, only telomerase reverse transcriptase (TERT) promoter mutations were observed in the founding clone in all tumors. Reconstruction of the clonal architecture in different sectors revealed regionally divergent evolution, and integration of data from 2 sectors increased the resolution of inferred clonal architecture in a given tumor. Predicted therapeutic mutations differed in presence and frequency between tumor regions. Similarly, different sectors exhibited significant divergence in the predicted neoantigen landscape. Conclusions The substantial spatial heterogeneity observed in different GBM tumor sectors, especially in spatially restricted hypermutator cases, raises important caveats to our current dependence on single-sector molecular information to guide either targeted or immune-based treatments.
Collapse
Affiliation(s)
- Tatenda Mahlokozera
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Ananth K Vellimana
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Tiandao Li
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri
| | - Diane D Mao
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Zohny S Zohny
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri
| | - David H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri
| | - David D Tran
- Lillian S. Wells Department of Neurosurgery, University of Florida College of Medicine, Gainesville, Florida
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Sarah J Fouke
- Department of Neurosurgery, St Luke's Hospital, St Louis, Missouri
| | - Jian L Campian
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri.,Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Christopher A Miller
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St Louis, Missouri.,Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri.,Department of Neurology, Washington University School of Medicine, St Louis, Missouri.,Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
183
|
Gershkovich P, Platt J, Knopf J, Tasoulis MK, Shi W, Pusztai L, Hatzis C. TQuest, A Web-Based Platform to Enable Precision Medicine by Linking a Tumor's Genetic Defects to Therapeutic Options. JCO Clin Cancer Inform 2019; 2:1-13. [PMID: 30652574 DOI: 10.1200/cci.17.00120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Currently, there are only a few software tools designed to assist physicians to translate molecular abnormalities in the cancer genome into potential treatment options. There is a pressing need to develop software to reliably identify known targeted therapies and experimental treatments for patients on the basis of the results of tumor DNA sequencing. METHODS The TQuest platform includes a data layer, data acquisition layer, search engine, and user interface. It identifies associations between one or more molecular targets and therapeutic options. The data layer consists of indexed interventional clinical trials and an expert-curated database of clinically or experimentally validated associations between mutations and drug response. The data acquisition layer includes an information-harvesting module that keeps an up-to-date full-text index of clinical trials by crawling clinicaltrials.gov and combining it with US Food and Drug Administration label data. The user interface is a Web-based module that allows users to upload genomic variants, tumor morphology, and diagnosis. The search results are qualified and ranked by a relevance score. RESULTS We have manually curated information for 368 distinct genomic variants of 162 gene targets corresponding to 863 drug and target interactions. The platform currently contains a full-text index of approximately 80,000 interventional clinical trials. We applied TQuest to molecular data from 73 metastatic breast cancers. TQuest identified a total of 276 drugs as potential therapeutic options, ranging from one to 103 per patient. CONCLUSION TQuest correctly identified all US Food and Drug Administration-approved drugs and routine indications for all cases and also identified many additional drugs that were used in the context of a given molecular abnormality in various clinical trials. The prototype Web application is available at www.tquest.us , and the source code is open and available on GitHub.
Collapse
Affiliation(s)
- Peter Gershkovich
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - James Platt
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - Joshua Knopf
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - Marios K Tasoulis
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - Weiwei Shi
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - Lajos Pusztai
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| | - Christos Hatzis
- Peter Gershkovich, Joshua Knopf, Lajos Pusztai, Christos Hatzis, Yale School of Medicine, New Haven; James Platt, Biomantica, West Haven, CT; Marios K. Tasoulis, The Royal Marsden National Health Services Foundation Trust, London, United Kingdom; and Weiwei Shi, Origimed, Shanghai, China
| |
Collapse
|
184
|
Demeure MJ. The Role of Precision Medicine in the Diagnosis and Treatment of Patients with Rare Cancers. Cancer Treat Res 2019; 178:81-108. [PMID: 31209842 DOI: 10.1007/978-3-030-16391-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Rare cancers pose unique challenges for patients and their physicians arising from a lack of information regarding the best therapeutic options. Very often, a lack of clinical trial data leads physicians to choose treatments based on small case series or case reports. Precision medicine based on genomic analysis of tumors may allow for selection of better treatments with greater efficacy and less toxicity. Physicians are increasingly using genetics to identify patients at high risk for certain cancers to allow for early detection or prophylactic interventions. Genomics can be used to inform prognosis and more accurately establish a diagnosis. Genomic analysis may also expose therapeutic targets for which drugs are currently available and approved for use in other cancers. Notable successes in the treatment of previously refractory cancers have resulted. New more advanced sequencing technologies, tools for interpretation, and an increasing array of targeted drugs offer additional hope, but challenges remain.
Collapse
Affiliation(s)
- Michael J Demeure
- Hoag Family Cancer Institute, Newport Beach, CA, USA.
- Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
185
|
Abstract
Tumor genomic profiling involves analyzing many data types to produce a molecular profile of a tumor. Many of these analyses result in a prioritized list of genes or variants for further study. Interpretation of these lists relies upon annotating and extracting biological meaning through literature and manually curated knowledge bases. This chapter will describe several of these approaches including gene annotation, variant annotation, clinical annotation, functional enrichment analyses, and network analyses. Taken together or individually, these analyses will result in a biological understanding of complex genomic data to improve clinical decision making.
Collapse
Affiliation(s)
- Kathleen M Fisch
- Department of Medicine, Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
186
|
Abstract
Network-aided in silico approaches have been widely used for prediction of drug-target interactions and evaluation of drug safety to increase the clinical efficiency and productivity during drug discovery and development. Here we review the advances and new progress in this field and summarize the translational applications of several new network-aided in silico approaches we developed recently. In addition, we describe the detailed protocols for a network-aided drug repositioning infrastructure for identification of new targets for old drugs, failed drugs in clinical trials, and new chemical entities. These state-of-the-art network-aided in silico approaches have been used for the discovery and development of broad-acting and targeted clinical therapies for various complex diseases, in particular for oncology drug repositioning. In this chapter, the described network-aided in silico protocols are appropriate for target-centric drug repositioning to various complex diseases, but expertise is still necessary to perform the specific oncology projects based on the cancer targets of interest.
Collapse
|
187
|
de Ávila PM, e Silva DCV, de Melo Bernardo PC, da Silva RGTM, Fachin AL, Marins M, Caritá EC. CANCROX: a cross-species cancer therapy database. Database (Oxford) 2019; 2019:baz044. [PMID: 31032838 PMCID: PMC6482323 DOI: 10.1093/database/baz044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/12/2019] [Accepted: 03/14/2019] [Indexed: 12/21/2022]
Abstract
Cancer comprises a set of more than 200 diseases resulting from the uncontrolled growth of cells that invade tissues and organs, which can spread to other regions of the body. The types of cancer found in humans are also described in animal models, a fact that has raised the interest of the scientific community in comparative oncology studies. In this study, bioinformatics tools were used to implement a computational model that uses text mining and natural language processing to construct a reference database that relates human and canine genes potentially associated with cancer, defining genetic pathways and information about cancer and cancer therapies. The CANCROX reference database was constructed by processing the scientific literature and lists more than 1300 drugs and therapies used to treat cancer, in addition to over 10 000 combinations of these drugs, including 40 types of cancer. A user-friendly interface was developed that enables researchers to search for different types of information about therapies, drug combinations, genes and types of cancer. In addition, data visualization tools allow to explore and relate different drugs and therapies for the treatment of cancer, providing information for groups studying animal models, in this case the dog, as well as groups studying cancer in humans.
Collapse
Affiliation(s)
- Paulo Muniz de Ávila
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Federal Institute of Education, Science and Technology of South of Minas Gerais
| | - Diego Cesar Valente e Silva
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Federal Institute Of Education, Science and Technology of São Paulo
| | - Paulo Cesar de Melo Bernardo
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Federal Institute Of Education, Science and Technology of São Paulo
| | - Ramon Gustavo Teodoro Marques da Silva
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Federal Institute of Education, Science and Technology of South of Minas Gerais
| | - Ana Lúcia Fachin
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
| | - Mozart Marins
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Medicine School, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
| | - Edilson Carlos Caritá
- Biotechnology Unit, University of Ribeirão Preto, Av. Costábile Romano, Ribeirão Preto, SP, Brazil
- Center for Exact, Natural and Technological Sciences, University of Ribeirão Preto, Ribeirão Preto SP, Brazil
| |
Collapse
|
188
|
Cheng F. Cardio-oncology: Network-Based Prediction of Cancer Therapy-Induced Cardiotoxicity. CHALLENGES AND ADVANCES IN COMPUTATIONAL CHEMISTRY AND PHYSICS 2019:75-97. [DOI: 10.1007/978-3-030-16443-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
189
|
Lv BM, Tong XY, Quan Y, Liu MY, Zhang QY, Song YF, Zhang HY. Drug Repurposing for Japanese Encephalitis Virus Infection by Systems Biology Methods. Molecules 2018; 23:molecules23123346. [PMID: 30567313 PMCID: PMC6320907 DOI: 10.3390/molecules23123346] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/14/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis is a zoonotic disease caused by the Japanese encephalitis virus (JEV). It is mainly epidemic in Asia with an estimated 69,000 cases occurring per year. However, no approved agents are available for the treatment of JEV infection, and existing vaccines cannot control various types of JEV strains. Drug repurposing is a new concept for finding new indication of existing drugs, and, recently, the concept has been used to discover new antiviral agents. Identifying host proteins involved in the progress of JEV infection and using these proteins as targets are the center of drug repurposing for JEV infection. In this study, based on the gene expression data of JEV infection and the phenome-wide association study (PheWAS) data, we identified 286 genes that participate in the progress of JEV infection using systems biology methods. The enrichment analysis of these genes suggested that the genes identified by our methods were predominantly related to viral infection pathways and immune response-related pathways. We found that bortezomib, which can target these genes, may have an effect on the treatment of JEV infection. Subsequently, we evaluated the antiviral activity of bortezomib using a JEV-infected mouse model. The results showed that bortezomib can lower JEV-induced lethality in mice, alleviate suffering in JEV-infected mice and reduce the damage in brains caused by JEV infection. This work provides an agent with new indication to treat JEV infection.
Collapse
Affiliation(s)
- Bo-Min Lv
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xin-Yu Tong
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Meng-Yuan Liu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Qing-Ye Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yun-Feng Song
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
190
|
Fang J, Liu C, Wang Q, Lin P, Cheng F. In silico polypharmacology of natural products. Brief Bioinform 2018; 19:1153-1171. [PMID: 28460068 DOI: 10.1093/bib/bbx045] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Indexed: 01/03/2025] Open
Abstract
Natural products with polypharmacological profiles have demonstrated promise as novel therapeutics for various complex diseases, including cancer. Currently, many gaps exist in our knowledge of which compounds interact with which targets, and experimentally testing all possible interactions is infeasible. Recent advances and developments of systems pharmacology and computational (in silico) approaches provide powerful tools for exploring the polypharmacological profiles of natural products. In this review, we introduce recent progresses and advances of computational tools and systems pharmacology approaches for identifying drug targets of natural products by focusing on the development of targeted cancer therapy. We survey the polypharmacological and systems immunology profiles of five representative natural products that are being considered as cancer therapies. We summarize various chemoinformatics, bioinformatics and systems biology resources for reconstructing drug-target networks of natural products. We then review currently available computational approaches and tools for prediction of drug-target interactions by focusing on five domains: target-based, ligand-based, chemogenomics-based, network-based and omics-based systems biology approaches. In addition, we describe a practical example of the application of systems pharmacology approaches by integrating the polypharmacology of natural products and large-scale cancer genomics data for the development of precision oncology under the systems biology framework. Finally, we highlight the promise of cancer immunotherapies and combination therapies that target tumor ecosystems (e.g. clones or 'selfish' sub-clones) via exploiting the immunological and inflammatory 'side' effects of natural products in the cancer post-genomics era.
Collapse
Affiliation(s)
- Jiansong Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuang Liu
- Alibaba Research Center for Complexity Sciences at the Hangzhou Normal University, Hangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Lin
- National Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Feixiong Cheng
- Department of Biomedical Informatics, Vanderbilt University Medical Center in Nashville (United States)
| |
Collapse
|
191
|
Integrated transcriptome interactome study of oncogenes and tumor suppressor genes in breast cancer. Genes Dis 2018; 6:78-87. [PMID: 30906836 PMCID: PMC6411624 DOI: 10.1016/j.gendis.2018.10.004] [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] [Received: 05/28/2018] [Accepted: 10/31/2018] [Indexed: 01/14/2023] Open
Abstract
Breast cancer is the leading cause for mortality among women worldwide. Dysregulation of oncogenes and tumor suppressor genes is the major reason for the cause of cancer. Understanding these genes will provide clues and insights about their regulatory mechanism and their interplay in cancer. In the present study, an attempt is made to compare the functional characteristics and interactions of oncogenes and tumor suppressor genes to understand their biological role. 431 breast cancer samples from seven publicly available microarray datasets were collected and analysed using GEO2R tool. The identified 416 differentially expressed genes were classified into five gene sets as oncogenes (OG), tumor suppressor genes (TSG), druggable genes, essential genes and other genes. The gene sets were subjected to various analysis such as enrichment analysis (viz., GO, Pathways, Diseases and Drugs), network analysis, calculation of mutation frequencies and Guanine-Cytosine (GC) content. From the results, it was observed that the OG were having high GC content as well as high interactions than TSG. Moreover, the OG are found to have frequent mutations than TSG. The enrichment analysis results suggest that the oncogenes are involved in positive regulation of cellular protein metabolic process, macromolecule biosynthetic process and majorly in cell cycle and focal adhesion pathway in cancer. It was also found that these oncogenes are involved in other diseases such as skin diseases and viral infections. Collagenase, paclitaxel and docetaxel are some of the drugs found to be enriched for oncogenes.
Collapse
|
192
|
Chen J, Sun J, Liu X, Liu F, Liu R, Wang J. Structure-based prediction of West Nile virus-human protein-protein interactions. J Biomol Struct Dyn 2018; 37:2310-2321. [PMID: 30044201 DOI: 10.1080/07391102.2018.1479659] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In recent years, West Nile virus (WNV) has posed a great threat to global human health due to its explosive spread. Studying the protein-protein interactions (PPIs) between WNV and human is beneficial for understanding the pathogenesis of WNV and the immune response mechanism of human against WNV infection at the molecular level. In this study, we identified the human target proteins which interact with WNV based on protein structure similarity, and then the interacting pairs were filtered by the subcellular co-localization information. As a result, a network of 3346 interactions was constructed, involving 6 WNV proteins and 1970 human target proteins. To our knowledge, this is the first predicted interactome for WNV-human. By analyzing the topological properties and evolutionary rates of the human target proteins, it was demonstrated that these proteins tend to be the hub and bottleneck proteins in the human PPI network and are more conserved than the non-target ones. Triplet analysis showed that the target proteins are adjacent to each other in the human PPI network, suggesting that these proteins may have similar biological functions. Further, the functional enrichment analysis indicated that the target proteins are mainly involved in virus process, transcription regulation, cell adhesion, and so on. In addition, the common and specific targets were identified and compared based on the networks between WNV-human and Dengue virus II (DENV2)-human. Finally, by combining topological features and existing drug target information, we identified 30 potential anti-WNV human targets, among which 11 ones were reported to be associated with WNV infection. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jing Chen
- a Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics , Huazhong Agricultural University , Wuhan , People's Republic of China
| | - Jun Sun
- a Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics , Huazhong Agricultural University , Wuhan , People's Republic of China
| | - Xiangming Liu
- b Gongqing Institute of Science and Technology , Gongqing , People's Republic of China
| | - Feng Liu
- a Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics , Huazhong Agricultural University , Wuhan , People's Republic of China
| | - Rong Liu
- a Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics , Huazhong Agricultural University , Wuhan , People's Republic of China
| | - Jia Wang
- a Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics , Huazhong Agricultural University , Wuhan , People's Republic of China
| |
Collapse
|
193
|
Network integration of multi-tumour omics data suggests novel targeting strategies. Nat Commun 2018; 9:4514. [PMID: 30375513 PMCID: PMC6207774 DOI: 10.1038/s41467-018-06992-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
We characterize different tumour types in search for multi-tumour drug targets, in particular aiming for drug repurposing and novel drug combinations. Starting from 11 tumour types from The Cancer Genome Atlas, we obtain three clusters based on transcriptomic correlation profiles. A network-based analysis, integrating gene expression profiles and protein interactions of cancer-related genes, allows us to define three cluster-specific signatures, with genes belonging to NF-κB signaling, chromosomal instability, ubiquitin-proteasome system, DNA metabolism, and apoptosis biological processes. These signatures have been characterized by different approaches based on mutational, pharmacological and clinical evidences, demonstrating the validity of our selection. Moreover, we define new pharmacological strategies validated by in vitro experiments that show inhibition of cell growth in two tumour cell lines, with significant synergistic effect. Our study thus provides a list of genes and pathways that could possibly be used, singularly or in combination, for the design of novel treatment strategies. Tumours of different tissues can show similarities in genomic alterations. Here, the authors combine tumour transcriptome and protein interaction data in a network-based analysis of 11 tumours types, and identify clusters of tumours with specific signatures for multi-tumour drug targeting and survival prognosis.
Collapse
|
194
|
Singer F, Irmisch A, Toussaint NC, Grob L, Singer J, Thurnherr T, Beerenwinkel N, Levesque MP, Dummer R, Quagliata L, Rothschild SI, Wicki A, Beisel C, Stekhoven DJ. SwissMTB: establishing comprehensive molecular cancer diagnostics in Swiss clinics. BMC Med Inform Decis Mak 2018; 18:89. [PMID: 30373609 PMCID: PMC6206832 DOI: 10.1186/s12911-018-0680-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022] Open
Abstract
Background Molecular precision oncology is an emerging practice to improve cancer therapy by decreasing the risk of choosing treatments that lack efficacy or cause adverse events. However, the challenges of integrating molecular profiling into routine clinical care are manifold. From a computational perspective these include the importance of a short analysis turnaround time, the interpretation of complex drug-gene and gene-gene interactions, and the necessity of standardized high-quality workflows. In addition, difficulties faced when integrating molecular diagnostics into clinical practice are ethical concerns, legal requirements, and limited availability of treatment options beyond standard of care as well as the overall lack of awareness of their existence. Methods To the best of our knowledge, we are the first group in Switzerland that established a workflow for personalized diagnostics based on comprehensive high-throughput sequencing of tumors at the clinic. Our workflow, named SwissMTB (Swiss Molecular Tumor Board), links genetic tumor alterations and gene expression to therapeutic options and clinical trial opportunities. The resulting treatment recommendations are summarized in a clinical report and discussed in a molecular tumor board at the clinic to support therapy decisions. Results Here we present results from an observational pilot study including 22 late-stage cancer patients. In this study we were able to identify actionable variants and corresponding therapies for 19 patients. Half of the patients were analyzed retrospectively. In two patients we identified resistance-associated variants explaining lack of therapy response. For five out of eleven patients analyzed before treatment the SwissMTB diagnostic influenced treatment decision. Conclusions SwissMTB enables the analysis and clinical interpretation of large numbers of potentially actionable molecular targets. Thus, our workflow paves the way towards a more frequent use of comprehensive molecular diagnostics in Swiss hospitals. Electronic supplementary material The online version of this article (10.1186/s12911-018-0680-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franziska Singer
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Anja Irmisch
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Linda Grob
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Jochen Singer
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Thomas Thurnherr
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Luca Quagliata
- Department of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4056, Basel, Switzerland
| | - Sacha I Rothschild
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Andreas Wicki
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Daniel J Stekhoven
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland. .,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.
| |
Collapse
|
195
|
Abstract
Large-scale genomic data highlight the complexity and diversity of the molecular changes that drive cancer progression. Statistical analysis of cancer data from different tissues can guide drug repositioning as well as the design of targeted treatments. Here, we develop an improved Bayesian network model for tumour mutational profiles and apply it to 8198 patient samples across 22 cancer types from TCGA. For each cancer type, we identify the interactions between mutated genes, capturing signatures beyond mere mutational frequencies. When comparing mutation networks, we find genes which interact both within and across cancer types. To detach cancer classification from the tissue type we perform de novo clustering of the pancancer mutational profiles based on the Bayesian network models. We find 22 novel clusters which significantly improve survival prediction beyond clinical information. The models highlight key gene interactions for each cluster potentially allowing genomic stratification for clinical trials and identifying drug targets. Tumour heterogeneity hinders translation of large-scale genomic data into the clinic. Here the authors develop a method for the stratification of cancer patients based on the molecular gene status, including genetic interactions, rather than clinico-histological data, and apply it to TCGA data for over 8000 cases across 22 cancer types.
Collapse
|
196
|
Spooner W, McLaren W, Slidel T, Finch DK, Butler R, Campbell J, Eghobamien L, Rider D, Kiefer CM, Robinson MJ, Hardman C, Cunningham F, Vaughan T, Flicek P, Huntington CC. Haplosaurus computes protein haplotypes for use in precision drug design. Nat Commun 2018; 9:4128. [PMID: 30297836 PMCID: PMC6175845 DOI: 10.1038/s41467-018-06542-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 09/07/2018] [Indexed: 01/08/2023] Open
Abstract
Selecting the most appropriate protein sequences is critical for precision drug design. Here we describe Haplosaurus, a bioinformatic tool for computation of protein haplotypes. Haplosaurus computes protein haplotypes from pre-existing chromosomally-phased genomic variation data. Integration into the Ensembl resource provides rapid and detailed protein haplotypes retrieval. Using Haplosaurus, we build a database of unique protein haplotypes from the 1000 Genomes dataset reflecting real-world protein sequence variability and their prevalence. For one in seven genes, their most common protein haplotype differs from the reference sequence and a similar number differs on their most common haplotype between human populations. Three case studies show how knowledge of the range of commonly encountered protein forms predicted in populations leads to insights into therapeutic efficacy. Haplosaurus and its associated database is expected to find broad applications in many disciplines using protein sequences and particularly impactful for therapeutics design.
Collapse
Affiliation(s)
- William Spooner
- Eagle Genomics Ltd., Biodata Innovation Centre, Wellcome Genome Campus, Hinxton, Cambridge, CB10 3DR UK
- Genomics England, QMUL Dawson Hall, London, EC1M 6BQ UK
| | - William McLaren
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
| | | | | | - Robin Butler
- MedImmune Ltd., Granta Park, Cambridge, CB21 4QR UK
| | | | | | - David Rider
- MedImmune Ltd., Granta Park, Cambridge, CB21 4QR UK
| | | | | | | | - Fiona Cunningham
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
| | | | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD UK
| | | |
Collapse
|
197
|
Lin P, Zhong XZ, Wang XD, Li JJ, Zhao RQ, He Y, Jiang YQ, Huang XW, Chen G, He Y, Yang H. Survival analysis of genome-wide profiles coupled with Connectivity Map database mining to identify potential therapeutic targets for cholangiocarcinoma. Oncol Rep 2018; 40:3189-3198. [PMID: 30272356 PMCID: PMC6196639 DOI: 10.3892/or.2018.6710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 09/03/2018] [Indexed: 12/25/2022] Open
Abstract
Cholangiocarcinoma (CCA) is one of the most common epithelial cell malignancies worldwide. However, its prognosis is poor. The aim of the present study was to examine the prognostic landscape and potential therapeutic targets for CCA. RNA sequencing data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) dataset and processed. A total of 172 genes that were significantly associated with overall survival of patients with CCA were identified using the univariate Cox regression method. Bioinformatics tools were applied using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO). It was identified that ‘Wnt signaling pathway’, ‘cytoplasm’ and ‘AT DNA binding’ were the three most significant GO categories of CCA survival-associated genes. ‘Transcriptional misregulation in cancer’ was the most significant pathway identified in the KEGG analysis. Using the Drug-Gene Interaction database, a drug-gene interaction network was constructed, and 31 identified genes were involved in it. The most meaningful potential therapeutic targets were selected via protein-protein and gene-drug interactions. Among these genes, polo-like kinase 1 (PLK1) was identified to be a potential target due to its significant upregulation in CCA. To rapidly find molecules that may affect these genes, the Connectivity Map was queried. A series of molecules were selected for their potential anti-CCA functions. 0297417-0002B and tribenoside exhibited the highest connection scores with PLK1 via molecular docking. These findings may offer novel insights into treatment and perspectives on the future innovative treatment of CCA.
Collapse
Affiliation(s)
- Peng Lin
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Zhu Zhong
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiao-Dong Wang
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Jian-Jun Li
- Department of General Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rui-Qi Zhao
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yu He
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yan-Qiu Jiang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xian-Wen Huang
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yun He
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Hong Yang
- Department of Medical Ultrasonics, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
198
|
Kovaliov M, Cohen-Karni D, Burridge KA, Mambelli D, Sloane S, Daman N, Xu C, Guth J, Kenneth Wickiser J, Tomycz N, Page RC, Konkolewicz D, Averick S. Grafting strategies for the synthesis of active DNase I polymer biohybrids. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2018.07.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
199
|
Podder A, Pandit M, Narayanan L. Drug Target Prioritization for Alzheimer's Disease Using Protein Interaction Network Analysis. ACTA ACUST UNITED AC 2018; 22:665-677. [DOI: 10.1089/omi.2018.0131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Avijit Podder
- Bioinformatics Infrastructure Facility, Sri Venkateswara College (University of Delhi), Delhi, India
| | - Mansi Pandit
- Bioinformatics Infrastructure Facility, Sri Venkateswara College (University of Delhi), Delhi, India
| | - Latha Narayanan
- Bioinformatics Infrastructure Facility, Sri Venkateswara College (University of Delhi), Delhi, India
| |
Collapse
|
200
|
Sayles LC, Breese MR, Koehne AL, Leung SG, Lee AG, Liu HY, Spillinger A, Shah AT, Tanasa B, Straessler K, Hazard FK, Spunt SL, Marina N, Kim GE, Cho SJ, Avedian RS, Mohler DG, Kim MO, DuBois SG, Hawkins DS, Sweet-Cordero EA. Genome-Informed Targeted Therapy for Osteosarcoma. Cancer Discov 2018; 9:46-63. [PMID: 30266815 DOI: 10.1158/2159-8290.cd-17-1152] [Citation(s) in RCA: 265] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 11/16/2022]
Abstract
Osteosarcoma is a highly aggressive cancer for which treatment has remained essentially unchanged for more than 30 years. Osteosarcoma is characterized by widespread and recurrent somatic copy-number alterations (SCNA) and structural rearrangements. In contrast, few recurrent point mutations in protein-coding genes have been identified, suggesting that genes within SCNAs are key oncogenic drivers in this disease. SCNAs and structural rearrangements are highly heterogeneous across osteosarcoma cases, suggesting the need for a genome-informed approach to targeted therapy. To identify patient-specific candidate drivers, we used a simple heuristic based on degree and rank order of copy-number amplification (identified by whole-genome sequencing) and changes in gene expression as identified by RNA sequencing. Using patient-derived tumor xenografts, we demonstrate that targeting of patient-specific SCNAs leads to significant decrease in tumor burden, providing a road map for genome-informed treatment of osteosarcoma. SIGNIFICANCE: Osteosarcoma is treated with a chemotherapy regimen established 30 years ago. Although osteosarcoma is genomically complex, we hypothesized that tumor-specific dependencies could be identified within SCNAs. Using patient-derived tumor xenografts, we found a high degree of response for "genome-matched" therapies, demonstrating the utility of a targeted genome-informed approach.This article is highlighted in the In This Issue feature, p. 1.
Collapse
Affiliation(s)
- Leanne C Sayles
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Marcus R Breese
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Amanda L Koehne
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Stanley G Leung
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Alex G Lee
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Heng-Yi Liu
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Aviv Spillinger
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Avanthi T Shah
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Bogdan Tanasa
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Krystal Straessler
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California
| | - Florette K Hazard
- Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Sheri L Spunt
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Neyssa Marina
- Division of Hematology and Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Grace E Kim
- Department of Pathology, University of California, San Francisco, California
| | - Soo-Jin Cho
- Department of Pathology, University of California, San Francisco, California
| | - Raffi S Avedian
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - David G Mohler
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Mi-Ok Kim
- Biostatistics Core, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California.,Division of Biostatistics, Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Steven G DuBois
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center and Harvard Medical School, Boston, Massachusetts
| | - Douglas S Hawkins
- Seattle Children's Hospital, University of Washington, Fred Hutchison Cancer Research Center, Seattle, Washington
| | - E Alejandro Sweet-Cordero
- Division of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, California.
| |
Collapse
|