251
|
Tsherniak A, Vazquez F, Montgomery PG, Weir BA, Kryukov G, Cowley GS, Gill S, Harrington WF, Pantel S, Krill-Burger JM, Meyers RM, Ali L, Goodale A, Lee Y, Jiang G, Hsiao J, Gerath WFJ, Howell S, Merkel E, Ghandi M, Garraway LA, Root DE, Golub TR, Boehm JS, Hahn WC. Defining a Cancer Dependency Map. Cell 2017; 170:564-576.e16. [PMID: 28753430 DOI: 10.1016/j.cell.2017.06.010] [Citation(s) in RCA: 1907] [Impact Index Per Article: 238.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/09/2017] [Accepted: 06/07/2017] [Indexed: 12/15/2022]
Abstract
Most human epithelial tumors harbor numerous alterations, making it difficult to predict which genes are required for tumor survival. To systematically identify cancer dependencies, we analyzed 501 genome-scale loss-of-function screens performed in diverse human cancer cell lines. We developed DEMETER, an analytical framework that segregates on- from off-target effects of RNAi. 769 genes were differentially required in subsets of these cell lines at a threshold of six SDs from the mean. We found predictive models for 426 dependencies (55%) by nonlinear regression modeling considering 66,646 molecular features. Many dependencies fall into a limited number of classes, and unexpectedly, in 82% of models, the top biomarkers were expression based. We demonstrated the basis behind one such predictive model linking hypermethylation of the UBB ubiquitin gene to a dependency on UBC. Together, these observations provide a foundation for a cancer dependency map that facilitates the prioritization of therapeutic targets.
Collapse
Affiliation(s)
- Aviad Tsherniak
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Francisca Vazquez
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Phil G Montgomery
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Barbara A Weir
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Gregory Kryukov
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Stanley Gill
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA
| | | | - Sasha Pantel
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | | | - Robin M Meyers
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Levi Ali
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Amy Goodale
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Yenarae Lee
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Guozhi Jiang
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Jessica Hsiao
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | | | - Sara Howell
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Erin Merkel
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Mahmoud Ghandi
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Levi A Garraway
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, USA
| | - David E Root
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - Todd R Golub
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA; Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD, USA
| | - Jesse S Boehm
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA, USA; Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA, USA.
| |
Collapse
|
252
|
Sharma A, Cinti C, Capobianco E. Multitype Network-Guided Target Controllability in Phenotypically Characterized Osteosarcoma: Role of Tumor Microenvironment. Front Immunol 2017; 8:918. [PMID: 28824643 PMCID: PMC5536125 DOI: 10.3389/fimmu.2017.00918] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022] Open
Abstract
This study highlights the relevance of network-guided controllability analysis as a precision oncology tool. Target controllability through networks is potentially relevant to cancer research for the identification of therapeutic targets. With reference to a recent study on multiple phenotypes from 22 osteosarcoma (OS) cell lines characterized both in vitro and in vivo, we found that a variety of critical proteins in OS regulation circuits were in part phenotype specific and in part shared. To generalize our inference approach and match cancer phenotypic heterogeneity, we employed multitype networks and identified targets in correspondence with protein sub-complexes. Therefore, we established the relevance for diagnostic and therapeutic purposes of inspecting interactive targets, namely those enriched by significant connectivity patterns in protein sub-complexes. Emerging targets appeared with reference to the OS microenvironment, and relatively to small leucine-rich proteoglycan members and D-type cyclins, among other collagen, laminin, and keratin proteins. These described were evidences shared across all phenotypes; instead, specific evidences were provided by critical proteins including IGFBP7 and PDGFRA in the invasive phenotype, and FGFR3 and THBS1 in the colony forming phenotype.
Collapse
Affiliation(s)
- Ankush Sharma
- Experimental Oncology Unit, UOS - Institute of Clinical Physiology, CNR, Siena, Italy.,Center for Computational Science, University of Miami, Miami, FL, United States
| | - Caterina Cinti
- Experimental Oncology Unit, UOS - Institute of Clinical Physiology, CNR, Siena, Italy
| | - Enrico Capobianco
- Center for Computational Science, University of Miami, Miami, FL, United States.,Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
253
|
Velmurugan KR, Varghese RT, Fonville NC, Garner HR. High-depth, high-accuracy microsatellite genotyping enables precision lung cancer risk classification. Oncogene 2017; 36:6383-6390. [PMID: 28759038 PMCID: PMC5701090 DOI: 10.1038/onc.2017.256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/01/2017] [Accepted: 06/13/2017] [Indexed: 12/23/2022]
Abstract
There remains a large discrepancy between the known genetic contributions to cancer and that which can be explained by genomic variants, both inherited and somatic. Recently, understudied repetitive DNA regions called microsatellites have been identified as genetic risk markers for a number of diseases including various cancers (breast, ovarian and brain). In this study, we demonstrate an integrated process for identifying and further evaluating microsatellite-based risk markers for lung cancer using data from the cancer genome atlas and the 1000 genomes project. Comparing whole-exome germline sequencing data from 488 TCGA lung cancer samples to germline exome data from 390 control samples from the 1000 genomes project, we identified 119 potentially informative microsatellite loci. These loci were found to be able to distinguish between cancer and control samples with sensitivity and specificity ratios over 0.8. Then these loci, supplemented with additional loci from other cancers and controls, were evaluated using a target enrichment kit and sample-multiplexed nextgen sequencing. Thirteen of the 119 risk markers were found to be informative in a well powered study (>0.99 for a 0.95 confidence interval) using high-depth (579x±315) nextgen sequencing of 30 lung cancer and 89 control samples, resulting in sensitivity and specificity ratios of 0.90 and 0.94, respectively. When 8 loci harvested from the bioinformatic analysis of other cancers are added to the classifier, then the sensitivity and specificity rise to 0.93 and 0.97, respectively. Analysis of the genes harboring these loci revealed two genes (ARID1B and REL) and two significantly enriched pathways (chromatin organization and cellular stress response) suggesting that the process of lung carcinogenesis is linked to chromatin remodeling, inflammation, and tumor microenvironment restructuring. We illustrate that high-depth sequencing enables a high-precision microsatellite-based risk classifier analysis approach. This microsatellite-based platform confirms the potential to create clinically actionable diagnostics for lung cancer.
Collapse
Affiliation(s)
- K R Velmurugan
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - R T Varghese
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| | - N C Fonville
- Department of Biological Sciences, Riverside Law, LLP Glenhardie Corporate Center, Wayne, PA, USA
| | - H R Garner
- Department of Biological Sciences, Center for Bioinformatics and Genetics and the Primary Care Research Network, Edward Via College of Osteopathic Medicine, Blacksburg, VA, USA.,Department of Biological Sciences, Gibbs Cancer Center and Research Institute, Spartanburg, SC, USA
| |
Collapse
|
254
|
Abstract
Most human epithelial tumors harbor numerous alterations, making it difficult to predict which genes are required for tumor survival. To systematically identify cancer dependencies, we analyzed 501 genome-scale loss-of-function screens performed in diverse human cancer cell lines. We developed DEMETER, an analytical framework that segregates on- from off-target effects of RNAi. 769 genes were differentially required in subsets of these cell lines at a threshold of six SDs from the mean. We found predictive models for 426 dependencies (55%) by nonlinear regression modeling considering 66,646 molecular features. Many dependencies fall into a limited number of classes, and unexpectedly, in 82% of models, the top biomarkers were expression based. We demonstrated the basis behind one such predictive model linking hypermethylation of the UBB ubiquitin gene to a dependency on UBC. Together, these observations provide a foundation for a cancer dependency map that facilitates the prioritization of therapeutic targets.
Collapse
|
255
|
Bridgett S, Campbell J, Lord CJ, Ryan CJ. CancerGD: A Resource for Identifying and Interpreting Genetic Dependencies in Cancer. Cell Syst 2017; 5:82-86.e3. [PMID: 28711281 PMCID: PMC5531859 DOI: 10.1016/j.cels.2017.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/04/2017] [Accepted: 06/08/2017] [Indexed: 12/26/2022]
Abstract
Genes whose function is selectively essential in the presence of cancer-associated genetic aberrations represent promising targets for the development of precision therapeutics. Here, we present CancerGD, a resource that integrates genotypic profiling with large-scale loss-of-function genetic screens in tumor cell lines to identify such genetic dependencies. CancerGD provides tools for searching, visualizing, and interpreting these genetic dependencies through the integration of functional interaction networks. CancerGD includes different screen types (siRNA, shRNA, CRISPR), and we describe a simple format for submitting new datasets.
Collapse
Affiliation(s)
- Stephen Bridgett
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - James Campbell
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Colm J Ryan
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
256
|
Zhao Y, Forst CV, Sayegh CE, Wang IM, Yang X, Zhang B. Molecular and genetic inflammation networks in major human diseases. MOLECULAR BIOSYSTEMS 2017; 12:2318-41. [PMID: 27303926 DOI: 10.1039/c6mb00240d] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
It has been well-recognized that inflammation alongside tissue repair and damage maintaining tissue homeostasis determines the initiation and progression of complex diseases. Albeit with the accomplishment of having captured the most critical inflammation-involved molecules, genetic susceptibilities, epigenetic factors, and environmental factors, our schemata on the role of inflammation in complex diseases remain largely patchy, in part due to the success of reductionism in terms of research methodology per se. Omics data alongside the advances in data integration technologies have enabled reconstruction of molecular and genetic inflammation networks which shed light on the underlying pathophysiology of complex diseases or clinical conditions. Given the proven beneficial role of anti-inflammation in coronary heart disease as well as other complex diseases and immunotherapy as a revolutionary transition in oncology, it becomes timely to review our current understanding of the molecular and genetic inflammation networks underlying major human diseases. In this review, we first briefly discuss the complexity of infectious diseases and then highlight recently uncovered molecular and genetic inflammation networks in other major human diseases including obesity, type II diabetes, coronary heart disease, late onset Alzheimer's disease, Parkinson's disease, and sporadic cancer. The commonality and specificity of these molecular networks are addressed in the context of genetics based on genome-wide association study (GWAS). The double-sword role of inflammation, such as how the aberrant type 1 and/or type 2 immunity leads to chronic and severe clinical conditions, remains open in terms of the inflammasome and the core inflammatome network features. Increasingly available large Omics and clinical data in tandem with systems biology approaches have offered an exciting yet challenging opportunity toward reconstruction of more comprehensive and dynamic molecular and genetic inflammation networks, which hold great promise in transiting network snapshots to video-style multi-scale interplays of disease mechanisms, in turn leading to effective clinical intervention.
Collapse
Affiliation(s)
- Yongzhong Zhao
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA. and Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA
| | - Christian V Forst
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA. and Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA
| | - Camil E Sayegh
- Vertex Pharmaceuticals (Canada) Incorporated, 275 Armand-Frappier, Laval, Quebec H7V 4A7, Canada
| | - I-Ming Wang
- Informatics and Analysis, Merck Research Laboratories, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA.
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90025, USA.
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA. and Institute of Genomics and Multiscale Biology, Mount Sinai School of Medicine, 1425 Madison Avenue, NY 10029, USA
| |
Collapse
|
257
|
Barardo DG, Newby D, Thornton D, Ghafourian T, de Magalhães JP, Freitas AA. Machine learning for predicting lifespan-extending chemical compounds. Aging (Albany NY) 2017; 9:1721-1737. [PMID: 28783712 PMCID: PMC5559171 DOI: 10.18632/aging.101264] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/12/2017] [Indexed: 12/12/2022]
Abstract
Increasing age is a risk factor for many diseases; therefore developing pharmacological interventions that slow down ageing and consequently postpone the onset of many age-related diseases is highly desirable. In this work we analyse data from the DrugAge database, which contains chemical compounds and their effect on the lifespan of model organisms. Predictive models were built using the machine learning method random forests to predict whether or not a chemical compound will increase Caenorhabditis elegans' lifespan, using as features Gene Ontology (GO) terms annotated for proteins targeted by the compounds and chemical descriptors calculated from each compound's chemical structure. The model with the best predictive accuracy used both biological and chemical features, achieving a prediction accuracy of 80%. The top 20 most important GO terms include those related to mitochondrial processes, to enzymatic and immunological processes, and terms related to metabolic and transport processes. We applied our best model to predict compounds which are more likely to increase C. elegans' lifespan in the DGIdb database, where the effect of the compounds on an organism's lifespan is unknown. The top hit compounds can be broadly divided into four groups: compounds affecting mitochondria, compounds for cancer treatment, anti-inflammatories, and compounds for gonadotropin-releasing hormone therapies.
Collapse
Affiliation(s)
- Diogo G. Barardo
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, UK
| | - Daniel Thornton
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
258
|
Abdul SN, Ab Mutalib NS, Sean KS, Syafruddin SE, Ishak M, Sagap I, Mazlan L, Rose IM, Abu N, Mokhtar NM, Jamal R. Molecular Characterization of Somatic Alterations in Dukes' B and C Colorectal Cancers by Targeted Sequencing. Front Pharmacol 2017; 8:465. [PMID: 28769798 PMCID: PMC5513919 DOI: 10.3389/fphar.2017.00465] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/30/2017] [Indexed: 12/12/2022] Open
Abstract
Despite global progress in research, improved screening and refined treatment strategies, colorectal cancer (CRC) remains as the third most common malignancy. As each type of cancer is different and exhibits unique alteration patterns, identifying and characterizing gene alterations in CRC that may serve as biomarkers might help to improve diagnosis, prognosis and predict potential response to therapy. With the emergence of next generation sequencing technologies (NGS), it is now possible to extensively and rapidly identify the gene profile of individual tumors. In this study, we aimed to identify actionable somatic alterations in Dukes’ B and C in CRC via NGS. Targeted sequencing of 409 cancer-related genes using the Ion AmpliseqTM Comprehensive Cancer Panel was performed on genomic DNA obtained from paired fresh frozen tissues, cancer and normal, of Dukes’ B (n = 10) and Dukes’ C (n = 9) CRC. The sequencing results were analyzed using Torrent Suite, annotated using ANNOVAR and validated using Sanger sequencing. A total of 141 somatic non-synonymous sequence variations were identified in 86 genes. Among these, 64 variants (45%) were predicted to be deleterious, 38 variants (27%) possibly deleterious while the other 39 variants (28%) have low or neutral protein impact. Seventeen genes have alterations with frequencies of ≥10% in the patient cohort and with 14 overlapped genes in both Dukes’ B and C. The adenomatous polyposis coli gene (APC) was the most frequently altered gene in both groups (n = 6 in Dukes’ B and C). In addition, TP53 was more frequently altered in Dukes’ C (n = 7) compared to Dukes’ B (n = 4). Ten variants in APC, namely p.R283∗, p.N778fs, p.R805∗, p.Y935fs, p.E941fs, p.E1057∗, p.I1401fs, p.Q1378∗, p.E1379∗, and p.A1485fs were predicted to be driver variants. APC remains as the most frequently altered gene in the intermediate stages of CRC. Wnt signaling pathway is the major affected pathway followed by P53, RAS, TGF-β, and PI3K signaling. We reported the alteration profiles in each of the patient which has the potential to affect the clinical decision. We believe that this study will add further to the understanding of CRC molecular landscape.
Collapse
Affiliation(s)
- Shafina-Nadiawati Abdul
- UKM Medical Molecular Biology InstituteUniversiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | | | - Saiful E Syafruddin
- UKM Medical Molecular Biology InstituteUniversiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Muhiddin Ishak
- UKM Medical Molecular Biology InstituteUniversiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ismail Sagap
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan MalaysiaKuala Lumpur, Malaysia
| | - Luqman Mazlan
- Department of Surgery, Faculty of Medicine, Universiti Kebangsaan MalaysiaKuala Lumpur, Malaysia
| | - Isa M Rose
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan MalaysiaKuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology InstituteUniversiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Norfilza M Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan MalaysiaKuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology InstituteUniversiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
259
|
Detecting protein variants by mass spectrometry: a comprehensive study in cancer cell-lines. Genome Med 2017; 9:62. [PMID: 28716134 PMCID: PMC5514513 DOI: 10.1186/s13073-017-0454-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/22/2017] [Indexed: 02/07/2023] Open
Abstract
Background Onco-proteogenomics aims to understand how changes in a cancer’s genome influences its proteome. One challenge in integrating these molecular data is the identification of aberrant protein products from mass-spectrometry (MS) datasets, as traditional proteomic analyses only identify proteins from a reference sequence database. Methods We established proteomic workflows to detect peptide variants within MS datasets. We used a combination of publicly available population variants (dbSNP and UniProt) and somatic variations in cancer (COSMIC) along with sample-specific genomic and transcriptomic data to examine proteome variation within and across 59 cancer cell-lines. Results We developed a set of recommendations for the detection of variants using three search algorithms, a split target-decoy approach for FDR estimation, and multiple post-search filters. We examined 7.3 million unique variant tryptic peptides not found within any reference proteome and identified 4771 mutations corresponding to somatic and germline deviations from reference proteomes in 2200 genes among the NCI60 cell-line proteomes. Conclusions We discuss in detail the technical and computational challenges in identifying variant peptides by MS and show that uncovering these variants allows the identification of druggable mutations within important cancer genes. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0454-9) contains supplementary material, which is available to authorized users.
Collapse
|
260
|
Wang JY, Chen LL, Zhou XH. Identifying prognostic signature in ovarian cancer using DirGenerank. Oncotarget 2017; 8:46398-46413. [PMID: 28615526 PMCID: PMC5542276 DOI: 10.18632/oncotarget.18189] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 04/26/2017] [Indexed: 12/27/2022] Open
Abstract
Identifying the prognostic genes in cancer is essential not only for the treatment of cancer patients, but also for drug discovery. However, it's still a big challenge to select the prognostic genes that can distinguish the risk of cancer patients across various data sets because of tumor heterogeneity. In this situation, the selected genes whose expression levels are statistically related to prognostic risks may be passengers. In this paper, based on gene expression data and prognostic data of ovarian cancer patients, we used conditional mutual information to construct gene dependency network in which the nodes (genes) with more out-degrees have more chances to be the modulators of cancer prognosis. After that, we proposed DirGenerank (Generank in direct netowrk) algorithm, which concerns both the gene dependency network and genes' correlations to prognostic risks, to identify the gene signature that can predict the prognostic risks of ovarian cancer patients. Using ovarian cancer data set from TCGA (The Cancer Genome Atlas) as training data set, 40 genes with the highest importance were selected as prognostic signature. Survival analysis of these patients divided by the prognostic signature in testing data set and four independent data sets showed the signature can distinguish the prognostic risks of cancer patients significantly. Enrichment analysis of the signature with curated cancer genes and the drugs selected by CMAP showed the genes in the signature may be drug targets for therapy. In summary, we have proposed a useful pipeline to identify prognostic genes of cancer patients.
Collapse
Affiliation(s)
- Jian-Yong Wang
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Ling-Ling Chen
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Xiong-Hui Zhou
- College of Informatics, Huazhong Agricultural University, Wuhan 430070, P.R. China
| |
Collapse
|
261
|
Ge S, Li B, Li Y, Li Z, Liu Z, Chen Z, Wu J, Gao J, Shen L. Genomic alterations in advanced gastric cancer endoscopic biopsy samples using targeted next-generation sequencing. Am J Cancer Res 2017; 7:1540-1553. [PMID: 28744403 PMCID: PMC5523034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 05/17/2017] [Indexed: 06/07/2023] Open
Abstract
Gastric cancer (GC) remains the second tumor caused death threat worldwide, and personalized medicine for GC is far from expectation. Finding novel, recurrently mutated genes through next-generation sequencing (NGS) is a powerful and productive approach. However, previous genomic data for GC are based on surgical resected samples while a large proportion of advanced gastric cancer (AGC) patients have already missed the chance for operation. The aim of this study is to assess frequent genomic alteration in AGC via biopsy samples. Here we performed targeted genomic sequencing of 78 AGC patients' tumor biopsies along with matched lymphocyte samples based on a 118 cancer related gene panel. In total, we observed 301 somatic nonsynonymous genomic alterations in 92 different genes, as well as 37 copy number gain events among 15 different genes (fold change 2-12), and validated the fold changes of ERBB2 copy number gains with IHC and FISH test showed an accuracy of 81.8%. Previously reported driver genes for gastric cancer (TP53, KMT2D, KMT2B, EGFR, PIK3CA, GNAQ, and ARID1A), and several unreported mutations (TGFBR2, RNF213, NF1, NSD1, and LRP2) showed high non-silent mutation prevalence (7.7%-34.6%). When comparing intestinal-type gastric cancer (IGC) with diffuse-type gastric cancer (DGC), TP53 and GNAQ appear to be more frequently mutated in IGC (P=0.028 and P=0.023, respectively), whereas LRP2, BRCA2 and FGFR3 mutations are not observed in IGC, but have 12.8%, 7.7% and 7.7% mutation rates, respectively, in DGC patients. Patients with one or more mutations in adherens junction pathway (CREBBP, EP300, CDH1, CTNNB1, EGFR, MET, TGFBR2 and ERBB2) or TGF-β signaling pathway (CREBBP, EP300, MYST4, KRAS and TGFBR2) showed significantly better overall survival (P=0.007 and P=0.014, respectively), consistent with The Cancer Genome Atlas (TCGA) cohort data. Importantly, 57 (73.1%) patients harbored at least one genomic alteration with potential treatments, making NGS-based drug target screening a viable option for AGC patients. Our study established a comprehensive genomic portrait of AGC, and identified several mutation signatures highly associated with clinical features, survival outcomes, which may be used to design future personalized treatments.
Collapse
Affiliation(s)
- Sai Ge
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Beifang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Yanyan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Zhongwu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Zhentao Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Zuhua Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Jian Wu
- My Genostics Inc801 West Baltimore Street, Suite 502L, Baltimore, MD 21205, USA
| | - Jing Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Lin Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and InstituteBeijing 100142, China
| |
Collapse
|
262
|
Raja K, Patrick M, Elder JT, Tsoi LC. Machine learning workflow to enhance predictions of Adverse Drug Reactions (ADRs) through drug-gene interactions: application to drugs for cutaneous diseases. Sci Rep 2017. [PMID: 28623363 PMCID: PMC5473874 DOI: 10.1038/s41598-017-03914-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adverse drug reactions (ADRs) pose critical public health issues, affecting over 6% of hospitalized patients. While knowledge of potential drug-drug interactions (DDI) is necessary to prevent ADR, the rapid pace of drug discovery makes it challenging to maintain a strong insight into DDIs. In this study, we present a novel literature-mining framework for enhancing the predictions of DDIs and ADR types by integrating drug-gene interactions (DGIs). The ADR types were adapted from a DDI corpus, including i) adverse effect; ii) effect at molecular level; iii) effect related to pharmacokinetics; and iv) DDIs without known ADRs. By using random forest classifier our approach achieves an F-score of 0.87 across the ADRs classification using only the DDI features. We then enhanced the performance of the classifier by including DGIs (F-score = 0.90), and applied the classification model trained with the DDI corpus to identify the drugs that might interact with the drugs for cutaneous diseases. We successfully predict previously known ADRs for drugs prescribed to cutaneous diseases, and are also able to identify promising new ADRs.
Collapse
Affiliation(s)
- Kalpana Raja
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James T Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
263
|
Identification of Pharmacologically Tractable Protein Complexes in Cancer Using the R-Based Network Clustering and Visualization Program MCODER. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1016305. [PMID: 28691013 PMCID: PMC5485287 DOI: 10.1155/2017/1016305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/21/2017] [Accepted: 05/23/2017] [Indexed: 12/27/2022]
Abstract
Current multiomics assay platforms facilitate systematic identification of functional entities that are mappable in a biological network, and computational methods that are better able to detect densely connected clusters of signals within a biological network are considered increasingly important. One of the most famous algorithms for detecting network subclusters is Molecular Complex Detection (MCODE). MCODE, however, is limited in simultaneous analyses of multiple, large-scale data sets, since it runs on the Cytoscape platform, which requires extensive computational resources and has limited coding flexibility. In the present study, we implemented the MCODE algorithm in R programming language and developed a related package, which we called MCODER. We found the MCODER package to be particularly useful in analyzing multiple omics data sets simultaneously within the R framework. Thus, we applied MCODER to detect pharmacologically tractable protein-protein interactions selectively elevated in molecular subtypes of ovarian and colorectal tumors. In doing so, we found that a single molecular subtype representing epithelial-mesenchymal transition in both cancer types exhibited enhanced production of the collagen-integrin protein complex. These results suggest that tumors of this molecular subtype could be susceptible to pharmacological inhibition of integrin signaling.
Collapse
|
264
|
Precision medicine for hepatocelluar carcinoma using molecular pattern diagnostics: results from a preclinical pilot study. Cell Death Dis 2017; 8:e2867. [PMID: 28594404 PMCID: PMC5520889 DOI: 10.1038/cddis.2017.229] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/07/2017] [Accepted: 04/11/2017] [Indexed: 02/07/2023]
Abstract
The aim of this study was to design a road map for personalizing cancer therapy in hepatocellular carcinoma (HCC) by using molecular pattern diagnostics. As an exploratory study, we investigated molecular patterns of tissues of two tumors from individual HCC patients, which in previous experiments had shown contrasting reactions to the phase 2 transforming growth factor beta receptor 1 inhibitor galunisertib. Cancer-driving molecular patterns encompass – inter alias – altered transcription profiles and somatic mutations in coding regions differentiating tumors from their respective peritumoral tissues and from each other. Massive analysis of cDNA ends and all-exome sequencing demonstrate a highly divergent transcriptional and mutational landscape, respectively, for the two tumors, that offers potential explanations for the tumors contrasting responses to galunisertib. Molecular pattern diagnostics (MPDs) suggest alternative, individual-tumor-specific therapies, which in both cases deviate from the standard sorafenib treatment and from each other. Suggested personalized therapies use kinase inhibitors and immune-focused drugs as well as low-toxicity natural compounds identified using an advanced bioinformatics routine included in the MPD protocol. The MPD pipeline we describe here for the prediction of suitable drugs for treatment of two contrasting HCCs may serve as a blueprint for the design of therapies for various types of cancer.
Collapse
|
265
|
Barardo D, Thornton D, Thoppil H, Walsh M, Sharifi S, Ferreira S, Anžič A, Fernandes M, Monteiro P, Grum T, Cordeiro R, De-Souza EA, Budovsky A, Araujo N, Gruber J, Petrascheck M, Fraifeld VE, Zhavoronkov A, Moskalev A, de Magalhães JP. The DrugAge database of aging-related drugs. Aging Cell 2017; 16:594-597. [PMID: 28299908 PMCID: PMC5418190 DOI: 10.1111/acel.12585] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2017] [Indexed: 11/30/2022] Open
Abstract
Aging is a major worldwide medical challenge. Not surprisingly, identifying drugs and compounds that extend lifespan in model organisms is a growing research area. Here, we present DrugAge (http://genomics.senescence.info/drugs/), a curated database of lifespan‐extending drugs and compounds. At the time of writing, DrugAge contains 1316 entries featuring 418 different compounds from studies across 27 model organisms, including worms, flies, yeast and mice. Data were manually curated from 324 publications. Using drug–gene interaction data, we also performed a functional enrichment analysis of targets of lifespan‐extending drugs. Enriched terms include various functional categories related to glutathione and antioxidant activity, ion transport and metabolic processes. In addition, we found a modest but significant overlap between targets of lifespan‐extending drugs and known aging‐related genes, suggesting that some but not most aging‐related pathways have been targeted pharmacologically in longevity studies. DrugAge is freely available online for the scientific community and will be an important resource for biogerontologists.
Collapse
Affiliation(s)
- Diogo Barardo
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Daniel Thornton
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Harikrishnan Thoppil
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
- Amrita School of Biotechnology; Amrita Vishwa Vidyapeetham (Amrita University); Coimbatore India
| | - Michael Walsh
- Energy Metabolism Laboratory; Swiss Federal Institute of Technology (ETH) Zurich; Zurich Switzerland
| | - Samim Sharifi
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Susana Ferreira
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Andreja Anžič
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Maria Fernandes
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Patrick Monteiro
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Tjaša Grum
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Rui Cordeiro
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool 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 Israel
- Judea Regional Research & Development Center; Carmel 90404 Israel
| | - Natali Araujo
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
| | - Jan Gruber
- Department of Science; Yale- NUS College; Singapore City 138527 Singapore
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore City 117597 Singapore
| | - Michael Petrascheck
- Department of Chemical Physiology; The Scripps Research Institute; La Jolla CA USA
| | - 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 Israel
| | - Alexander Zhavoronkov
- Pharmaceutical Artificial Intelligence Research Division; Emerging Technology Centers; Insilico Medicine, Inc; Johns Hopkins University at Eastern; B301, 1101 33rd Street Baltimore MD 21218 USA
- The Biogerontology Research Foundation; Oxford UK
| | - Alexey Moskalev
- Moscow Institute of Physics and Technology; Dolgoprudny 141700 Russia
- Laboratory of Molecular Radiobiology and Gerontology; Institute of Biology of Komi Science Center of Ural Branch of Russian Academy of Sciences; Syktyvkar 167982 Russia
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences; Moscow 119991 Russia
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing Group; Institute of Ageing and Chronic Disease; University of Liverpool; Liverpool UK
- The Biogerontology Research Foundation; Oxford UK
| |
Collapse
|
266
|
Priedigkeit N, Hartmaier RJ, Chen Y, Vareslija D, Basudan A, Watters RJ, Thomas R, Leone JP, Lucas PC, Bhargava R, Hamilton RL, Chmielecki J, Puhalla SL, Davidson NE, Oesterreich S, Brufsky AM, Young L, Lee AV. Intrinsic Subtype Switching and Acquired ERBB2/HER2 Amplifications and Mutations in Breast Cancer Brain Metastases. JAMA Oncol 2017; 3:666-671. [PMID: 27926948 PMCID: PMC5508875 DOI: 10.1001/jamaoncol.2016.5630] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
IMPORTANCE Patients with breast cancer (BrCa) brain metastases (BrM) have limited therapeutic options. A better understanding of molecular alterations acquired in BrM could identify clinically actionable metastatic dependencies. OBJECTIVE To determine whether there are intrinsic subtype differences between primary tumors and matched BrM and to uncover BrM-acquired alterations that are clinically actionable. DESIGN, SETTING, AND PARTICIPANTS In total, 20 cases of primary breast cancer tissue and resected BrM (10 estrogen receptor [ER]-negative and 10 ER-positive) from 2 academic institutions were included. Eligible cases in the discovery cohort harbored patient-matched primary breast cancer tissue and resected BrM. Given the rarity of patient-matched samples, no exclusion criteria were enacted. Two validation sequencing cohorts were used-a published data set of 17 patient-matched cases of BrM and a cohort of 7884 BrCa tumors enriched for metastatic samples. MAIN OUTCOMES AND MEASURES Brain metastases expression changes in 127 genes within BrCa signatures, PAM50 assignments, and ERBB2/HER2 DNA-level gains. RESULTS Overall, 17 of 20 BrM retained the PAM50 subtype of the primary BrCa. Despite this concordance, 17 of 20 BrM harbored expression changes (<2-fold or >2-fold) in clinically actionable genes including gains of FGFR4 (n = 6 [30%]), FLT1 (n = 4 [20%]), AURKA (n = 2 [10%]) and loss of ESR1 expression (n = 9 [45%]). The most recurrent expression gain was ERBB2/HER2, which showed a greater than 2-fold expression increase in 7 of 20 BrM (35%). Three of these 7 cases were ERBB2/HER2-negative out of 13 ERBB2/HER2-negative in the primary BrCa cohort and became immunohistochemical positive (3+) in the paired BrM with metastasis-specific amplification of the ERBB2/HER2 locus. In an independent data set, 2 of 9 (22.2%) ERBB2/HER2-negative BrCa switched to ERBB2/HER2-positive with 1 BrM acquiring ERBB2/HER2 amplification and the other showing metastatic enrichment of the activating V777L ERBB2/HER2 mutation. An expanded cohort revealed that ERBB2/HER2 amplification and/or mutation frequency was unchanged between local disease and metastases across all sites; however, a significant enrichment was appreciated for BrM (13% local vs 24% BrM; P < .001). CONCLUSIONS AND RELEVANCE Breast cancer BrM commonly acquire alterations in clinically actionable genes, with metastasis-acquired ERBB2/HER2 alterations in approximately 20% of ERBB2/HER2-negative cases. These observations have immediate clinical implications for patients with ERBB2/HER2-negative breast cancer and support comprehensive profiling of metastases to inform clinical care.
Collapse
Affiliation(s)
- Nolan Priedigkeit
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | | | - Yijing Chen
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Damir Vareslija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ahmed Basudan
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Rebecca J. Watters
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Roby Thomas
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Jose P. Leone
- University of Iowa Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, C32 GH. 200 Hawkins Drive, Iowa City, IA, USA
| | - Peter C. Lucas
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Rohit Bhargava
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Ronald L. Hamilton
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | | | - Shannon L. Puhalla
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Nancy E. Davidson
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Steffi Oesterreich
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Adam M. Brufsky
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| | - Leonie Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Adrian V. Lee
- Departments of Pharmacology and Chemical Biology, Human Genetics, Medicine, and Pathology, Women’s Cancer Research Center, Magee-Women’s Research Institute, University of Pittsburgh Cancer Institute, PA, USA
| |
Collapse
|
267
|
Hamilton DJ, White CM, Rees CL, Wheeler DW, Ascoli GA. Molecular fingerprinting of principal neurons in the rodent hippocampus: A neuroinformatics approach. J Pharm Biomed Anal 2017; 144:269-278. [PMID: 28549853 DOI: 10.1016/j.jpba.2017.03.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 03/05/2017] [Accepted: 03/29/2017] [Indexed: 12/17/2022]
Abstract
Neurons are often classified by their morphological and molecular properties. The online knowledge base Hippocampome.org primarily defines neuron types from the rodent hippocampal formation based on their main neurotransmitter (glutamate or GABA) and the spatial distributions of their axons and dendrites. For each neuron type, this open-access resource reports any and all published information regarding the presence or absence of known molecular markers, including calcium-binding proteins, neuropeptides, receptors, channels, transcription factors, and other molecules of biomedical relevance. The resulting chemical profile is relatively sparse: even for the best studied neuron types, the expression or lack thereof of fewer than 70 molecules has been firmly established to date. The mouse genome-wide in situ hybridization mapping of the Allen Brain Atlas provides a wealth of data that, when appropriately analyzed, can substantially augment the molecular marker knowledge in Hippocampome.org. Here we focus on the principal cell layers of dentate gyrus (DG), CA3, CA2, and CA1, which together contain approximately 90% of hippocampal neurons. These four anatomical parcels are densely packed with somata of mostly excitatory projection neurons. Thus, gene expression data for those layers can be justifiably linked to the respective principal neuron types: granule cells in DG and pyramidal cells in CA3, CA2, and CA1. In order to enable consistent interpretation across genes and regions, we screened the whole-genome dataset against known molecular markers of those neuron types. The resulting threshold values allow over 6000 very-high confidence (>99.5%) expressed/not-expressed assignments, expanding the biochemical information content of Hippocampome.org more than five-fold. Many of these newly identified molecular markers are potential pharmacological targets for major neurological and psychiatric conditions. Furthermore, our approach yields reasonable expression/non-expression estimates for every single gene in each of these four neuron types with >90% average confidence, providing a considerably complete genetic characterization of hippocampal principal neurons.
Collapse
Affiliation(s)
- D J Hamilton
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States.
| | - C M White
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States
| | - C L Rees
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States
| | - D W Wheeler
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States
| | - G A Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States.
| |
Collapse
|
268
|
Dalleau K, Marzougui Y, Da Silva S, Ringot P, Ndiaye NC, Coulet A. Learning from biomedical linked data to suggest valid pharmacogenes. J Biomed Semantics 2017; 8:16. [PMID: 28427468 PMCID: PMC5399403 DOI: 10.1186/s13326-017-0125-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 03/29/2017] [Indexed: 12/15/2022] Open
Abstract
Background A standard task in pharmacogenomics research is identifying genes that may be involved in drug response variability, i.e., pharmacogenes. Because genomic experiments tended to generate many false positives, computational approaches based on the use of background knowledge have been proposed. Until now, only molecular networks or the biomedical literature were used, whereas many other resources are available. Method We propose here to consume a diverse and larger set of resources using linked data related either to genes, drugs or diseases. One of the advantages of linked data is that they are built on a standard framework that facilitates the joint use of various sources, and thus facilitates considering features of various origins. We propose a selection and linkage of data sources relevant to pharmacogenomics, including for example DisGeNET and Clinvar. We use machine learning to identify and prioritize pharmacogenes that are the most probably valid, considering the selected linked data. This identification relies on the classification of gene–drug pairs as either pharmacogenomically associated or not and was experimented with two machine learning methods –random forest and graph kernel–, which results are compared in this article. Results We assembled a set of linked data relative to pharmacogenomics, of 2,610,793 triples, coming from six distinct resources. Learning from these data, random forest enables identifying valid pharmacogenes with a F-measure of 0.73, on a 10 folds cross-validation, whereas graph kernel achieves a F-measure of 0.81. A list of top candidates proposed by both approaches is provided and their obtention is discussed. Electronic supplementary material The online version of this article (doi:10.1186/s13326-017-0125-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin Dalleau
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Yassine Marzougui
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France.,Ecole nationale supérieure des mines de Nancy, Campus Artem, Nancy, France
| | - Sébastien Da Silva
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Patrice Ringot
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France
| | - Ndeye Coumba Ndiaye
- UMR U1122 IGE-PCV (INSERM, University of Lorraine), 30 Rue Lionnois, Nancy, France
| | - Adrien Coulet
- LORIA (CNRS, Inria Nancy-Grand Est, University of Lorraine), Campus Scientifique, Nancy, France.
| |
Collapse
|
269
|
Finan C, Gaulton A, Kruger FA, Lumbers RT, Shah T, Engmann J, Galver L, Kelley R, Karlsson A, Santos R, Overington JP, Hingorani AD, Casas JP. The druggable genome and support for target identification and validation in drug development. Sci Transl Med 2017; 9:eaag1166. [PMID: 28356508 PMCID: PMC6321762 DOI: 10.1126/scitranslmed.aag1166] [Citation(s) in RCA: 518] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/27/2017] [Indexed: 12/11/2022]
Abstract
Target identification (determining the correct drug targets for a disease) and target validation (demonstrating an effect of target perturbation on disease biomarkers and disease end points) are important steps in drug development. Clinically relevant associations of variants in genes encoding drug targets model the effect of modifying the same targets pharmacologically. To delineate drug development (including repurposing) opportunities arising from this paradigm, we connected complex disease- and biomarker-associated loci from genome-wide association studies to an updated set of genes encoding druggable human proteins, to agents with bioactivity against these targets, and, where there were licensed drugs, to clinical indications. We used this set of genes to inform the design of a new genotyping array, which will enable association studies of druggable genes for drug target selection and validation in human disease.
Collapse
Affiliation(s)
- Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Anna Gaulton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - Felix A Kruger
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- BenevolentAI, 40 Churchway, London, U.K
| | - R Thomas Lumbers
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Tina Shah
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Jorgen Engmann
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Luana Galver
- Illumina Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Ryan Kelley
- Illumina Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Anneli Karlsson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - Rita Santos
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K
| | - John P Overington
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, U.K.
- BenevolentAI, 40 Churchway, London, U.K
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London WC1E 6BT, U.K.
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K
| | - Juan P Casas
- Farr Institute of Health Informatics, University College London, London WC1E 6BT, U.K.
| |
Collapse
|
270
|
Pepke S, Ver Steeg G. Comprehensive discovery of subsample gene expression components by information explanation: therapeutic implications in cancer. BMC Med Genomics 2017; 10:12. [PMID: 28292312 PMCID: PMC5351169 DOI: 10.1186/s12920-017-0245-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 02/08/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND De novo inference of clinically relevant gene function relationships from tumor RNA-seq remains a challenging task. Current methods typically either partition patient samples into a few subtypes or rely upon analysis of pairwise gene correlations that will miss some groups in noisy data. Leveraging higher dimensional information can be expected to increase the power to discern targetable pathways, but this is commonly thought to be an intractable computational problem. METHODS In this work we adapt a recently developed machine learning algorithm for sensitive detection of complex gene relationships. The algorithm, CorEx, efficiently optimizes over multivariate mutual information and can be iteratively applied to generate a hierarchy of relatively independent latent factors. The learned latent factors are used to stratify patients for survival analysis with respect to both single factors and combinations. These analyses are performed and interpreted in the context of biological function annotations and protein network interactions that might be utilized to match patients to multiple therapies. RESULTS Analysis of ovarian tumor RNA-seq samples demonstrates the algorithm's power to infer well over one hundred biologically interpretable gene cohorts, several times more than standard methods such as hierarchical clustering and k-means. The CorEx factor hierarchy is also informative, with related but distinct gene clusters grouped by upper nodes. Some latent factors correlate with patient survival, including one for a pathway connected with the epithelial-mesenchymal transition in breast cancer that is regulated by a microRNA that modulates epigenetics. Further, combinations of factors lead to a synergistic survival advantage in some cases. CONCLUSIONS In contrast to studies that attempt to partition patients into a small number of subtypes (typically 4 or fewer) for treatment purposes, our approach utilizes subgroup information for combinatoric transcriptional phenotyping. Considering only the 66 gene expression groups that are found to both have significant Gene Ontology enrichment and are small enough to indicate specific drug targets implies a computational phenotype for ovarian cancer that allows for 366 possible patient profiles, enabling truly personalized treatment. The findings here demonstrate a new technique that sheds light on the complexity of gene expression dependencies in tumors and could eventually enable the use of patient RNA-seq profiles for selection of personalized and effective cancer treatments.
Collapse
Affiliation(s)
| | - Greg Ver Steeg
- Information Sciences Institute, University of Southern California, Marina Del Rey, USA
| |
Collapse
|
271
|
C Yuen RK, Merico D, Bookman M, L Howe J, Thiruvahindrapuram B, Patel RV, Whitney J, Deflaux N, Bingham J, Wang Z, Pellecchia G, Buchanan JA, Walker S, Marshall CR, Uddin M, Zarrei M, Deneault E, D'Abate L, Chan AJS, Koyanagi S, Paton T, Pereira SL, Hoang N, Engchuan W, Higginbotham EJ, Ho K, Lamoureux S, Li W, MacDonald JR, Nalpathamkalam T, Sung WWL, Tsoi FJ, Wei J, Xu L, Tasse AM, Kirby E, Van Etten W, Twigger S, Roberts W, Drmic I, Jilderda S, Modi BM, Kellam B, Szego M, Cytrynbaum C, Weksberg R, Zwaigenbaum L, Woodbury-Smith M, Brian J, Senman L, Iaboni A, Doyle-Thomas K, Thompson A, Chrysler C, Leef J, Savion-Lemieux T, Smith IM, Liu X, Nicolson R, Seifer V, Fedele A, Cook EH, Dager S, Estes A, Gallagher L, Malow BA, Parr JR, Spence SJ, Vorstman J, Frey BJ, Robinson JT, Strug LJ, Fernandez BA, Elsabbagh M, Carter MT, Hallmayer J, Knoppers BM, Anagnostou E, Szatmari P, Ring RH, Glazer D, Pletcher MT, Scherer SW. Whole genome sequencing resource identifies 18 new candidate genes for autism spectrum disorder. Nat Neurosci 2017; 20:602-611. [PMID: 28263302 DOI: 10.1038/nn.4524] [Citation(s) in RCA: 575] [Impact Index Per Article: 71.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/01/2017] [Indexed: 12/13/2022]
Abstract
We are performing whole-genome sequencing of families with autism spectrum disorder (ASD) to build a resource (MSSNG) for subcategorizing the phenotypes and underlying genetic factors involved. Here we report sequencing of 5,205 samples from families with ASD, accompanied by clinical information, creating a database accessible on a cloud platform and through a controlled-access internet portal. We found an average of 73.8 de novo single nucleotide variants and 12.6 de novo insertions and deletions or copy number variations per ASD subject. We identified 18 new candidate ASD-risk genes and found that participants bearing mutations in susceptibility genes had significantly lower adaptive ability (P = 6 × 10-4). In 294 of 2,620 (11.2%) of ASD cases, a molecular basis could be determined and 7.2% of these carried copy number variations and/or chromosomal abnormalities, emphasizing the importance of detecting all forms of genetic variation as diagnostic and therapeutic targets in ASD.
Collapse
Affiliation(s)
- Ryan K C Yuen
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Daniele Merico
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Deep Genomics Inc., Toronto, Canada
| | - Matt Bookman
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Jennifer L Howe
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Bhooma Thiruvahindrapuram
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Rohan V Patel
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Joe Whitney
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Nicole Deflaux
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Jonathan Bingham
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | - Zhuozhi Wang
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Janet A Buchanan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Susan Walker
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
| | - Mohammed Uddin
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Mehdi Zarrei
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Eric Deneault
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Lia D'Abate
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Ada J S Chan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Stephanie Koyanagi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Tara Paton
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Sergio L Pereira
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Ny Hoang
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Worrawat Engchuan
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Edward J Higginbotham
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Karen Ho
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Sylvia Lamoureux
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Weili Li
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Jeffrey R MacDonald
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Thomas Nalpathamkalam
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Wilson W L Sung
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Fiona J Tsoi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - John Wei
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Lizhen Xu
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Anne-Marie Tasse
- Public Population Project in Genomics and Society, McGill University, Montreal, Canada
| | - Emily Kirby
- Public Population Project in Genomics and Society, McGill University, Montreal, Canada
| | | | | | - Wendy Roberts
- Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Irene Drmic
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Sanne Jilderda
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Bonnie MacKinnon Modi
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Autism Research Unit, The Hospital for Sick Children, Toronto, Canada
| | - Barbara Kellam
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Michael Szego
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Dalla Lana School of Public Health and the Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl Cytrynbaum
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Dalla Lana School of Public Health and the Department of Family and Community Medicine, University of Toronto, Toronto, Ontario, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada
| | - Rosanna Weksberg
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada
| | | | - Marc Woodbury-Smith
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Jessica Brian
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | - Lili Senman
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | - Alana Iaboni
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | | | - Ann Thompson
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Christina Chrysler
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Canada
| | - Jonathan Leef
- Bloorview Research Institute, University of Toronto, Toronto, Canada.
| | | | - Isabel M Smith
- Departments of Pediatrics and of Psychology &Neuroscience, Dalhousie University and Autism Research Centre, IWK Health Centre, Halifax, Canada
| | - Xudong Liu
- Department of Psychiatry, Queen's University, Kinston, Canada
| | - Rob Nicolson
- Children's Health Research Institute, London, Ontario, Canada.,Western University, London, Ontario, Canada
| | | | | | - Edwin H Cook
- Institute for Juvenile Research, Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Stephen Dager
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington, Seattle, Washington, USA
| | - Louise Gallagher
- Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Beth A Malow
- Sleep Disorders Division, Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jeremy R Parr
- Institute of Neuroscience, Newcastle University, Newcastle Upon Tyne, UK
| | - Sarah J Spence
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jacob Vorstman
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Brendan J Frey
- Deep Genomics Inc., Toronto, Canada.,Department of Electrical and Computer Engineering and Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Canada
| | - James T Robinson
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lisa J Strug
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Canada
| | - Bridget A Fernandez
- Disciplines of Genetics and Medicine, Memorial University of Newfoundland and Provincial Medical Genetic Program, Eastern Health, St. John's, Canada
| | | | - Melissa T Carter
- Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, Canada.,Regional Genetics Program, The Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Joachim Hallmayer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California, USA
| | | | | | - Peter Szatmari
- Child Youth and Family Services, Centre for Addiction and Mental Health, Toronto, Canada.,Department of Psychiatry, University of Toronto, Toronto, Canada.,Department of Psychiatry, The Hospital for Sick Children, Toronto, Canada
| | - Robert H Ring
- Department of Pharmacology &Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - David Glazer
- Google, Mountain View, California, USA.,Verily Life Sciences, South San Francisco, California, USA
| | | | - Stephen W Scherer
- The Centre for Applied Genomics, Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,McLaughlin Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
272
|
Nguyen DT, Mathias S, Bologa C, Brunak S, Fernandez N, Gaulton A, Hersey A, Holmes J, Jensen LJ, Karlsson A, Liu G, Ma'ayan A, Mandava G, Mani S, Mehta S, Overington J, Patel J, Rouillard AD, Schürer S, Sheils T, Simeonov A, Sklar LA, Southall N, Ursu O, Vidovic D, Waller A, Yang J, Jadhav A, Oprea TI, Guha R. Pharos: Collating protein information to shed light on the druggable genome. Nucleic Acids Res 2017; 45:D995-D1002. [PMID: 27903890 PMCID: PMC5210555 DOI: 10.1093/nar/gkw1072] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023] Open
Abstract
The 'druggable genome' encompasses several protein families, but only a subset of targets within them have attracted significant research attention and thus have information about them publicly available. The Illuminating the Druggable Genome (IDG) program was initiated in 2014, has the goal of developing experimental techniques and a Knowledge Management Center (KMC) that would collect and organize information about protein targets from four families, representing the most common druggable targets with an emphasis on understudied proteins. Here, we describe two resources developed by the KMC: the Target Central Resource Database (TCRD) which collates many heterogeneous gene/protein datasets and Pharos (https://pharos.nih.gov), a multimodal web interface that presents the data from TCRD. We briefly describe the types and sources of data considered by the KMC and then highlight features of the Pharos interface designed to enable intuitive access to the IDG knowledgebase. The aim of Pharos is to encourage 'serendipitous browsing', whereby related, relevant information is made easily discoverable. We conclude by describing two use cases that highlight the utility of Pharos and TCRD.
Collapse
Affiliation(s)
- Dac-Trung Nguyen
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Stephen Mathias
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Cristian Bologa
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Soren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Nicolas Fernandez
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Anna Gaulton
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Anne Hersey
- European Molecular Biology Laboratory - European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Jayme Holmes
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | | | - Guixia Liu
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
- East China University of Science and Technology, Dept. Pharmaceutical Sciences, Shanghai, China
| | - Avi Ma'ayan
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Geetha Mandava
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Subramani Mani
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Saurabh Mehta
- Center for Computational Science, University of Miami, Coral Gables, FL 33146, USA
- Department of Applied Chemistry, Delhi Technological University, Delhi 110042, India
| | | | - Juhee Patel
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
- BA/MD Program, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Andrew D Rouillard
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Stephan Schürer
- Center for Computational Science, University of Miami, Coral Gables, FL 33146, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Timothy Sheils
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Larry A Sklar
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
- Center for Molecular Discovery, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Noel Southall
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Oleg Ursu
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Dusica Vidovic
- Center for Computational Science, University of Miami, Coral Gables, FL 33146, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anna Waller
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
- Center for Molecular Discovery, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Jeremy Yang
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Ajit Jadhav
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Tudor I Oprea
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Rajarshi Guha
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| |
Collapse
|
273
|
Rees CL, White CM, Ascoli GA. Neurochemical Markers in the Mammalian Brain: Structure, Roles in Synaptic Communication, and Pharmacological Relevance. Curr Med Chem 2017; 24:3077-3103. [PMID: 28413962 PMCID: PMC5646670 DOI: 10.2174/0929867324666170414163506] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/15/2017] [Accepted: 04/10/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Knowledge of molecular marker (typically protein or mRNA) expression in neural systems can provide insight to the chemical blueprint of signal processing and transmission, assist in tracking developmental or pathological progressions, and yield key information regarding potential medicinal targets. These markers are particularly relevant in the mammalian brain in the light of its unsurpassed cellular diversity. Accordingly, molecular expression profiling is rapidly becoming a major approach to classify neuron types. Despite a profusion of research, however, the biological functions of molecular markers commonly used to distinguish neuron types remain incompletely understood. Furthermore, most molecular markers of mammalian neuron types are also present in other organs, therefore complicating considerations of their potential pharmacological interactions. OBJECTIVE Here, we survey 15 prominent neurochemical markers from five categories, namely membrane transporters, calcium-binding proteins, neuropeptides, receptors, and extracellular matrix proteins, explaining their relation and relevance to synaptic communication. METHOD For each marker, we summarize fundamental structural features, cellular functionality, distributions within and outside the brain, as well as known drug effectors and mechanisms of action. CONCLUSION This essential primer thus links together the cellular complexity of the brain, the chemical properties of key molecular players in neurotransmission, and possible biomedical opportunities.
Collapse
Affiliation(s)
- Christopher L. Rees
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Charise M. White
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Giorgio A. Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| |
Collapse
|
274
|
Jorge P, Pérez-Pérez M, Pérez Rodríguez G, Fdez-Riverola F, Pereira MO, Lourenço A. Construction of antimicrobial peptide-drug combination networks from scientific literature based on a semi-automated curation workflow. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:baw143. [PMID: 28025336 PMCID: PMC5199187 DOI: 10.1093/database/baw143] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 10/02/2016] [Accepted: 10/07/2016] [Indexed: 01/25/2023]
Abstract
Considerable research efforts are being invested in the development of novel antimicrobial therapies effective against the growing number of multi-drug resistant pathogens. Notably, the combination of different agents is increasingly explored as means to exploit and improve individual agent actions while minimizing microorganism resistance. Although there are several databases on antimicrobial agents, scientific literature is the primary source of information on experimental antimicrobial combination testing. This work presents a semi-automated database curation workflow that supports the mining of scientific literature and enables the reconstruction of recently documented antimicrobial combinations. Currently, the database contains data on antimicrobial combinations that have been experimentally tested against Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli, Listeria monocytogenes and Candida albicans, which are prominent pathogenic organisms and are well-known for their wide and growing resistance to conventional antimicrobials. Researchers are able to explore the experimental results for a single organism or across organisms. Likewise, researchers may look into indirect network associations and identify new potential combinations to be tested. The database is available without charges. Database URL:http://sing.ei.uvigo.es/antimicrobialCombination/
Collapse
Affiliation(s)
- Paula Jorge
- CEB - Centre of Biological Engineering LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Martín Pérez-Pérez
- ESEI - Department of Computer Science, University of Vigo, Ourense, Spain
| | | | | | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering LIBRO - Laboratory of Research in Biofilms Rosário Oliveira, University of Minho, Braga, Portugal
| | - Anália Lourenço
- ESEI - Department of Computer Science, University of Vigo, Ourense, Spain .,CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
| |
Collapse
|
275
|
Cheng S, Andrew AS, Andrews PC, Moore JH. Complex systems analysis of bladder cancer susceptibility reveals a role for decarboxylase activity in two genome-wide association studies. BioData Min 2016; 9:40. [PMID: 27999618 PMCID: PMC5154053 DOI: 10.1186/s13040-016-0119-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 12/02/2016] [Indexed: 11/24/2022] Open
Abstract
Background Bladder cancer is common disease with a complex etiology that is likely due to many different genetic and environmental factors. The goal of this study was to embrace this complexity using a bioinformatics analysis pipeline designed to use machine learning to measure synergistic interactions between single nucleotide polymorphisms (SNPs) in two genome-wide association studies (GWAS) and then to assess their enrichment within functional groups defined by Gene Ontology. The significance of the results was evaluated using permutation testing and those results that replicated between the two GWAS data sets were reported. Results In the first step of our bioinformatics pipeline, we estimated the pairwise synergistic effects of SNPs on bladder cancer risk in both GWAS data sets using Multifactor Dimensionality Reduction (MDR) machine learning method that is designed specifically for this purpose. Statistical significance was assessed using a 1000-fold permutation test. Each single SNP was assigned a p-value based on its strongest pairwise association. Each SNP was then mapped to one or more genes using a window of 500 kb upstream and downstream from each gene boundary. This window was chosen to capture as many regulatory variants as possible. Using Exploratory Visual Analysis (EVA), we then carried out a gene set enrichment analysis at the gene level to identify those genes with an overabundance of significant SNPs relative to the size of their mapped regions. Each gene was assigned to a biological functional group defined by Gene Ontology (GO). We next used EVA to evaluate the overabundance of significant genes in biological functional groups. Our study yielded one GO category, carboxy-lysase activity (GO:0016831), that was significant in analyses from both GWAS data sets. Interestingly, only the gamma-glutamyl carboxylase (GGCX) gene from this GO group was significant in both the detection and replication data, highlighting the complexity of the pathway-level effects on risk. The GGCX gene is expressed in the bladder, but has not been previously associated with bladder cancer in univariate GWAS. However, there is some experimental evidence that carboxy-lysase activity might play a role in cancer and that genes in this pathway should be explored as drug targets. This study provides a genetic basis for that observation. Conclusions Our machine learning analysis of genetic associations in two GWAS for bladder cancer identified numerous associations with pairs of SNPs. Gene set enrichment analysis found aggregation of risk-associated SNPs in genes and significant genes in GO functional groups. This study supports a role for decarboxylase protein complexes in bladder cancer susceptibility. Previous research has implicated decarboxylases in bladder cancer etiology; however, the genes that we found to be significant in the detection and replication data are not known to have direct influence on bladder cancer, suggesting some novel hypotheses. This study highlights the need for a complex systems approach to the genetic and genomic analysis of common diseases such as cancer.
Collapse
Affiliation(s)
- Samantha Cheng
- Department of Biostatistics and Epidemiology, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6116 USA
| | - Angeline S Andrew
- Department of Epidemiology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755 USA
| | - Peter C Andrews
- Department of Biostatistics and Epidemiology, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6116 USA
| | - Jason H Moore
- Department of Biostatistics and Epidemiology, Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6116 USA
| |
Collapse
|
276
|
Rahmati S, Abovsky M, Pastrello C, Jurisica I. pathDIP: an annotated resource for known and predicted human gene-pathway associations and pathway enrichment analysis. Nucleic Acids Res 2016; 45:D419-D426. [PMID: 27899558 PMCID: PMC5210562 DOI: 10.1093/nar/gkw1082] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 09/30/2016] [Accepted: 10/25/2016] [Indexed: 01/06/2023] Open
Abstract
Molecular pathway data are essential in current computational and systems biology research. While there are many primary and integrated pathway databases, several challenges remain, including low proteome coverage (57%), low overlap across different databases, unavailability of direct information about underlying physical connectivity of pathway members, and high fraction of protein-coding genes without any pathway annotations, i.e. ‘pathway orphans’. In order to address all these challenges, we developed pathDIP, which integrates data from 20 source pathway databases, ‘core pathways’, with physical protein–protein interactions to predict biologically relevant protein–pathway associations, referred to as ‘extended pathways’. Cross-validation determined 71% recovery rate of our predictions. Data integration and predictions increase coverage of pathway annotations for protein-coding genes to 86%, and provide novel annotations for 5732 pathway orphans. PathDIP (http://ophid.utoronto.ca/pathdip) annotates 17 070 protein-coding genes with 4678 pathways, and provides multiple query, analysis and output options.
Collapse
Affiliation(s)
- Sara Rahmati
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Mark Abovsky
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, TMDT, Room 11-314, Toronto, ON M5G 1L7, Canada
| | - Chiara Pastrello
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, TMDT, Room 11-314, Toronto, ON M5G 1L7, Canada
| | - Igor Jurisica
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada .,Princess Margaret Cancer Centre, University Health Network, 101 College Street, TMDT, Room 11-314, Toronto, ON M5G 1L7, Canada.,Department of Computer Science, University of Toronto, Toronto, ON, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
277
|
Suhaimi SS, Ab Mutalib NS, Jamal R. Understanding Molecular Landscape of Endometrial Cancer through Next Generation Sequencing: What We Have Learned so Far? Front Pharmacol 2016; 7:409. [PMID: 27847479 PMCID: PMC5088199 DOI: 10.3389/fphar.2016.00409] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 10/14/2016] [Indexed: 01/06/2023] Open
Abstract
Endometrial cancer (EC) is among the most common gynecological cancers affecting women worldwide. Despite the early detection and rather high overall survival rate, around 20% of the cases recur with poor prognosis. The Next Generation Sequencing (NGS) technology, also known as massively parallel sequencing, symbolizes a high-throughput, fast, sensitive and accurate way to study the molecular landscape of a cancer and this has indeed revolutionized endometrial cancer research. Understanding the potential, advantages, and limitations of NGS will be crucial for the healthcare providers and scientists in providing the genome-driven care in this era of precision medicine and pharmacogenomics. This mini review aimed to compile and critically summarize the recent findings contributed by NGS technology pertaining to EC. Importantly, we also discussed the potential of this technology for fundamental discovery research, individualized therapy, screening of at-risk individual and early diagnosis.
Collapse
Affiliation(s)
- Siti-Syazani Suhaimi
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia Cheras, Malaysia
| | | | - Rahman Jamal
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia Cheras, Malaysia
| |
Collapse
|
278
|
Kosarek N, Ho ES. IDICAP: A Novel Tool for Integrating Drug Intervention Based on Cancer Panel. J Pers Med 2016; 6:jpm6040019. [PMID: 27801815 PMCID: PMC5198058 DOI: 10.3390/jpm6040019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/20/2023] Open
Abstract
Cancer is a heterogeneous disease afflicting millions of people of all ages and their families worldwide. Tremendous resources have been and continue to be devoted to the development of cancer treatments that target the unique mutation profiles of patients, namely targeted cancer therapy. However, the sheer volume of drugs coupled with cancer heterogeneity becomes a challenge for physicians to prescribe effective therapies targeting patients’ unique genetic mutations. Developing a web service that allows clinicians as well as patients to identify effective drug therapies, both approved and experimental, would be helpful for both parties. We have developed an innovative web service, IDICAP, which stands for Integrated Drug Intervention for CAncer Panel. It uses genes that have been linked to a cancer type to search for drug and clinical trial information from ClinicalTrials.gov and DrugBank. IDICAP selects and integrates information pertaining to clinical trials, disease conditions, drugs under trial, locations of trials, drugs that are known to target the queried gene, and any known single nucleotide polymorphism (SNP) effects. We tested IDICAP by gene panels that contribute to breast cancer, ovarian cancer, and cancer in general. Clinical trials and drugs listed by our tool showed improved precision compared to the results from ClinicalTrials.gov and Drug Gene Interaction Database (DGIdb). Furthermore, IDICAP provides patients and doctors with a list of clinical facilities in their proximity, a characteristic that lends credence to the Precision Medicine Initiative launched by the White House in the United States in 2015. URL:http://idicap.lafayette.edu:8000
Collapse
Affiliation(s)
- Noelle Kosarek
- Department of Biology, Lafayette College, Easton, PA 18042, USA.
- Current address: Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA.
| | - Eric S Ho
- Department of Biology, Lafayette College, Easton, PA 18042, USA.
| |
Collapse
|
279
|
Hu B, Yang YCT, Huang Y, Zhu Y, Lu ZJ. POSTAR: a platform for exploring post-transcriptional regulation coordinated by RNA-binding proteins. Nucleic Acids Res 2016; 45:D104-D114. [PMID: 28053162 PMCID: PMC5210617 DOI: 10.1093/nar/gkw888] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 01/01/2023] Open
Abstract
We present POSTAR (http://POSTAR.ncrnalab.org), a resource of POST-trAnscriptional Regulation coordinated by RNA-binding proteins (RBPs). Precise characterization of post-transcriptional regulatory maps has accelerated dramatically in the past few years. Based on new studies and resources, POSTAR supplies the largest collection of experimentally probed (∼23 million) and computationally predicted (approximately 117 million) RBP binding sites in the human and mouse transcriptomes. POSTAR annotates every transcript and its RBP binding sites using extensive information regarding various molecular regulatory events (e.g., splicing, editing, and modification), RNA secondary structures, disease-associated variants, and gene expression and function. Moreover, POSTAR provides a friendly, multi-mode, integrated search interface, which helps users to connect multiple RBP binding sites with post-transcriptional regulatory events, phenotypes, and diseases. Based on our platform, we were able to obtain novel insights into post-transcriptional regulation, such as the putative association between CPSF6 binding, RNA structural domains, and Li-Fraumeni syndrome SNPs. In summary, POSTAR represents an early effort to systematically annotate post-transcriptional regulatory maps and explore the putative roles of RBPs in human diseases.
Collapse
Affiliation(s)
- Boqin Hu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Center for Plant Biology and Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yu-Cheng T Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Center for Plant Biology and Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.,Department of Statistics, University of California Los Angeles, Los Angeles, CA 90095-1554, USA
| | - Yiming Huang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Center for Plant Biology and Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yumin Zhu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Center for Plant Biology and Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Center for Plant Biology and Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
280
|
Thurnherr T, Singer F, Stekhoven DJ, Beerenwinkel N. Genomic variant annotation workflow for clinical applications. F1000Res 2016; 5:1963. [PMID: 27990260 PMCID: PMC5130070 DOI: 10.12688/f1000research.9357.2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/17/2016] [Indexed: 12/31/2022] Open
Abstract
Annotation and interpretation of DNA aberrations identified through next-generation sequencing is becoming an increasingly important task. Even more so in the context of data analysis pipelines for medical applications, where genomic aberrations are associated with phenotypic and clinical features. Here we describe a workflow to identify potential gene targets in aberrated genes or pathways and their corresponding drugs. To this end, we provide the R/Bioconductor package rDGIdb, an R wrapper to query the drug-gene interaction database (DGIdb). DGIdb accumulates drug-gene interaction data from 15 different resources and allows filtering on different levels. The rDGIdb package makes these resources and tools available to R users. Moreover, rDGIdb queries can be automated through incorporation of the rDGIdb package into NGS sequencing pipelines.
Collapse
Affiliation(s)
- Thomas Thurnherr
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Daniel J. Stekhoven
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
- NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
281
|
Thurnherr T, Singer F, Stekhoven DJ, Beerenwinkel N. Genomic variant annotation workflow for clinical applications. F1000Res 2016; 5:1963. [PMID: 27990260 DOI: 10.12688/f1000research.9357.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/11/2016] [Indexed: 03/27/2024] Open
Abstract
Annotation and interpretation of DNA aberrations identified through next-generation sequencing is becoming an increasingly important task. Even more so in the context of data analysis pipelines for medical applications, where genomic aberrations are associated with phenotypic and clinical features. Here we describe a workflow to identify potential gene targets in aberrated genes or pathways and their corresponding drugs. To this end, we provide the R/Bioconductor package rDGIdb, an R wrapper to query the drug-gene interaction database (DGIdb). DGIdb accumulates drug-gene interaction data from 15 different source databases and allows filtering on different levels. The rDGIdb package makes these resources and tools available to R users. Moreover, DGIdb queries can be automated through incorporation of the rDGIdb package into NGS sequencing pipelines.
Collapse
Affiliation(s)
- Thomas Thurnherr
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Franziska Singer
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland; NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Daniel J Stekhoven
- SIB Swiss Institute of Bioinformatics, Basel, Switzerland; NEXUS Personalized Health Technologies, ETH Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland; SIB Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
282
|
Samatov TR, Senyavina NV, Galatenko VV, Trushkin EV, Tonevitskaya SA, Alexandrov DE, Shibukhova GP, Schumacher U, Tonevitsky AG. Tumour-like druggable gene expression pattern of CaCo2 cells in microfluidic chip. BIOCHIP JOURNAL 2016. [DOI: 10.1007/s13206-016-0308-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
283
|
Griffith M, Griffith OL, Krysiak K, Skidmore ZL, Christopher MJ, Klco JM, Ramu A, Lamprecht TL, Wagner AH, Campbell KM, Lesurf R, Hundal J, Zhang J, Spies NC, Ainscough BJ, Larson DE, Heath SE, Fronick C, O'Laughlin S, Fulton RS, Magrini V, McGrath S, Smith SM, Miller CA, Maher CA, Payton JE, Walker JR, Eldred JM, Walter MJ, Link DC, Graubert TA, Westervelt P, Kulkarni S, DiPersio JF, Mardis ER, Wilson RK, Ley TJ. Comprehensive genomic analysis reveals FLT3 activation and a therapeutic strategy for a patient with relapsed adult B-lymphoblastic leukemia. Exp Hematol 2016; 44:603-13. [PMID: 27181063 PMCID: PMC4914477 DOI: 10.1016/j.exphem.2016.04.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 10/21/2022]
Abstract
The genomic events responsible for the pathogenesis of relapsed adult B-lymphoblastic leukemia (B-ALL) are not yet clear. We performed integrative analysis of whole-genome, whole-exome, custom capture, whole-transcriptome (RNA-seq), and locus-specific genomic assays across nine time points from a patient with primary de novo B-ALL. Comprehensive genome and transcriptome characterization revealed a dramatic tumor evolution during progression, yielding a tumor with complex clonal architecture at second relapse. We observed and validated point mutations in EP300 and NF1, a highly expressed EP300-ZNF384 gene fusion, a microdeletion in IKZF1, a focal deletion affecting SETD2, and large deletions affecting RB1, PAX5, NF1, and ETV6. Although the genome analysis revealed events of potential biological relevance, no clinically actionable treatment options were evident at the time of the second relapse. However, transcriptome analysis identified aberrant overexpression of the targetable protein kinase encoded by the FLT3 gene. Although the patient had refractory disease after salvage therapy for the second relapse, treatment with the FLT3 inhibitor sunitinib rapidly induced a near complete molecular response, permitting the patient to proceed to a matched-unrelated donor stem cell transplantation. The patient remains in complete remission more than 4 years later. Analysis of this patient's relapse genome revealed an unexpected, actionable therapeutic target that led to a specific therapy associated with a rapid clinical response. For some patients with relapsed or refractory cancers, this approach may indicate a novel therapeutic intervention that could alter outcome.
Collapse
Affiliation(s)
- Malachi Griffith
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Genetics, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA.
| | - Obi L Griffith
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Genetics, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Kilannin Krysiak
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | | | | | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Avinash Ramu
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Tamara L Lamprecht
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alex H Wagner
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Katie M Campbell
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Robert Lesurf
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Jin Zhang
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Nicholas C Spies
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Benjamin J Ainscough
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA
| | - David E Larson
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Sharon E Heath
- Department of Medicine, Washington University, St. Louis, MO, USA
| | - Catrina Fronick
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Shelly O'Laughlin
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Robert S Fulton
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Vincent Magrini
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Sean McGrath
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Scott M Smith
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Christopher A Miller
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Christopher A Maher
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Jacqueline E Payton
- Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA; Department of Pathology, Washington University, St. Louis, MO, USA
| | - Jason R Walker
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - James M Eldred
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA
| | - Matthew J Walter
- Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Daniel C Link
- Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | | | - Peter Westervelt
- Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | | | - John F DiPersio
- Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Genetics, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Genetics, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA
| | - Timothy J Ley
- McDonnell Genome Institute, Washington University, St. Louis, MO, USA; Department of Genetics, Washington University, St. Louis, MO, USA; Siteman Cancer Center, Washington University, St. Louis, MO, USA; Department of Medicine, Washington University, St. Louis, MO, USA.
| |
Collapse
|
284
|
Skidmore ZL, Wagner AH, Lesurf R, Campbell KM, Kunisaki J, Griffith OL, Griffith M. GenVisR: Genomic Visualizations in R. Bioinformatics 2016; 32:3012-4. [PMID: 27288499 PMCID: PMC5039916 DOI: 10.1093/bioinformatics/btw325] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/18/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Visualizing and summarizing data from genomic studies continues to be a challenge. Here, we introduce the GenVisR package to addresses this challenge by providing highly customizable, publication-quality graphics focused on cohort level genome analyses. GenVisR provides a rapid and easy-to-use suite of genomic visualization tools, while maintaining a high degree of flexibility by leveraging the abilities of ggplot2 and Bioconductor. AVAILABILITY AND IMPLEMENTATION GenVisR is an R package available via Bioconductor (https://bioconductor.org/packages/GenVisR) under GPLv3. Support is available via GitHub (https://github.com/griffithlab/GenVisR/issues) and the Bioconductor support website. CONTACTS obigriffith@wustl.edu or mgriffit@wustl.edu SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zachary L Skidmore
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Alex H Wagner
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Robert Lesurf
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Katie M Campbell
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jason Kunisaki
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Obi L Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA Department of Medicine Siteman Cancer Center Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA Siteman Cancer Center Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
285
|
Tajti J, Szok D, Majláth Z, Csáti A, Petrovics-Balog A, Vécsei L. Alleviation of pain in painful diabetic neuropathy. Expert Opin Drug Metab Toxicol 2016; 12:753-64. [PMID: 27149100 DOI: 10.1080/17425255.2016.1184648] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Painful diabetic neuropathy (PDN) is a disabling pain condition. Its pathomechanism remains unknown, but a sensitization and neuronal hyperexcitabilty have been suggested. Only symptomatic pharmacological pain management treatment is currently available. AREAS COVERED The origin of PDN is enigmatic, and the evidence-based therapeutic guidelines therefore consist only of antidepressants and antiepileptics as first-line recommended drugs. This article relates to a MEDLINE/PubMed systematic search (2005-2015). EXPERT OPINION The results of the meta-analysis from the aspect of the efficacy of amitriptyline, duloxetine, venlafaxine, gabapentin and pregabalin are favorable, but the placebo response rate is relatively high in patients with neuropathic pain. For personalization of the medication of PDN patients, the optimum dosing, the genotyping of the metabolizing enzymes and optimum biomarkers are needed. As concerns the future perspectives, specific sodium channel subtype inhibitors acting on peripheral nociceptive neurons or modified T-type voltage-gated calcium channel blockers may be promising targets for pharmaceutical innovations. Another attractive strategy for the treatment is based on the effects of monoclonal antibodies against nerve growth factor, sodium channels, specific receptor and cytokines. Botulinum toxin A, capsaicin patch and spinal cord stimulation therapies are the nearest future therapeutic options for the treatment of PDN patients.
Collapse
Affiliation(s)
- János Tajti
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary
| | - Délia Szok
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary
| | - Zsófia Majláth
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary
| | - Anett Csáti
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary
| | - Anna Petrovics-Balog
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary
| | - László Vécsei
- a Department of Neurology, Faculty of Medicine , University of Szeged , Szeged , Hungary.,b MTA - SZTE Neuroscience Research Group , Szeged , Hungary
| |
Collapse
|
286
|
Griffith OL, Griffith M, Krysiak K, Magrini V, Ramu A, Skidmore ZL, Kunisaki J, Austin R, McGrath S, Zhang J, Demeter R, Graves T, Eldred JM, Walker J, Larson DE, Maher CA, Lin Y, Chapman W, Mahadevan A, Miksad R, Nasser I, Hanto DW, Mardis ER. A genomic case study of mixed fibrolamellar hepatocellular carcinoma. Ann Oncol 2016; 27:1148-1154. [PMID: 27029710 PMCID: PMC4880064 DOI: 10.1093/annonc/mdw135] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/07/2016] [Indexed: 12/28/2022] Open
Abstract
We report the first comprehensive genomic analysis of a case of mixed conventional and fibrolamellar HCC (mFL-HCC). This study confirms the expression of DNAJB1:PRKACA, a fusion previously associated with pure FL-HCC but not conventional HCC, in mFL-HCC. These results indicate the DNAJB1:PRKACA fusion has diagnostic utility for both pure and mixed FL-HCC. Background Mixed fibrolamellar hepatocellular carcinoma (mFL-HCC) is a rare liver tumor defined by the presence of both pure FL-HCC and conventional HCC components, represents up to 25% of cases of FL-HCC, and has been associated with worse prognosis. Recent genomic characterization of pure FL-HCC identified a highly recurrent transcript fusion (DNAJB1:PRKACA) not found in conventional HCC. Patients and Methods We performed exome and transcriptome sequencing of a case of mFL-HCC. A novel BAC-capture approach was developed to identify a 400 kb deletion as the underlying genomic mechanism for a DNAJB1:PRKACA fusion in this case. A sensitive Nanostring Elements assay was used to screen for this transcript fusion in a second case of mFL-HCC, 112 additional HCC samples and 44 adjacent non-tumor liver samples. Results We report the first comprehensive genomic analysis of a case of mFL-HCC. No common HCC-associated mutations were identified. The very low mutation rate of this case, large number of mostly single-copy, long-range copy number variants, and high expression of ERBB2 were more consistent with previous reports of pure FL-HCC than conventional HCC. In particular, the DNAJB1:PRKACA fusion transcript specifically associated with pure FL-HCC was detected at very high expression levels. Subsequent analysis revealed the presence of this fusion in all primary and metastatic samples, including those with mixed or conventional HCC pathology. A second case of mFL-HCC confirmed our finding that the fusion was detectable in conventional components. An expanded screen identified a third case of fusion-positive HCC, which upon review, also had both conventional and fibrolamellar features. This screen confirmed the absence of the fusion in all conventional HCC and adjacent non-tumor liver samples. Conclusion These results indicate that mFL-HCC is similar to pure FL-HCC at the genomic level and the DNAJB1:PRKACA fusion can be used as a diagnostic tool for both pure and mFL-HCC.
Collapse
Affiliation(s)
- O L Griffith
- McDonnell Genome Institute; Department of Medicine; Siteman Cancer Center; Department of Genetics.
| | - M Griffith
- McDonnell Genome Institute; Siteman Cancer Center; Department of Genetics
| | | | - V Magrini
- McDonnell Genome Institute; Department of Genetics
| | - A Ramu
- McDonnell Genome Institute
| | | | | | | | | | | | | | | | | | | | - D E Larson
- McDonnell Genome Institute; Department of Genetics
| | - C A Maher
- McDonnell Genome Institute; Department of Medicine; Siteman Cancer Center
| | - Y Lin
- Department of Surgery, Washington University School of Medicine, St Louis
| | - W Chapman
- Department of Surgery, Washington University School of Medicine, St Louis
| | | | | | - I Nasser
- Pathology, Harvard Medical School, Boston
| | - D W Hanto
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, USA
| | - E R Mardis
- McDonnell Genome Institute; Department of Medicine; Siteman Cancer Center; Department of Genetics
| |
Collapse
|
287
|
Piscuoglio S, Ng CKY, Murray MP, Guerini-Rocco E, Martelotto LG, Geyer FC, Bidard FC, Berman S, Fusco N, Sakr RA, Eberle CA, De Mattos-Arruda L, Macedo GS, Akram M, Baslan T, Hicks JB, King TA, Brogi E, Norton L, Weigelt B, Hudis CA, Reis-Filho JS. The Genomic Landscape of Male Breast Cancers. Clin Cancer Res 2016; 22:4045-56. [PMID: 26960396 DOI: 10.1158/1078-0432.ccr-15-2840] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/29/2016] [Indexed: 01/07/2023]
Abstract
PURPOSE Male breast cancer is rare, and its genomic landscape has yet to be fully characterized. Lacking studies in men, treatment of males with breast cancer is extrapolated from results in females with breast cancer. We sought to define whether male breast cancers harbor somatic genetic alterations in genes frequently altered in female breast cancers. EXPERIMENTAL DESIGN All male breast cancers were estrogen receptor-positive, and all but two were HER2-negative. Fifty-nine male breast cancers were subtyped by immunohistochemistry, and tumor-normal pairs were microdissected and subjected to massively parallel sequencing targeting all exons of 241 genes frequently mutated in female breast cancers or DNA-repair related. The repertoires of somatic mutations and copy number alterations of male breast cancers were compared with that of subtype-matched female breast cancers. RESULTS Twenty-nine percent and 71% of male breast cancers were immunohistochemically classified as luminal A-like or luminal B-like, respectively. Male breast cancers displayed a heterogeneous repertoire of somatic genetic alterations that to some extent recapitulated that of estrogen receptor (ER)-positive/HER2-negative female breast cancers, including recurrent mutations affecting PIK3CA (20%) and GATA3 (15%). ER-positive/HER2-negative male breast cancers, however, less frequently harbored 16q losses, and PIK3CA and TP53 mutations than ER-positive/HER2-negative female breast cancers. In addition, male breast cancers were found to be significantly enriched for mutations affecting DNA repair-related genes. CONCLUSIONS Male breast cancers less frequently harbor somatic genetic alterations typical of ER-positive/HER2-negative female breast cancers, such as PIK3CA and TP53 mutations and losses of 16q, suggesting that at least a subset of male breast cancers are driven by a distinct repertoire of somatic changes. Given the genomic differences, caution may be needed in the application of biologic and therapeutic findings from studies of female breast cancers to male breast cancers. Clin Cancer Res; 22(16); 4045-56. ©2016 AACR.
Collapse
Affiliation(s)
- Salvatore Piscuoglio
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charlotte K Y Ng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Melissa P Murray
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elena Guerini-Rocco
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pathology, European Institute of Oncology, Milan, Italy
| | - Luciano G Martelotto
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Felipe C Geyer
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Pathology, Hospital Israelita Albert Einstein, Instituto Israelita de Ensino e Pesquisa, São Paulo, Brazil
| | - Francois-Clement Bidard
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medical Oncology, Institut Curie, Paris, France
| | - Samuel Berman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nicola Fusco
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York. Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Rita A Sakr
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Carey A Eberle
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Gabriel S Macedo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Muzaffar Akram
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timour Baslan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Department of Molecular and Cellular Biology, Stony Brook University, Stony Brook, New York. Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - James B Hicks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | - Tari A King
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Clifford A Hudis
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
288
|
Network Biomarkers of Bladder Cancer Based on a Genome-Wide Genetic and Epigenetic Network Derived from Next-Generation Sequencing Data. DISEASE MARKERS 2016; 2016:4149608. [PMID: 27034531 PMCID: PMC4789422 DOI: 10.1155/2016/4149608] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/02/2016] [Indexed: 12/15/2022]
Abstract
Epigenetic and microRNA (miRNA) regulation are associated with carcinogenesis and the development of cancer. By using the available omics data, including those from next-generation sequencing (NGS), genome-wide methylation profiling, candidate integrated genetic and epigenetic network (IGEN) analysis, and drug response genome-wide microarray analysis, we constructed an IGEN system based on three coupling regression models that characterize protein-protein interaction networks (PPINs), gene regulatory networks (GRNs), miRNA regulatory networks (MRNs), and epigenetic regulatory networks (ERNs). By applying system identification method and principal genome-wide network projection (PGNP) to IGEN analysis, we identified the core network biomarkers to investigate bladder carcinogenic mechanisms and design multiple drug combinations for treating bladder cancer with minimal side-effects. The progression of DNA repair and cell proliferation in stage 1 bladder cancer ultimately results not only in the derepression of miR-200a and miR-200b but also in the regulation of the TNF pathway to metastasis-related genes or proteins, cell proliferation, and DNA repair in stage 4 bladder cancer. We designed a multiple drug combination comprising gefitinib, estradiol, yohimbine, and fulvestrant for treating stage 1 bladder cancer with minimal side-effects, and another multiple drug combination comprising gefitinib, estradiol, chlorpromazine, and LY294002 for treating stage 4 bladder cancer with minimal side-effects.
Collapse
|