651
|
Heterozygosity Ratio, a Robust Global Genomic Measure of Autozygosity and Its Association with Height and Disease Risk. Genetics 2016; 204:893-904. [PMID: 27585849 DOI: 10.1534/genetics.116.189936] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023] Open
Abstract
Greater genetic variability in an individual is protective against recessive disease. However, existing quantifications of autozygosity, such as runs of homozygosity (ROH), have proved highly sensitive to genotyping density and have yielded inconclusive results about the relationship of diversity and disease risk. Using genotyping data from three data sets with >43,000 subjects, we demonstrated that an alternative approach to quantifying genetic variability, the heterozygosity ratio, is a robust measure of diversity and is positively associated with the nondisease trait height and several disease phenotypes in subjects of European ancestry. The heterozygosity ratio is the number of heterozygous sites in an individual divided by the number of nonreference homozygous sites and is strongly affected by the degree of genetic admixture of the population and varies across human populations. Unlike quantifications of ROH, the heterozygosity ratio is not sensitive to the density of genotyping performed. Our results establish the heterozygosity ratio as a powerful new statistic for exploring the patterns and phenotypic effects of different levels of genetic variation in populations.
Collapse
|
652
|
Denny JC, Bastarache L, Roden DM. Phenome-Wide Association Studies as a Tool to Advance Precision Medicine. Annu Rev Genomics Hum Genet 2016; 17:353-73. [PMID: 27147087 PMCID: PMC5480096 DOI: 10.1146/annurev-genom-090314-024956] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Beginning in the early 2000s, the accumulation of biospecimens linked to electronic health records (EHRs) made possible genome-phenome studies (i.e., comparative analyses of genetic variants and phenotypes) using only data collected as a by-product of typical health care. In addition to disease and trait genetics, EHRs proved a valuable resource for analyzing pharmacogenetic traits and developing reverse genetics approaches such as phenome-wide association studies (PheWASs). PheWASs are designed to survey which of many phenotypes may be associated with a given genetic variant. PheWAS methods have been validated through replication of hundreds of known genotype-phenotype associations, and their use has differentiated between true pleiotropy and clinical comorbidity, added context to genetic discoveries, and helped define disease subtypes, and may also help repurpose medications. PheWAS methods have also proven to be useful with research-collected data. Future efforts that integrate broad, robust collection of phenotype data (e.g., EHR data) with purpose-collected research data in combination with a greater understanding of EHR data will create a rich resource for increasingly more efficient and detailed genome-phenome analysis to usher in new discoveries in precision medicine.
Collapse
Affiliation(s)
- Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
| | - Dan M Roden
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37203;
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| |
Collapse
|
653
|
Prieto ML, Ryu E, Jenkins GD, Batzler A, Nassan MM, Cuellar-Barboza AB, Pathak J, McElroy SL, Frye MA, Biernacka JM. Leveraging electronic health records to study pleiotropic effects on bipolar disorder and medical comorbidities. Transl Psychiatry 2016; 6:e870. [PMID: 27529678 PMCID: PMC5022084 DOI: 10.1038/tp.2016.138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 05/13/2016] [Accepted: 06/15/2016] [Indexed: 01/27/2023] Open
Abstract
Patients with bipolar disorder (BD) have a high prevalence of comorbid medical illness. However, the mechanisms underlying these comorbidities with BD are not well known. Certain genetic variants may have pleiotropic effects, increasing the risk of BD and other medical illnesses simultaneously. In this study, we evaluated the association of BD-susceptibility genetic variants with various medical conditions that tend to co-exist with BD, using electronic health records (EHR) data linked to genome-wide single-nucleotide polymorphism (SNP) data. Data from 7316 Caucasian subjects were used to test the association of 19 EHR-derived phenotypes with 34 SNPs that were previously reported to be associated with BD. After Bonferroni multiple testing correction, P<7.7 × 10(-5) was considered statistically significant. The top association findings suggested that the BD risk alleles at SNP rs4765913 in CACNA1C gene and rs7042161 in SVEP1 may be associated with increased risk of 'cardiac dysrhythmias' (odds ratio (OR)=1.1, P=3.4 × 10(-3)) and 'essential hypertension' (OR=1.1, P=3.5 × 10(-3)), respectively. Although these associations are not statistically significant after multiple testing correction, both genes have been previously implicated with cardiovascular phenotypes. Moreover, we present additional evidence supporting these associations, particularly the association of the SVEP1 SNP with hypertension. This study shows the potential for EHR-based analyses of large cohorts to discover pleiotropic effects contributing to complex psychiatric traits and commonly co-occurring medical conditions.
Collapse
Affiliation(s)
- M L Prieto
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Universidad de los Andes, Facultad de Medicina, Departamento de Psiquiatría, Santiago, Chile
| | - E Ryu
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - G D Jenkins
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Batzler
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - M M Nassan
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A B Cuellar-Barboza
- Department of Psychiatry, Universidad Autónoma de Nuevo León, Nuevo León, Mexico
| | - J Pathak
- Division of Health Informatics, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - S L McElroy
- Lindner Center of HOPE, Mason, OH, USA
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M A Frye
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - J M Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic College of Medicine, Rochester, MN, USA
- Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
654
|
eMERGE Phenome-Wide Association Study (PheWAS) identifies clinical associations and pleiotropy for stop-gain variants. BMC Med Genomics 2016; 9 Suppl 1:32. [PMID: 27535653 PMCID: PMC4989894 DOI: 10.1186/s12920-016-0191-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We explored premature stop-gain variants to test the hypothesis that variants, which are likely to have a consequence on protein structure and function, will reveal important insights with respect to the phenotypes associated with them. We performed a phenome-wide association study (PheWAS) exploring the association between a selected list of functional stop-gain genetic variants (variation resulting in truncated proteins or in nonsense-mediated decay) and an extensive group of diagnoses to identify novel associations and uncover potential pleiotropy. RESULTS In this study, we selected 25 stop-gain variants: 5 stop-gain variants with previously reported phenotypic associations, and a set of 20 putative stop-gain variants identified using dbSNP. For the PheWAS, we used data from the electronic MEdical Records and GEnomics (eMERGE) Network across 9 sites with a total of 41,057 unrelated patients. We divided all these samples into two datasets by equal proportion of eMERGE site, sex, race, and genotyping platform. We calculated single effect associations between these 25 stop-gain variants and ICD-9 defined case-control diagnoses. We also performed stratified analyses for samples of European and African ancestry. Associations were adjusted for sex, site, genotyping platform and the first three principal components to account for global ancestry. We identified previously known associations, such as variants in LPL associated with hyperglyceridemia indicating that our approach was robust. We also found a total of three significant associations with p < 0.01 in both datasets, with the most significant replicating result being LPL SNP rs328 and ICD-9 code 272.1 "Disorder of Lipoid metabolism" (pdiscovery = 2.59x10-6, preplicating = 2.7x10-4). The other two significant replicated associations identified by this study are: variant rs1137617 in KCNH2 gene associated with ICD-9 code category 244 "Acquired Hypothyroidism" (pdiscovery = 5.31x103, preplicating = 1.15x10-3) and variant rs12060879 in DPT gene associated with ICD-9 code category 996 "Complications peculiar to certain specified procedures" (pdiscovery = 8.65x103, preplicating = 4.16x10-3). CONCLUSION In conclusion, this PheWAS revealed novel associations of stop-gained variants with interesting phenotypes (ICD-9 codes) along with pleiotropic effects.
Collapse
|
655
|
Phenome-Wide Association Study to Explore Relationships between Immune System Related Genetic Loci and Complex Traits and Diseases. PLoS One 2016; 11:e0160573. [PMID: 27508393 PMCID: PMC4980020 DOI: 10.1371/journal.pone.0160573] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/16/2016] [Indexed: 12/21/2022] Open
Abstract
We performed a Phenome-Wide Association Study (PheWAS) to identify interrelationships between the immune system genetic architecture and a wide array of phenotypes from two de-identified electronic health record (EHR) biorepositories. We selected variants within genes encoding critical factors in the immune system and variants with known associations with autoimmunity. To define case/control status for EHR diagnoses, we used International Classification of Diseases, Ninth Revision (ICD-9) diagnosis codes from 3,024 Geisinger Clinic MyCode® subjects (470 diagnoses) and 2,899 Vanderbilt University Medical Center BioVU biorepository subjects (380 diagnoses). A pooled-analysis was also carried out for the replicating results of the two data sets. We identified new associations with potential biological relevance including SNPs in tumor necrosis factor (TNF) and ankyrin-related genes associated with acute and chronic sinusitis and acute respiratory tract infection. The two most significant associations identified were for the C6orf10 SNP rs6910071 and “rheumatoid arthritis” (ICD-9 code category 714) (pMETAL = 2.58 x 10−9) and the ATN1 SNP rs2239167 and “diabetes mellitus, type 2” (ICD-9 code category 250) (pMETAL = 6.39 x 10−9). This study highlights the utility of using PheWAS in conjunction with EHRs to discover new genotypic-phenotypic associations for immune-system related genetic loci.
Collapse
|
656
|
Teixeira PL, Wei WQ, Cronin RM, Mo H, VanHouten JP, Carroll RJ, LaRose E, Bastarache LA, Rosenbloom ST, Edwards TL, Roden DM, Lasko TA, Dart RA, Nikolai AM, Peissig PL, Denny JC. Evaluating electronic health record data sources and algorithmic approaches to identify hypertensive individuals. J Am Med Inform Assoc 2016; 24:162-171. [PMID: 27497800 DOI: 10.1093/jamia/ocw071] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 04/03/2016] [Accepted: 04/07/2016] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Phenotyping algorithms applied to electronic health record (EHR) data enable investigators to identify large cohorts for clinical and genomic research. Algorithm development is often iterative, depends on fallible investigator intuition, and is time- and labor-intensive. We developed and evaluated 4 types of phenotyping algorithms and categories of EHR information to identify hypertensive individuals and controls and provide a portable module for implementation at other sites. MATERIALS AND METHODS We reviewed the EHRs of 631 individuals followed at Vanderbilt for hypertension status. We developed features and phenotyping algorithms of increasing complexity. Input categories included International Classification of Diseases, Ninth Revision (ICD9) codes, medications, vital signs, narrative-text search results, and Unified Medical Language System (UMLS) concepts extracted using natural language processing (NLP). We developed a module and tested portability by replicating 10 of the best-performing algorithms at the Marshfield Clinic. RESULTS Random forests using billing codes, medications, vitals, and concepts had the best performance with a median area under the receiver operator characteristic curve (AUC) of 0.976. Normalized sums of all 4 categories also performed well (0.959 AUC). The best non-NLP algorithm combined normalized ICD9 codes, medications, and blood pressure readings with a median AUC of 0.948. Blood pressure cutoffs or ICD9 code counts alone had AUCs of 0.854 and 0.908, respectively. Marshfield Clinic results were similar. CONCLUSION This work shows that billing codes or blood pressure readings alone yield good hypertension classification performance. However, even simple combinations of input categories improve performance. The most complex algorithms classified hypertension with excellent recall and precision.
Collapse
Affiliation(s)
- Pedro L Teixeira
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert M Cronin
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Huan Mo
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jacob P VanHouten
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eric LaRose
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, 1000 N Oak Ave - ML8, Marshfield, WI 54449, USA
| | - Lisa A Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - S Trent Rosenbloom
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Todd L Edwards
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Dan M Roden
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Thomas A Lasko
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Richard A Dart
- Center for Human Genetics, Marshfield Clinic Research Foundation, 1000 N Oak Ave-MLR, Marshfield, WI 54449, USA
| | - Anne M Nikolai
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, 1000 N Oak Ave - ML8, Marshfield, WI 54449, USA
| | - Peggy L Peissig
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, 1000 N Oak Ave - ML8, Marshfield, WI 54449, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA .,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
657
|
Wang L, Fang Y, Aref D, Rathi S, Shen L, Jiang X, Wang S. PALME: PAtients Like My gEnome. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2016; 2016:219-24. [PMID: 27570674 PMCID: PMC5001758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
PAtients Like My gEnome (PALME) is a webservice that matches patients based on their genome and healthcare profiles. We support two types of inputs: (1) dual query (a variant + phenotype), and (2) genome sequences. For the first type of queries, we will show the patient profile matching the inputs. For the second type of queries, we will calculate similarity (based on Hamming distance) and show the distribution of phenotypes of similar patients given the input sequences of a target patient. Using the publicly available Personal Genome Project (PGP) dataset, we retrieved 4,360 patients' profiles along with their genome data, medical conditions, and treatments. We used a subset of these profiles to build PALME to be an interactive system to support healthcare profile matching. PALME is designed not only for biomedical researchers to support their studies on human genome but also for individuals to explore their own genetics and health. The webservice is accessible at (http://pgp.ucsd-dbmi.org:3838/GenAnaly/PatientGen/#) and the demo videos are available at (https://youtu.be/ycP0rXQizlc).
Collapse
Affiliation(s)
- Lichang Wang
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Fang
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Dima Aref
- New Jersey Institute of Technology, Newark, New Jersey
| | - Suyash Rathi
- Electrical Engineering & Computer Science, Syracuse University, Syracuse, New York
| | - Li Shen
- Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai, China
| | - Xiaoqian Jiang
- Department of Biomedical Informatics, University of California San Diego, La Jolla, California
| | - Shuang Wang
- Department of Biomedical Informatics, University of California San Diego, La Jolla, California
| |
Collapse
|
658
|
In Search of 'Birth Month Genes': Using Existing Data Repositories to Locate Genes Underlying Birth Month-Disease Relationships. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2016; 2016:189-98. [PMID: 27570668 PMCID: PMC5001771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prenatal and perinatal exposures vary seasonally (e.g., sunlight, allergens) and many diseases are linked with variance in exposure. Epidemiologists often measure these changes using birth month as a proxy for seasonal variance. Likewise, Genome-Wide Association Studies have associated or implicated these same diseases with many genes. Both disparate data types (epidemiological and genetic) can provide key insights into the underlying disease biology. We developed an algorithm that links 1) epidemiological data from birth month studies with 2) genetic data from published gene-disease association studies. Our framework uses existing data repositories - PubMed, DisGeNET and Gene Ontology - to produce a bipartite network that connects enriched seasonally varying biofactorss with birth month dependent diseases (BMDDs) through their overlapping developmental gene sets. As a proof-of-concept, we investigate 7 known BMDDs and highlight three important biological networks revealed by our algorithm and explore some interesting genetic mechanisms potentially responsible for the seasonal contribution to BMDDs.
Collapse
|
659
|
Richesson RL, Sun J, Pathak J, Kho AN, Denny JC. Clinical phenotyping in selected national networks: demonstrating the need for high-throughput, portable, and computational methods. Artif Intell Med 2016; 71:57-61. [PMID: 27506131 PMCID: PMC5480212 DOI: 10.1016/j.artmed.2016.05.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/30/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The combination of phenomic data from electronic health records (EHR) and clinical data repositories with dense biological data has enabled genomic and pharmacogenomic discovery, a first step toward precision medicine. Computational methods for the identification of clinical phenotypes from EHR data will advance our understanding of disease risk and drug response, and support the practice of precision medicine on a national scale. METHODS Based on our experience within three national research networks, we summarize the broad approaches to clinical phenotyping and highlight the important role of these networks in the progression of high-throughput phenotyping and precision medicine. We provide supporting literature in the form of a non-systematic review. RESULTS The practice of clinical phenotyping is evolving to meet the growing demand for scalable, portable, and data driven methods and tools. The resources required for traditional phenotyping algorithms from expert defined rules are significant. In contrast, machine learning approaches that rely on data patterns will require fewer clinical domain experts and resources. CONCLUSIONS Machine learning approaches that generate phenotype definitions from patient features and clinical profiles will result in truly computational phenotypes, derived from data rather than experts. Research networks and phenotype developers should cooperate to develop methods, collaboration platforms, and data standards that will enable computational phenotyping and truly modernize biomedical research and precision medicine.
Collapse
Affiliation(s)
- Rachel L Richesson
- Duke University School of Nursing, 311 Trent Drive, Durham, NC 27710 USA.
| | - Jimeng Sun
- School of Computational Science and Engineering, Georgia Institute of Technology, 266 Ferst Drive, Atlanta, GA 30313, USA.
| | - Jyotishman Pathak
- Department of Health Sciences Research, 200 1st Street SW, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Abel N Kho
- Departments of Medicine and Preventive Medicine, Northwestern University, 633 N St. Clair St. 20th floor. Chicago IL 60611, USA.
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, 2525 West End Ave, Suite 672, Nashville, TN 37203, USA.
| |
Collapse
|
660
|
Greene CS, Voight BF. Pathway and network-based strategies to translate genetic discoveries into effective therapies. Hum Mol Genet 2016; 25:R94-R98. [PMID: 27340225 DOI: 10.1093/hmg/ddw160] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/19/2016] [Indexed: 11/13/2022] Open
Abstract
One way to design a drug is to attempt to phenocopy a genetic variant that is known to have the desired effect. In general, drugs that are supported by genetic associations progress further in the development pipeline. However, the number of associations that are candidates for development into drugs is limited because many associations are in non-coding regions or difficult to target genes. Approaches that overlay information from pathway databases or biological networks can expand the potential target list. In cases where the initial variant is not targetable or there is no variant with the desired effect, this may reveal new means to target a disease. In this review, we discuss recent examples in the domain of pathway and network-based drug repositioning from genetic associations. We highlight important caveats and challenges for the field, and we discuss opportunities for further development.
Collapse
Affiliation(s)
- Casey S Greene
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine Institute for Translational Medicine and Therapeutics, Perelman School of Medicine
| | - Benjamin F Voight
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine Institute for Translational Medicine and Therapeutics, Perelman School of Medicine Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19103 USA
| |
Collapse
|
661
|
Fisher HM, Hoehndorf R, Bazelato BS, Dadras SS, King LE, Gkoutos GV, Sundberg JP, Schofield PN. DermO; an ontology for the description of dermatologic disease. J Biomed Semantics 2016; 7:38. [PMID: 27296450 PMCID: PMC4907256 DOI: 10.1186/s13326-016-0085-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 06/03/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There have been repeated initiatives to produce standard nosologies and terminologies for cutaneous disease, some dedicated to the domain and some part of bigger terminologies such as ICD-10. Recently, formally structured terminologies, ontologies, have been widely developed in many areas of biomedical research. Primarily, these address the aim of providing comprehensive working terminologies for domains of knowledge, but because of the knowledge contained in the relationships between terms they can also be used computationally for many purposes. RESULTS We have developed an ontology of cutaneous disease, constructed manually by domain experts. With more than 3000 terms, DermO represents the most comprehensive formal dermatological disease terminology available. The disease entities are categorized in 20 upper level terms, which use a variety of features such as anatomical location, heritability, affected cell or tissue type, or etiology, as the features for classification, in line with professional practice and nosology in dermatology. Available in OBO flatfile and OWL 2 formats, it is integrated semantically with other ontologies and terminologies describing diseases and phenotypes. We demonstrate the application of DermO to text mining the biomedical literature and in the creation of a network describing the phenotypic relationships between cutaneous diseases. CONCLUSIONS DermO is an ontology with broad coverage of the domain of dermatologic disease and we demonstrate here its utility for text mining and investigation of phenotypic relationships between dermatologic disorders. We envision that in the future it may be applied to the creation and mining of electronic health records, clinical training and basic research, as it supports automated inference and reasoning, and for the broader integration of skin disease information with that from other domains.
Collapse
Affiliation(s)
- Hannah M Fisher
- Dept. of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Robert Hoehndorf
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, 23955-6900, Thuwal, Kingdom of Saudi Arabia
| | - Bruno S Bazelato
- Dept. of Computer Science, Llandinam Building, Aberystwyth University, Aberystwyth, Ceredigion, SY23 3DB, UK
| | - Soheil S Dadras
- Dept. Dermatology and Pathology, University of Connecticut Health Center, 263, Farmington Avenue, Farmington, CT, 06030, USA
| | - Lloyd E King
- Dept. of Medicine, Div. Dermatology, Vanderbilt University, Nashville, Tennessee, USA
| | - Georgios V Gkoutos
- Dept. of Computer Science, Llandinam Building, Aberystwyth University, Aberystwyth, Ceredigion, SY23 3DB, UK.,College of Medical and Dental Sciences, Institute of Cancer and Genomic Sciences, Centre for Computational Biology, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TT, UK
| | - John P Sundberg
- The Jackson Laboratory, 600, Main Street, Bar Harbor Maine, ME 04609-1500, USA
| | - Paul N Schofield
- Dept. of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK. .,The Jackson Laboratory, 600, Main Street, Bar Harbor Maine, ME 04609-1500, USA.
| |
Collapse
|
662
|
Liu J, Ye Z, Mayer JG, Hoch BA, Green C, Rolak L, Cold C, Khor SS, Zheng X, Miyagawa T, Tokunaga K, Brilliant MH, Hebbring SJ. Phenome-wide association study maps new diseases to the human major histocompatibility complex region. J Med Genet 2016; 53:681-9. [PMID: 27287392 DOI: 10.1136/jmedgenet-2016-103867] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/19/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Over 160 disease phenotypes have been mapped to the major histocompatibility complex (MHC) region on chromosome 6 by genome-wide association study (GWAS), suggesting that the MHC region as a whole may be involved in the aetiology of many phenotypes, including unstudied diseases. The phenome-wide association study (PheWAS), a powerful and complementary approach to GWAS, has demonstrated its ability to discover and rediscover genetic associations. The objective of this study is to comprehensively investigate the MHC region by PheWAS to identify new phenotypes mapped to this genetically important region. METHODS In the current study, we systematically explored the MHC region using PheWAS to associate 2692 MHC-linked variants (minor allele frequency ≥0.01) with 6221 phenotypes in a cohort of 7481 subjects from the Marshfield Clinic Personalized Medicine Research Project. RESULTS Findings showed that expected associations previously identified by GWAS could be identified by PheWAS (eg, psoriasis, ankylosing spondylitis, type I diabetes and coeliac disease) with some having strong cross-phenotype associations potentially driven by pleiotropic effects. Importantly, novel associations with eight diseases not previously assessed by GWAS (eg, lichen planus) were also identified and replicated in an independent population. Many of these associated diseases appear to be immune-related disorders. Further assessment of these diseases in 16 484 Marshfield Clinic twins suggests that some of these diseases, including lichen planus, may have genetic aetiologies. CONCLUSIONS These results demonstrate that the PheWAS approach is a powerful and novel method to discover SNP-disease associations, and is ideal when characterising cross-phenotype associations, and further emphasise the importance of the MHC region in human health and disease.
Collapse
Affiliation(s)
- Jixia Liu
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Zhan Ye
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - John G Mayer
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Brian A Hoch
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Clayton Green
- Department of Dermatology, Marshfield Clinic, Marshfield, Wisconsin, USA
| | - Loren Rolak
- Department of Neurology, Marshfield Clinic, Marshfield, Wisconsin, USA
| | - Christopher Cold
- Department of Pathology, Marshfield Clinic, Marshfield, Wisconsin, USA
| | - Seik-Soon Khor
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Xiuwen Zheng
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Taku Miyagawa
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Murray H Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| | - Scott J Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, USA
| |
Collapse
|
663
|
Oetjens MT, Bush WS, Denny JC, Birdwell K, Kodaman N, Verma A, Dilks HH, Pendergrass SA, Ritchie MD, Crawford DC. Evidence for extensive pleiotropy among pharmacogenes. Pharmacogenomics 2016; 17:853-66. [PMID: 27249515 DOI: 10.2217/pgs-2015-0007] [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: 12/12/2022] Open
Abstract
AIM We sought to identify potential pleiotropy involving pharmacogenes. METHODS We tested 184 functional variants in 34 pharmacogenes for associations using a custom grouping of International Classification and Disease, Ninth Revision billing codes extracted from deidentified electronic health records of 6892 patients. RESULTS We replicated several associations including ABCG2 (rs2231142) and gout (p = 1.73 × 10(-7); odds ratio [OR]: 1.73; 95% CI: 1.40-2.12); and SLCO1B1 (rs4149056) and jaundice (p = 2.50 × 10(-4); OR: 1.67; 95% CI: 1.27-2.20). CONCLUSION In this systematic screen for phenotypic associations with functional variants, several novel genotype-phenotype combinations also achieved phenome-wide significance, including SLC15A2 rs1143672 and renal osteodystrophy (p = 2.67 × 10(-) (6); OR: 0.61; 95% CI: 0.49-0.75).
Collapse
Affiliation(s)
- Matthew T Oetjens
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN 37232, USA
| | - William S Bush
- Department of Epidemiology & Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37203, USA
| | - Kelly Birdwell
- Department of Medicine, Vanderbilt University, Nashville, TN 37232, USA
| | - Nuri Kodaman
- Center for Human Genetics Research, Vanderbilt University, Nashville, TN 37232, USA
| | - Anurag Verma
- Center for Systems Genomics, Department of Biochemistry & Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Holli H Dilks
- Sarah Cannon Research Institute, Nashville, TN 37203 USA
| | - Sarah A Pendergrass
- Center for Systems Genomics, Department of Biochemistry & Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Marylyn D Ritchie
- Center for Systems Genomics, Department of Biochemistry & Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dana C Crawford
- Department of Epidemiology & Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
664
|
Han Y, Li L, Zhang Y, Yuan H, Ye L, Zhao J, Duan DD. Phenomics of Vascular Disease: The Systematic Approach to the Combination Therapy. Curr Vasc Pharmacol 2016; 13:433-40. [PMID: 25313004 PMCID: PMC4397150 DOI: 10.2174/1570161112666141014144829] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 02/15/2014] [Accepted: 05/21/2014] [Indexed: 12/28/2022]
Abstract
Vascular diseases are usually caused by multifactorial pathogeneses involving genetic and environmental factors. Our current understanding of vascular disease is, however, based on the focused genotype/phenotype studies driven by the “one-gene/one-phenotype” hypothesis. Drugs with “pure target” at individual molecules involved in the pathophysiological pathways are the mainstream of current clinical treatments and the basis of combination therapy of vascular diseases. Recently, the combination of genomics, proteomics, and metabolomics has unraveled the etiology and pathophysiology of vascular disease in a big-data fashion and also revealed unmatched relationships between the omic variability and the much narrower definition of various clinical phenotypes of vascular disease in individual patients. Here, we introduce the phenomics strategy that will change the conventional focused phenotype/genotype/genome study to a new systematic phenome/genome/proteome approach to the understanding of pathophysiology and combination therapy of vascular disease. A phenome is the sum total of an organism’s phenotypic traits that signify the expression of genome and specific environmental influence. Phenomics is the study of phenome to quantitatively correlate complex traits to variability not only in genome, but also in transcriptome, proteome, metabolome, interactome, and environmental factors by exploring the systems biology that links the genomic and phenomic spaces. The application of phenomics and the phenome-wide associated study (PheWAS) will not only identify a systemically-integrated set of biomarkers for diagnosis and prognosis of vascular disease but also provide novel treatment targets for combination therapy and thus make a revolutionary paradigm shift in the clinical treatment of these devastating diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dayue Darrel Duan
- Laboratory of Cardiovascular Phenomics, Department of Pharmacology, University of Nevada School of Medicine, Center for Molecular Medicine 303F, 1664 N Virginia Street/MS 318, Reno, Nevada 89557-0318, USA.
| |
Collapse
|
665
|
Zhang YP, Zhang YY, Duan DD. From Genome-Wide Association Study to Phenome-Wide Association Study: New Paradigms in Obesity Research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 140:185-231. [PMID: 27288830 DOI: 10.1016/bs.pmbts.2016.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity is a condition in which excess body fat has accumulated over an extent that increases the risk of many chronic diseases. The current clinical classification of obesity is based on measurement of body mass index (BMI), waist-hip ratio, and body fat percentage. However, these measurements do not account for the wide individual variations in fat distribution, degree of fatness or health risks, and genetic variants identified in the genome-wide association studies (GWAS). In this review, we will address this important issue with the introduction of phenome, phenomics, and phenome-wide association study (PheWAS). We will discuss the new paradigm shift from GWAS to PheWAS in obesity research. In the era of precision medicine, phenomics and PheWAS provide the required approaches to better definition and classification of obesity according to the association of obese phenome with their unique molecular makeup, lifestyle, and environmental impact.
Collapse
Affiliation(s)
- Y-P Zhang
- Pediatric Heart Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Y-Y Zhang
- Department of Cardiology, Changzhou Second People's Hospital, Changzhou, Jiangsu, China
| | - D D Duan
- Laboratory of Cardiovascular Phenomics, Center for Cardiovascular Research, Department of Pharmacology, and Center for Molecular Medicine, University of Nevada School of Medicine, Reno, NV, United States.
| |
Collapse
|
666
|
Mowery DL, Chapman BE, Conway M, South BR, Madden E, Keyhani S, Chapman WW. Extracting a stroke phenotype risk factor from Veteran Health Administration clinical reports: an information content analysis. J Biomed Semantics 2016; 7:26. [PMID: 27175226 PMCID: PMC4863379 DOI: 10.1186/s13326-016-0065-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 04/19/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the United States, 795,000 people suffer strokes each year; 10-15 % of these strokes can be attributed to stenosis caused by plaque in the carotid artery, a major stroke phenotype risk factor. Studies comparing treatments for the management of asymptomatic carotid stenosis are challenging for at least two reasons: 1) administrative billing codes (i.e., Current Procedural Terminology (CPT) codes) that identify carotid images do not denote which neurovascular arteries are affected and 2) the majority of the image reports are negative for carotid stenosis. Studies that rely on manual chart abstraction can be labor-intensive, expensive, and time-consuming. Natural Language Processing (NLP) can expedite the process of manual chart abstraction by automatically filtering reports with no/insignificant carotid stenosis findings and flagging reports with significant carotid stenosis findings; thus, potentially reducing effort, costs, and time. METHODS In this pilot study, we conducted an information content analysis of carotid stenosis mentions in terms of their report location (Sections), report formats (structures) and linguistic descriptions (expressions) from Veteran Health Administration free-text reports. We assessed an NLP algorithm, pyConText's, ability to discern reports with significant carotid stenosis findings from reports with no/insignificant carotid stenosis findings given these three document composition factors for two report types: radiology (RAD) and text integration utility (TIU) notes. RESULTS We observed that most carotid mentions are recorded in prose using categorical expressions, within the Findings and Impression sections for RAD reports and within neither of these designated sections for TIU notes. For RAD reports, pyConText performed with high sensitivity (88 %), specificity (84 %), and negative predictive value (95 %) and reasonable positive predictive value (70 %). For TIU notes, pyConText performed with high specificity (87 %) and negative predictive value (92 %), reasonable sensitivity (73 %), and moderate positive predictive value (58 %). pyConText performed with the highest sensitivity processing the full report rather than the Findings or Impressions independently. CONCLUSION We conclude that pyConText can reduce chart review efforts by filtering reports with no/insignificant carotid stenosis findings and flagging reports with significant carotid stenosis findings from the Veteran Health Administration electronic health record, and hence has utility for expediting a comparative effectiveness study of treatment strategies for stroke prevention.
Collapse
Affiliation(s)
- Danielle L. Mowery
- />Department of Biomedical Informatics, University of Utah, Salt Lake City, UT USA
- />IDEAS Center, Veteran Affair Health Care System, Salt Lake City, UT USA
| | - Brian E. Chapman
- />Department of Biomedical Informatics, University of Utah, Salt Lake City, UT USA
- />IDEAS Center, Veteran Affair Health Care System, Salt Lake City, UT USA
| | - Mike Conway
- />Department of Biomedical Informatics, University of Utah, Salt Lake City, UT USA
| | - Brett R. South
- />Department of Biomedical Informatics, University of Utah, Salt Lake City, UT USA
- />IDEAS Center, Veteran Affair Health Care System, Salt Lake City, UT USA
| | - Erin Madden
- />San Francisco Veteran Affair Health Care System, San Francisco, CA USA
| | - Salomeh Keyhani
- />San Francisco Veteran Affair Health Care System, San Francisco, CA USA
| | - Wendy W. Chapman
- />Department of Biomedical Informatics, University of Utah, Salt Lake City, UT USA
- />IDEAS Center, Veteran Affair Health Care System, Salt Lake City, UT USA
| |
Collapse
|
667
|
Claar DD, Larkin EK, Bastarache L, Blackwell TS, Loyd JE, Hartert TV, Denny JC, Kropski JA. A Phenome-Wide Association Study Identifies a Novel Asthma Risk Locus Near TERC. Am J Respir Crit Care Med 2016; 193:98-100. [PMID: 26720789 DOI: 10.1164/rccm.201507-1267le] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Dru D Claar
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | - Emma K Larkin
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | - Lisa Bastarache
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | - Timothy S Blackwell
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and.,2 Department of Veteran Affairs Medical Center Nashville, Tennessee
| | - James E Loyd
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | - Tina V Hartert
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | - Joshua C Denny
- 1 Vanderbilt University School of Medicine Nashville, Tennessee and
| | | |
Collapse
|
668
|
May JC, Gant-Branum RL, McLean JA. Targeting the untargeted in molecular phenomics with structurally-selective ion mobility-mass spectrometry. Curr Opin Biotechnol 2016; 39:192-197. [PMID: 27132126 DOI: 10.1016/j.copbio.2016.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/06/2016] [Accepted: 04/13/2016] [Indexed: 12/25/2022]
Abstract
Systems-wide molecular phenomics is rapidly expanding through technological advances in instrumentation and bioinformatics. Strategies such as structural mass spectrometry, which utilizes size and shape measurements with molecular weight, serve to characterize the sum of molecular expression in biological contexts, where broad-scale measurements are made that are interpreted through big data statistical techniques to reveal underlying patterns corresponding to phenotype. The data density, data dimensionality, data projection, and data interrogation are all critical aspects of these approaches to turn data into salient information. Untargeted molecular phenomics is already having a dramatic impact in discovery science from drug discovery to synthetic biology. It is evident that these emerging techniques will integrate closely in broad efforts aimed at precision medicine.
Collapse
Affiliation(s)
- Jody Christopher May
- Department of Chemistry, Center for Innovative Technology, Vanderbilt Institute of Chemical Biology, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
| | - Randi Lee Gant-Branum
- Department of Chemistry, Center for Innovative Technology, Vanderbilt Institute of Chemical Biology, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
| | - John Allen McLean
- Department of Chemistry, Center for Innovative Technology, Vanderbilt Institute of Chemical Biology, Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
669
|
Biological findings from the PheWAS catalog: focus on connective tissue-related disorders (pelvic floor dysfunction, abdominal hernia, varicose veins and hemorrhoids). Hum Genet 2016; 135:779-95. [DOI: 10.1007/s00439-016-1672-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/17/2016] [Indexed: 01/31/2023]
|
670
|
Integrative genomics analyses unveil downstream biological effectors of disease-specific polymorphisms buried in intergenic regions. NPJ Genom Med 2016; 1. [PMID: 27482468 PMCID: PMC4966659 DOI: 10.1038/npjgenmed.2016.6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Functionally altered biological mechanisms arising from disease-associated polymorphisms, remain difficult to characterise when those variants are intergenic, or, fall between genes. We sought to identify shared downstream mechanisms by which inter- and intragenic single-nucleotide polymorphisms (SNPs) contribute to a specific physiopathology. Using computational modelling of 2 million pairs of disease-associated SNPs drawn from genome-wide association studies (GWAS), integrated with expression Quantitative Trait Loci (eQTL) and Gene Ontology functional annotations, we predicted 3,870 inter–intra and inter–intra SNP pairs with convergent biological mechanisms (FDR<0.05). These prioritised SNP pairs with overlapping messenger RNA targets or similar functional annotations were more likely to be associated with the same disease than unrelated pathologies (OR>12). We additionally confirmed synergistic and antagonistic genetic interactions for a subset of prioritised SNP pairs in independent studies of Alzheimer’s disease (entropy P=0.046), bladder cancer (entropy P=0.039), and rheumatoid arthritis (PheWAS case–control P<10−4). Using ENCODE data sets, we further statistically validated that the biological mechanisms shared within prioritised SNP pairs are frequently governed by matching transcription factor binding sites and long-range chromatin interactions. These results provide a ‘roadmap’ of disease mechanisms emerging from GWAS and further identify candidate therapeutic targets among downstream effectors of intergenic SNPs.
Collapse
|
671
|
Mosley JD, Witte JS, Larkin EK, Bastarache L, Shaffer CM, Karnes JH, Stein CM, Phillips E, Hebbring SJ, Brilliant MH, Mayer J, Ye Z, Roden DM, Denny JC. Identifying genetically driven clinical phenotypes using linear mixed models. Nat Commun 2016; 7:11433. [PMID: 27109359 PMCID: PMC4848547 DOI: 10.1038/ncomms11433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 03/24/2016] [Indexed: 01/06/2023] Open
Abstract
We hypothesized that generalized linear mixed models (GLMMs), which estimate the additive genetic variance underlying phenotype variability, would facilitate rapid characterization of clinical phenotypes from an electronic health record. We evaluated 1,288 phenotypes in 29,349 subjects of European ancestry with single-nucleotide polymorphism (SNP) genotyping on the Illumina Exome Beadchip. We show that genetic liability estimates are primarily driven by SNPs identified by prior genome-wide association studies and SNPs within the human leukocyte antigen (HLA) region. We identify 44 (false discovery rate q<0.05) phenotypes associated with HLA SNP variation and show that hypothyroidism is genetically correlated with Type I diabetes (rG=0.31, s.e. 0.12, P=0.003). We also report novel SNP associations for hypothyroidism near HLA-DQA1/HLA-DQB1 at rs6906021 (combined odds ratio (OR)=1.2 (95% confidence interval (CI): 1.1-1.2), P=9.8 × 10(-11)) and for polymyalgia rheumatica near C6orf10 at rs6910071 (OR=1.5 (95% CI: 1.3-1.6), P=1.3 × 10(-10)). Phenome-wide application of GLMMs identifies phenotypes with important genetic drivers, and focusing on these phenotypes can identify novel genetic associations.
Collapse
Affiliation(s)
- Jonathan D. Mosley
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - John S. Witte
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California 94158, USA
| | - Emma K. Larkin
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Lisa Bastarache
- Biomedical Informatics, Vanderbilt University, Nashville, Tennessee 37203, USA
| | | | - Jason H. Karnes
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - C. Michael Stein
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Elizabeth Phillips
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Scott J. Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - Murray H. Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - John Mayer
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - Zhan Ye
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, Wisconsin 54449, USA
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Joshua C. Denny
- Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
- Biomedical Informatics, Vanderbilt University, Nashville, Tennessee 37203, USA
| |
Collapse
|
672
|
Cheng F, Zhao J, Fooksa M, Zhao Z. A network-based drug repositioning infrastructure for precision cancer medicine through targeting significantly mutated genes in the human cancer genomes. J Am Med Inform Assoc 2016; 23:681-91. [PMID: 27026610 DOI: 10.1093/jamia/ocw007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 01/13/2016] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE Development of computational approaches and tools to effectively integrate multidomain data is urgently needed for the development of newly targeted cancer therapeutics. METHODS We proposed an integrative network-based infrastructure to identify new druggable targets and anticancer indications for existing drugs through targeting significantly mutated genes (SMGs) discovered in the human cancer genomes. The underlying assumption is that a drug would have a high potential for anticancer indication if its up-/down-regulated genes from the Connectivity Map tended to be SMGs or their neighbors in the human protein interaction network. RESULTS We assembled and curated 693 SMGs in 29 cancer types and found 121 proteins currently targeted by known anticancer or noncancer (repurposed) drugs. We found that the approved or experimental cancer drugs could potentially target these SMGs in 33.3% of the mutated cancer samples, and this number increased to 68.0% by drug repositioning through surveying exome-sequencing data in approximately 5000 normal-tumor pairs from The Cancer Genome Atlas. Furthermore, we identified 284 potential new indications connecting 28 cancer types and 48 existing drugs (adjusted P < .05), with a 66.7% success rate validated by literature data. Several existing drugs (e.g., niclosamide, valproic acid, captopril, and resveratrol) were predicted to have potential indications for multiple cancer types. Finally, we used integrative analysis to showcase a potential mechanism-of-action for resveratrol in breast and lung cancer treatment whereby it targets several SMGs (ARNTL, ASPM, CTTN, EIF4G1, FOXP1, and STIP1). CONCLUSIONS In summary, we demonstrated that our integrative network-based infrastructure is a promising strategy to identify potential druggable targets and uncover new indications for existing drugs to speed up molecularly targeted cancer therapeutics.
Collapse
Affiliation(s)
- Feixiong Cheng
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Junfei Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA
| | - Michaela Fooksa
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA Chemical and Physical Biology Program, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Zhongming Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37203, USA Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA Department of Psychiatry, Vanderbilt University School of Medicine, Nashville, TN 37212, USA Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
673
|
Nissim N, Boland MR, Tatonetti NP, Elovici Y, Hripcsak G, Shahar Y, Moskovitch R. Improving condition severity classification with an efficient active learning based framework. J Biomed Inform 2016; 61:44-54. [PMID: 27016383 DOI: 10.1016/j.jbi.2016.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/31/2016] [Accepted: 03/21/2016] [Indexed: 02/07/2023]
Abstract
Classification of condition severity can be useful for discriminating among sets of conditions or phenotypes, for example when prioritizing patient care or for other healthcare purposes. Electronic Health Records (EHRs) represent a rich source of labeled information that can be harnessed for severity classification. The labeling of EHRs is expensive and in many cases requires employing professionals with high level of expertise. In this study, we demonstrate the use of Active Learning (AL) techniques to decrease expert labeling efforts. We employ three AL methods and demonstrate their ability to reduce labeling efforts while effectively discriminating condition severity. We incorporate three AL methods into a new framework based on the original CAESAR (Classification Approach for Extracting Severity Automatically from Electronic Health Records) framework to create the Active Learning Enhancement framework (CAESAR-ALE). We applied CAESAR-ALE to a dataset containing 516 conditions of varying severity levels that were manually labeled by seven experts. Our dataset, called the "CAESAR dataset," was created from the medical records of 1.9 million patients treated at Columbia University Medical Center (CUMC). All three AL methods decreased labelers' efforts compared to the learning methods applied by the original CAESER framework in which the classifier was trained on the entire set of conditions; depending on the AL strategy used in the current study, the reduction ranged from 48% to 64% that can result in significant savings, both in time and money. As for the PPV (precision) measure, CAESAR-ALE achieved more than 13% absolute improvement in the predictive capabilities of the framework when classifying conditions as severe. These results demonstrate the potential of AL methods to decrease the labeling efforts of medical experts, while increasing accuracy given the same (or even a smaller) number of acquired conditions. We also demonstrated that the methods included in the CAESAR-ALE framework (Exploitation and Combination_XA) are more robust to the use of human labelers with different levels of professional expertise.
Collapse
Affiliation(s)
- Nir Nissim
- Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel; Malware Lab, Cyber Security Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | - Mary Regina Boland
- Department of Biomedical Informatics, Columbia University, New York, NY, USA; Observational Health Data Sciences and Informatics, Columbia University, New York, NY, USA
| | - Nicholas P Tatonetti
- Department of Biomedical Informatics, Columbia University, New York, NY, USA; Department of Systems Biology, Columbia University, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA; Observational Health Data Sciences and Informatics, Columbia University, New York, NY, USA
| | - Yuval Elovici
- Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, NY, USA; Observational Health Data Sciences and Informatics, Columbia University, New York, NY, USA
| | - Yuval Shahar
- Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Robert Moskovitch
- Department of Biomedical Informatics, Columbia University, New York, NY, USA; Department of Systems Biology, Columbia University, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA; Observational Health Data Sciences and Informatics, Columbia University, New York, NY, USA.
| |
Collapse
|
674
|
Hall JL, Ryan JJ, Bray BE, Brown C, Lanfear D, Newby LK, Relling MV, Risch NJ, Roden DM, Shaw SY, Tcheng JE, Tenenbaum J, Wang TN, Weintraub WS. Merging Electronic Health Record Data and Genomics for Cardiovascular Research: A Science Advisory From the American Heart Association. ACTA ACUST UNITED AC 2016; 9:193-202. [PMID: 26976545 DOI: 10.1161/hcg.0000000000000029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The process of scientific discovery is rapidly evolving. The funding climate has influenced a favorable shift in scientific discovery toward the use of existing resources such as the electronic health record. The electronic health record enables long-term outlooks on human health and disease, in conjunction with multidimensional phenotypes that include laboratory data, images, vital signs, and other clinical information. Initial work has confirmed the utility of the electronic health record for understanding mechanisms and patterns of variability in disease susceptibility, disease evolution, and drug responses. The addition of biobanks and genomic data to the information contained in the electronic health record has been demonstrated. The purpose of this statement is to discuss the current challenges in and the potential for merging electronic health record data and genomics for cardiovascular research.
Collapse
|
675
|
Simonti CN, Vernot B, Bastarache L, Bottinger E, Carrell DS, Chisholm RL, Crosslin DR, Hebbring SJ, Jarvik GP, Kullo IJ, Li R, Pathak J, Ritchie MD, Roden DM, Verma SS, Tromp G, Prato JD, Bush WS, Akey JM, Denny JC, Capra JA. The phenotypic legacy of admixture between modern humans and Neandertals. Science 2016; 351:737-41. [PMID: 26912863 DOI: 10.1126/science.aad2149] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many modern human genomes retain DNA inherited from interbreeding with archaic hominins, such as Neandertals, yet the influence of this admixture on human traits is largely unknown. We analyzed the contribution of common Neandertal variants to over 1000 electronic health record (EHR)-derived phenotypes in ~28,000 adults of European ancestry. We discovered and replicated associations of Neandertal alleles with neurological, psychiatric, immunological, and dermatological phenotypes. Neandertal alleles together explained a significant fraction of the variation in risk for depression and skin lesions resulting from sun exposure (actinic keratosis), and individual Neandertal alleles were significantly associated with specific human phenotypes, including hypercoagulation and tobacco use. Our results establish that archaic admixture influences disease risk in modern humans, provide hypotheses about the effects of hundreds of Neandertal haplotypes, and demonstrate the utility of EHR data in evolutionary analyses.
Collapse
Affiliation(s)
- Corinne N Simonti
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Benjamin Vernot
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | | | - David S Carrell
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
| | - Rex L Chisholm
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David R Crosslin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA. Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
| | - Scott J Hebbring
- Center for Human Genetics, Marshfield Clinic, Marshfield, WI, USA
| | - Gail P Jarvik
- Department of Genome Sciences, University of Washington, Seattle, WA, USA. Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
| | - Iftikhar J Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Rongling Li
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jyotishman Pathak
- Division of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Marylyn D Ritchie
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA. Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Dan M Roden
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA. Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA. Department of Medicine, Vanderbilt University, Nashville, TN, USA. Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Shefali S Verma
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Gerard Tromp
- Weis Center for Research, Geisinger Health System, Danville, PA, USA. Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Health Science, Stellenbosch University, Tygerberg, South Africa
| | - Jeffrey D Prato
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - William S Bush
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA
| | - Joshua M Akey
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Joshua C Denny
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA. Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA. Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - John A Capra
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA. Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA. Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA. Center for Quantitative Sciences, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
676
|
Roden DM, Denny JC. Integrating electronic health record genotype and phenotype datasets to transform patient care. Clin Pharmacol Ther 2016; 99:298-305. [PMID: 26667791 PMCID: PMC4760864 DOI: 10.1002/cpt.321] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 12/16/2022]
Abstract
The Health Information Technology for Economic and Clinical Health (HITECH) Act of 2009 mandates the development and implementation of electronic health record (EHR) systems across the country. While a primary goal is to improve the care of individual patients, EHRs are also key enabling resources for a vision of individualized (or personalized or precision) medicine: the aggregation of multiple EHRs within or across healthcare systems should allow discovery of patient subsets that have unusual and definable clinical trajectories that deviate importantly from the expected response in a "typical" patient. The spectrum of such personalized care can then extend from prevention to choice of medication to intensity or nature of follow-up.
Collapse
Affiliation(s)
- D M Roden
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - J C Denny
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
677
|
Unravelling the human genome-phenome relationship using phenome-wide association studies. Nat Rev Genet 2016; 17:129-45. [PMID: 26875678 DOI: 10.1038/nrg.2015.36] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advances in genotyping technology have, over the past decade, enabled the focused search for common genetic variation associated with human diseases and traits. With the recently increased availability of detailed phenotypic data from electronic health records and epidemiological studies, the impact of one or more genetic variants on the phenome is starting to be characterized both in clinical and population-based settings using phenome-wide association studies (PheWAS). These studies reveal a number of challenges that will need to be overcome to unlock the full potential of PheWAS for the characterization of the complex human genome-phenome relationship.
Collapse
|
678
|
Wang X, Pandey AK, Mulligan MK, Williams EG, Mozhui K, Li Z, Jovaisaite V, Quarles LD, Xiao Z, Huang J, Capra JA, Chen Z, Taylor WL, Bastarache L, Niu X, Pollard KS, Ciobanu DC, Reznik AO, Tishkov AV, Zhulin IB, Peng J, Nelson SF, Denny JC, Auwerx J, Lu L, Williams RW. Joint mouse-human phenome-wide association to test gene function and disease risk. Nat Commun 2016; 7:10464. [PMID: 26833085 PMCID: PMC4740880 DOI: 10.1038/ncomms10464] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 12/11/2015] [Indexed: 01/22/2023] Open
Abstract
Phenome-wide association is a novel reverse genetic strategy to analyze genome-to-phenome relations in human clinical cohorts. Here we test this approach using a large murine population segregating for ∼5 million sequence variants, and we compare our results to those extracted from a matched analysis of gene variants in a large human cohort. For the mouse cohort, we amassed a deep and broad open-access phenome consisting of ∼4,500 metabolic, physiological, pharmacological and behavioural traits, and more than 90 independent expression quantitative trait locus (QTL), transcriptome, proteome, metagenome and metabolome data sets—by far the largest coherent phenome for any experimental cohort (www.genenetwork.org). We tested downstream effects of subsets of variants and discovered several novel associations, including a missense mutation in fumarate hydratase that controls variation in the mitochondrial unfolded protein response in both mouse and Caenorhabditis elegans, and missense mutations in Col6a5 that underlies variation in bone mineral density in both mouse and human. Phenome-wide association is a novel method that links sequence variants to a spectrum of phenotypes and diseases. Here the authors generate detailed mouse genetic and phenome data which links their phenome-wide association study (PheWAS) of mouse to corresponding PheWAS in human.
Collapse
Affiliation(s)
- Xusheng Wang
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.,St Jude Proteomics Facility, St Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Ashutosh K Pandey
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Megan K Mulligan
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Evan G Williams
- Laboratory of Integrative and Systems Physiology, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Khyobeni Mozhui
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Zhengsheng Li
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Virginija Jovaisaite
- Laboratory of Integrative and Systems Physiology, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Jinsong Huang
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.,Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - John A Capra
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Zugen Chen
- Department of Human Genetics, University of California, Los Angeles, California 90095, USA
| | - William L Taylor
- Molecular Resource Center, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Xinnan Niu
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, California 94158, USA.,Division of Biostatistics and Institute for Human Genetics, University of California, San Francisco, California 94158, USA
| | - Daniel C Ciobanu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.,Animal Science Department, University of Nebraska, Lincoln, Nebraska 68583, USA
| | - Alexander O Reznik
- Joint Institute for Computational Sciences, University of Tennessee-Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, USA
| | - Artem V Tishkov
- Joint Institute for Computational Sciences, University of Tennessee-Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, USA
| | - Igor B Zhulin
- Joint Institute for Computational Sciences, University of Tennessee-Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, USA
| | - Junmin Peng
- St Jude Proteomics Facility, St Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Stanley F Nelson
- Department of Human Genetics, University of California, Los Angeles, California 90095, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, USA
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| |
Collapse
|
679
|
Chen R, Sun J, Dittus RS, Fabbri D, Kirby J, Laffer CL, McNaughton CD, Malin B. Patient Stratification Using Electronic Health Records from a Chronic Disease Management Program. IEEE J Biomed Health Inform 2016:10.1109/JBHI.2016.2514264. [PMID: 26742152 PMCID: PMC4931988 DOI: 10.1109/jbhi.2016.2514264] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The goal of this study is to devise a machine learning framework to assist care coordination programs in prognostic stratification to design and deliver personalized care plans and to allocate financial and medical resources effectively. MATERIALS AND METHODS This study is based on a de-identified cohort of 2,521 hypertension patients from a chronic care coordination program at the Vanderbilt University Medical Center. Patients were modeled as vectors of features derived from electronic health records (EHRs) over a six-year period. We applied a stepwise regression to identify risk factors associated with a decrease in mean arterial pressure of at least 2 mmHg after program enrollment. The resulting features were subsequently validated via a logistic regression classifier. Finally, risk factors were applied to group the patients through model-based clustering. RESULTS We identified a set of predictive features that consisted of a mix of demographic, medication, and diagnostic concepts. Logistic regression over these features yielded an area under the ROC curve (AUC) of 0.71 (95% CI: [0.67, 0.76]). Based on these features, four clinically meaningful groups are identified through clustering - two of which represented patients with more severe disease profiles, while the remaining represented patients with mild disease profiles. DISCUSSION Patients with hypertension can exhibit significant variation in their blood pressure control status and responsiveness to therapy. Yet this work shows that a clustering analysis can generate more homogeneous patient groups, which may aid clinicians in designing and implementing customized care programs. CONCLUSION The study shows that predictive modeling and clustering using EHR data can be beneficial for providing a systematic, generalized approach for care providers to tailor their management approach based upon patient-level factors.
Collapse
Affiliation(s)
- Robert Chen
- School of Computational Science and Engineering at the Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Jimeng Sun
- School of Computational Science and Engineering at the Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Robert S. Dittus
- Institute for Medicine and Public Health, Vanderbilt University, Nashville, TN, the Geriatric Research, Education, and Clinical Center, VA Tennessee Valley Healthcare System, Nashville, TN, and the Department of Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Daniel Fabbri
- Department of Biomedical Informatics, School of Medicine, Vanderbilt University, Nashville, TN, and the Department of Electrical Engineering and Computer Science, School of Engineering, Vanderbilt University, Nashville, TN
| | - Jacqueline Kirby
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University, Nashville, TN
| | - Cheryl L. Laffer
- Department of Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Candace D. McNaughton
- Department of Emergency Medicine, School of Medicine, Vanderbilt University, Nashville, TN
| | - Bradley Malin
- Department of Biomedical Informatics, School of Medicine, Vanderbilt University, Nashville, TN, and the Department of Electrical Engineering and Computer Science, School of Engineering, Vanderbilt University, Nashville, TN
| |
Collapse
|
680
|
Verma A, Leader JB, Verma SS, Frase A, Wallace J, Dudek S, Lavage DR, Van Hout CV, Dewey FE, Penn J, Lopez A, Overton JD, Carey DJ, Ledbetter DH, Kirchner HL, Ritchie MD, Pendergrass SA. INTEGRATING CLINICAL LABORATORY MEASURES AND ICD-9 CODE DIAGNOSES IN PHENOME-WIDE ASSOCIATION STUDIES. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2016; 21:168-79. [PMID: 26776183 PMCID: PMC4718547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Electronic health records (EHR) provide a comprehensive resource for discovery, allowing unprecedented exploration of the impact of genetic architecture on health and disease. The data of EHRs also allow for exploration of the complex interactions between health measures across health and disease. The discoveries arising from EHR based research provide important information for the identification of genetic variation for clinical decision-making. Due to the breadth of information collected within the EHR, a challenge for discovery using EHR based data is the development of high-throughput tools that expose important areas of further research, from genetic variants to phenotypes. Phenome-Wide Association studies (PheWAS) provide a way to explore the association between genetic variants and comprehensive phenotypic measurements, generating new hypotheses and also exposing the complex relationships between genetic architecture and outcomes, including pleiotropy. EHR based PheWAS have mainly evaluated associations with case/control status from International Classification of Disease, Ninth Edition (ICD-9) codes. While these studies have highlighted discovery through PheWAS, the rich resource of clinical lab measures collected within the EHR can be better utilized for high-throughput PheWAS analyses and discovery. To better use these resources and enrich PheWAS association results we have developed a sound methodology for extracting a wide range of clinical lab measures from EHR data. We have extracted a first set of 21 clinical lab measures from the de-identified EHR of participants of the Geisinger MyCodeTM biorepository, and calculated the median of these lab measures for 12,039 subjects. Next we evaluated the association between these 21 clinical lab median values and 635,525 genetic variants, performing a genome-wide association study (GWAS) for each of 21 clinical lab measures. We then calculated the association between SNPs from these GWAS passing our Bonferroni defined p-value cutoff and 165 ICD-9 codes. Through the GWAS we found a series of results replicating known associations, and also some potentially novel associations with less studied clinical lab measures. We found the majority of the PheWAS ICD-9 diagnoses highly related to the clinical lab measures associated with same SNPs. Moving forward, we will be evaluating further phenotypes and expanding the methodology for successful extraction of clinical lab measurements for research and PheWAS use. These developments are important for expanding the PheWAS approach for improved EHR based discovery.
Collapse
Affiliation(s)
- Anurag Verma
- Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA3Center for Systems Genomics, The Pennsylvania State University, University Park, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
681
|
MORGAN ALEXANDERA, MOONEY SEAND, ARONOW BRUCEJ, BRENNER STEVENE. PRECISION MEDICINE: DATA AND DISCOVERY FOR IMPROVED HEALTH AND THERAPY. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2016; 21:243-8. [PMID: 26776190 PMCID: PMC5180448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Rapid advances in personal, cohort, and population-scale data acquisition, such as via sequencing, proteomics, mass spectroscopy, biosensors, mobile health devices and social network activity and other apps are opening up new vistas for personalized health biomedical data collection, analysis and insight. To achieve the vaunted goals of precision medicine and go from measurement to clinical translation, substantial gains still need to be made in methods of data and knowledge integration, analysis, discovery and interpretation. In this session of the 2016 Pacific Symposium on Biocomputing, we present sixteen papers to help accomplish this for precision medicine.
Collapse
Affiliation(s)
- ALEXANDER A. MORGAN
- Stanford University School of Medicine & Khosla Ventures, Stanford, CA, USA,
| | - SEAN D. MOONEY
- Department of Biomedical Informatics and Medical Education, Seattle, WA 98105, USA,
| | - BRUCE J. ARONOW
- Biomedical Informatics, Developmental Biology and Computer Science, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA,
| | - STEVEN E. BRENNER
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA, USA,
| |
Collapse
|
682
|
Abstract
Systems medicine promotes a range of approaches and strategies to study human health and disease at a systems level with the aim of improving the overall well-being of (healthy) individuals, and preventing, diagnosing, or curing disease. In this chapter we discuss how bioinformatics critically contributes to systems medicine. First, we explain the role of bioinformatics in the management and analysis of data. In particular we show the importance of publicly available biological and clinical repositories to support systems medicine studies. Second, we discuss how the integration and analysis of multiple types of omics data through integrative bioinformatics may facilitate the determination of more predictive and robust disease signatures, lead to a better understanding of (patho)physiological molecular mechanisms, and facilitate personalized medicine. Third, we focus on network analysis and discuss how gene networks can be constructed from omics data and how these networks can be decomposed into smaller modules. We discuss how the resulting modules can be used to generate experimentally testable hypotheses, provide insight into disease mechanisms, and lead to predictive models. Throughout, we provide several examples demonstrating how bioinformatics contributes to systems medicine and discuss future challenges in bioinformatics that need to be addressed to enable the advancement of systems medicine.
Collapse
Affiliation(s)
- Ulf Schmitz
- Dept of Systems Biology & Bioinformatics, University of Rostock, Rostock, Germany
| | - Olaf Wolkenhauer
- Dept of Systems Biology & Bioinformatics, University of Rostock, Rostock, Germany
| |
Collapse
|
683
|
Glueck M, Hamilton P, Chevalier F, Breslav S, Khan A, Wigdor D, Brudno M. PhenoBlocks: Phenotype Comparison Visualizations. IEEE TRANSACTIONS ON VISUALIZATION AND COMPUTER GRAPHICS 2016; 22:101-110. [PMID: 26529691 DOI: 10.1109/tvcg.2015.2467733] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The differential diagnosis of hereditary disorders is a challenging task for clinicians due to the heterogeneity of phenotypes that can be observed in patients. Existing clinical tools are often text-based and do not emphasize consistency, completeness, or granularity of phenotype reporting. This can impede clinical diagnosis and limit their utility to genetics researchers. Herein, we present PhenoBlocks, a novel visual analytics tool that supports the comparison of phenotypes between patients, or between a patient and the hallmark features of a disorder. An informal evaluation of PhenoBlocks with expert clinicians suggested that the visualization effectively guides the process of differential diagnosis and could reinforce the importance of complete, granular phenotypic reporting.
Collapse
|
684
|
Rouchka EC, Chariker JH, Harrison BJ. Proceedings of the Fourteenth Annual UT- KBRIN Bioinformatics Summit 2015. BMC Bioinformatics 2015; 16 Suppl 15:I1-P21. [PMID: 26510995 PMCID: PMC4625115 DOI: 10.1186/1471-2105-16-s15-i1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
685
|
Lu Y, Liu Y, Niu X, Yang Q, Hu X, Zhang HY, Xia J. Systems Genetic Validation of the SNP-Metabolite Association in Rice Via Metabolite-Pathway-Based Phenome-Wide Association Scans. FRONTIERS IN PLANT SCIENCE 2015; 6:1027. [PMID: 26640468 PMCID: PMC4661230 DOI: 10.3389/fpls.2015.01027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/05/2015] [Indexed: 06/05/2023]
Abstract
In the post-GWAS (Genome-Wide Association Scan) era, the interpretation of GWAS results is crucial to screen for highly relevant phenotype-genotype association pairs. Based on the single genotype-phenotype association test and a pathway enrichment analysis, we propose a Metabolite-pathway-based Phenome-Wide Association Scan (M-PheWAS) to analyze the key metabolite-SNP pairs in rice and determine the regulatory relationship by assessing similarities in the changes of enzymes and downstream products in a pathway. Two SNPs, sf0315305925 and sf0315308337, were selected using this approach, and their molecular function and regulatory relationship with Enzyme EC:5.5.1.6 and with flavonoids, a significant downstream regulatory metabolite product, were demonstrated. Moreover, a total of 105 crucial SNPs were screened using M-PheWAS, which may be important for metabolite associations.
Collapse
|
686
|
Dumitrescu L, Restrepo NA, Goodloe R, Boston J, Farber-Eger E, Pendergrass SA, Bush WS, Crawford DC. Towards a phenome-wide catalog of human clinical traits impacted by genetic ancestry. BioData Min 2015; 8:35. [PMID: 26566401 PMCID: PMC4642611 DOI: 10.1186/s13040-015-0068-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 11/02/2015] [Indexed: 01/13/2023] Open
Abstract
Background Racial/ethnic differences for commonly measured clinical variables are well documented, and it has been postulated that population-specific genetic factors may play a role. The genetic heterogeneity of admixed populations, such as African Americans, provides a unique opportunity to identify genomic regions and variants associated with the clinical variability observed for diseases and traits across populations. Method To begin a systematic search for these population-specific genomic regions at the phenome-wide scale, we determined the relationship between global genetic ancestry, specifically European and African ancestry, and clinical variables measured in a population of African Americans from BioVU, Vanderbilt University’s biorepository linked to de-identified electronic medical records (EMRs) as part of the Epidemiologic Architecture using Genomics and Epidemiology (EAGLE) study. Through billing (ICD-9) codes, procedure codes, labs, and clinical notes, 36 common clinical and laboratory variables were mined from the EMR, including body mass index (BMI), kidney traits, lipid levels, blood pressure, and electrocardiographic measurements. A total of 15,863 DNA samples from non-European Americans were genotyped on the Illumina Metabochip containing ~200,000 variants, of which 11,166 were from African Americans. Tests of association were performed to examine associations between global ancestry and the phenotype of interest. Results Increased European ancestry, and conversely decreased African ancestry, was most strongly correlated with an increase in QRS duration, consistent with previous observations that African Americans tend to have shorter a QRS duration compared with European Americans. Despite known racial/ethnic disparities in blood pressure, European and African ancestry was neither associated with diastolic nor systolic blood pressure measurements. Conclusion Collectively, these results suggest that this clinical population can be used to identify traits in which population differences may be due, in part, to population-specific genetics. Electronic supplementary material The online version of this article (doi:10.1186/s13040-015-0068-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Logan Dumitrescu
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Nicole A Restrepo
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Robert Goodloe
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Jonathan Boston
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Eric Farber-Eger
- Center for Human Genetics Research, Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232 USA
| | - Sarah A Pendergrass
- Center for Systems Genomics, Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802 USA
| | - William S Bush
- Institute for Computational Biology, Department of Epidemiology and Biostatistics, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Suite 2527, Cleveland, OH 44106 USA
| | - Dana C Crawford
- Institute for Computational Biology, Department of Epidemiology and Biostatistics, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road, Suite 2527, Cleveland, OH 44106 USA
| |
Collapse
|
687
|
Leader JB, Pendergrass SA, Verma A, Carey DJ, Hartzel DN, Ritchie MD, Kirchner HL. Contrasting Association Results between Existing PheWAS Phenotype Definition Methods and Five Validated Electronic Phenotypes. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2015; 2015:824-32. [PMID: 26958218 PMCID: PMC4765620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Phenome-Wide Association Studies (PheWAS) comprehensively investigate the association between genetic variation and a wide array of outcome traits. Electronic health record (EHR) based PheWAS uses various abstractions of International Classification of Diseases, Ninth Revision (ICD-9) codes to identify case/control status for diagnoses that are used as the phenotypic variables. However, there have not been comparisons within a PheWAS between results from high quality derived phenotypes and high-throughput but potentially inaccurate use of ICD-9 codes for case/control definition. For this study we first developed a group of high quality algorithms for five phenotypes. Next we evaluated the association of these "gold standard" phenotypes and 4,636,178 genetic variants with minor allele frequency > 0.01 and compared the results from high-throughput associations at the 3 digit, 5 digit, and PheWAS codes for defining case/control status. We found that certain diseases contained similar patient populations across phenotyping methods but had differences in PheWAS.
Collapse
Affiliation(s)
| | | | - Anurag Verma
- Biomedical and Translational Informatics Program, Danville, PA, USA; The Center for Systems Genomics, The Pennsylvania State University, University Park, PA USA
| | - David J Carey
- Weis Center for Research, Geisinger Health System, Danville, PA, USA
| | | | - Marylyn D Ritchie
- Biomedical and Translational Informatics Program, Danville, PA, USA; The Center for Systems Genomics, The Pennsylvania State University, University Park, PA USA
| | | |
Collapse
|
688
|
Chen ES, Melton GB, Wasserman RC, Rosenau PT, Howard DB, Sarkar IN. Mining and Visualizing Family History Associations in the Electronic Health Record: A Case Study for Pediatric Asthma. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2015; 2015:396-405. [PMID: 26958171 PMCID: PMC4765567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Asthma is the most common chronic childhood disease and has seen increasing prevalence worldwide. While there is existing evidence of familial and other risk factors for pediatric asthma, there is a need for further studies to explore and understand interactions among these risk factors. The goal of this study was to develop an approach for mining, visualizing, and evaluating association rules representing pairwise interactions among potential familial risk factors based on information documented as part of a patient's family history in the electronic health record. As a case study, 10,260 structured family history entries for a cohort of 1,531 pediatric asthma patients were extracted and analyzed to generate family history associations at different levels of granularity. The preliminary results highlight the potential of this approach for validating known knowledge and suggesting opportunities for further investigation that may contribute to improving prediction of asthma risk in children.
Collapse
Affiliation(s)
- Elizabeth S Chen
- Center for Biomedical Informatics, Warren Alpert Medical School of Brown University, Providence, RI; Center for Clinical and Translational Science, University of Vermont, Burlington, VT
| | - Genevieve B Melton
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN; Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Richard C Wasserman
- Department of Pediatrics, University of Vermont, Burlington, VT; University of Vermont Children's Hospital, Burlington, VT
| | - Paul T Rosenau
- Department of Pediatrics, University of Vermont, Burlington, VT; University of Vermont Children's Hospital, Burlington, VT
| | - Diantha B Howard
- Center for Clinical and Translational Science, University of Vermont, Burlington, VT
| | - Indra Neil Sarkar
- Center for Biomedical Informatics, Warren Alpert Medical School of Brown University, Providence, RI; Center for Clinical and Translational Science, University of Vermont, Burlington, VT
| |
Collapse
|
689
|
Seldin MF. The genetics of human autoimmune disease: A perspective on progress in the field and future directions. J Autoimmun 2015; 64:1-12. [PMID: 26343334 PMCID: PMC4628839 DOI: 10.1016/j.jaut.2015.08.015] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/23/2015] [Indexed: 12/18/2022]
Abstract
Progress in defining the genetics of autoimmune disease has been dramatically enhanced by large scale genetic studies. Genome-wide approaches, examining hundreds or for some diseases thousands of cases and controls, have been implemented using high throughput genotyping and appropriate algorithms to provide a wealth of data over the last decade. These studies have identified hundreds of non-HLA loci as well as further defining HLA variations that predispose to different autoimmune diseases. These studies to identify genetic risk loci are also complemented by progress in gene expression studies including definition of expression quantitative trait loci (eQTL), various alterations in chromatin structure including histone marks, DNase I sensitivity, repressed chromatin regions as well as transcript factor binding sites. Integration of this information can partially explain why particular variations can alter proclivity to autoimmune phenotypes. Despite our incomplete knowledge base with only partial definition of hereditary factors and possible functional connections, this progress has and will continue to facilitate a better understanding of critical pathways and critical changes in immunoregulation. Advances in defining and understanding functional variants potentially can lead to both novel therapeutics and personalized medicine in which therapeutic approaches are chosen based on particular molecular phenotypes and genomic alterations.
Collapse
Affiliation(s)
- Michael F Seldin
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Tupper Hall Room 4453, Davis, CA 95616, USA; Division of Rheumatology and Allergy, Department of Medicine, University of California, Davis, Tupper Hall Room 4453, Davis, CA 95616, USA.
| |
Collapse
|
690
|
Abstract
BACKGROUND The accuracy and utility of electronic health record (EHR)-derived phenotypes in replicating genotype-phenotype relationships have been infrequently examined. Low circulating vitamin D levels are associated with severe outcomes in inflammatory bowel disease (IBD); however, the genetic basis for vitamin D insufficiency in this population has not been examined previously. METHODS We compared the accuracy of physician-assigned phenotypes in a large prospective IBD registry to that identified by an EHR algorithm incorporating codified and structured data. Genotyping for IBD risk alleles was performed on the Immunochip and a genetic risk score calculated and compared between EHR-defined patients and those in the registry. Additionally, 4 vitamin D risk alleles were genotyped and serum 25-hydroxy vitamin D [25(OH)D] levels compared across genotypes. RESULTS A total of 1131 patients captured by our EHR algorithm were also included in our prospective registry (656 Crohn's disease, 475 ulcerative colitis). The overall genetic risk score for Crohn's disease (P = 0.13) and ulcerative colitis (P = 0.32) was similar between EHR-defined patients and a prospective registry. Three of the 4 vitamin D risk alleles were associated with low vitamin D levels in patients with IBD and contributed an additional 3% of the variance explained. Vitamin D genetic risk score did not predict normalization of vitamin D levels. CONCLUSIONS EHR cohorts form valuable data sources for examining genotype-phenotype relationships. Vitamin D risk alleles explain 3% of the variance in vitamin D levels in patients with IBD.
Collapse
|
691
|
Li YR, Zhao SD, Li J, Bradfield JP, Mohebnasab M, Steel L, Kobie J, Abrams DJ, Mentch FD, Glessner JT, Guo Y, Wei Z, Connolly JJ, Cardinale CJ, Bakay M, Li D, Maggadottir SM, Thomas KA, Qui H, Chiavacci RM, Kim CE, Wang F, Snyder J, Flatø B, Førre Ø, Denson LA, Thompson SD, Becker ML, Guthery SL, Latiano A, Perez E, Resnick E, Strisciuglio C, Staiano A, Miele E, Silverberg MS, Lie BA, Punaro M, Russell RK, Wilson DC, Dubinsky MC, Monos DS, Annese V, Munro JE, Wise C, Chapel H, Cunningham-Rundles C, Orange JS, Behrens EM, Sullivan KE, Kugathasan S, Griffiths AM, Satsangi J, Grant SFA, Sleiman PMA, Finkel TH, Polychronakos C, Baldassano RN, Luning Prak ET, Ellis JA, Li H, Keating BJ, Hakonarson H. Genetic sharing and heritability of paediatric age of onset autoimmune diseases. Nat Commun 2015; 6:8442. [PMID: 26450413 PMCID: PMC4633631 DOI: 10.1038/ncomms9442] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 08/21/2015] [Indexed: 12/21/2022] Open
Abstract
Autoimmune diseases (AIDs) are polygenic diseases affecting 7-10% of the population in the Western Hemisphere with few effective therapies. Here, we quantify the heritability of paediatric AIDs (pAIDs), including JIA, SLE, CEL, T1D, UC, CD, PS, SPA and CVID, attributable to common genomic variations (SNP-h(2)). SNP-h(2) estimates are most significant for T1D (0.863±s.e. 0.07) and JIA (0.727±s.e. 0.037), more modest for UC (0.386±s.e. 0.04) and CD (0.454±0.025), largely consistent with population estimates and are generally greater than that previously reported by adult GWAS. On pairwise analysis, we observed that the diseases UC-CD (0.69±s.e. 0.07) and JIA-CVID (0.343±s.e. 0.13) are the most strongly correlated. Variations across the MHC strongly contribute to SNP-h(2) in T1D and JIA, but does not significantly contribute to the pairwise rG. Together, our results partition contributions of shared versus disease-specific genomic variations to pAID heritability, identifying pAIDs with unexpected risk sharing, while recapitulating known associations between autoimmune diseases previously reported in adult cohorts.
Collapse
Affiliation(s)
- Yun R. Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Sihai D. Zhao
- Department of Statistics, University of Illinois at Urbana-Champaign, Champaign, Illinois 61820, USA
| | - Jin Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Jonathan P. Bradfield
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Maede Mohebnasab
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Laura Steel
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Julie Kobie
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Debra J. Abrams
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Frank D. Mentch
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Joseph T. Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Yiran Guo
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Zhi Wei
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey 07103, USA
| | - John J. Connolly
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Christopher J. Cardinale
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Marina Bakay
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - S. Melkorka Maggadottir
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Kelly A. Thomas
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Haijun Qui
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Rosetta M. Chiavacci
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Cecilia E. Kim
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Fengxiang Wang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - James Snyder
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Berit Flatø
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo 0372, Norway
| | - Øystein Førre
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo 0372, Norway
| | - Lee A. Denson
- Center for Inflammatory Bowel Disease, Division of Gastroenterology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Susan D. Thompson
- Divison of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Mara L. Becker
- Division of Rheumatology and Division of Clinical Pharmacology, Toxicology, and Therapeutic Innovation, Children's Mercy-Kansas City, Kansas City, Missouri 64108, USA
| | - Stephen L. Guthery
- Department of Pediatrics, University of Utah School of Medicine and Primary Children's Medical Center, Salt Lake City, Utah 84113, USA
| | - Anna Latiano
- RCCS ‘Casa Sollievo della Sofferenza', Division of Gastroenterology, San Giovanni Rotondo 71013, Italy
| | - Elena Perez
- Division of Pediatric Allergy and Immunology, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Elena Resnick
- Institute of Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, New York 10029, USA
| | - Caterina Strisciuglio
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples 80138, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples 80138, Italy
| | - Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II", Naples 80138, Italy
| | - Mark S. Silverberg
- IBD Centre, Mount Sinai Hospital, University of Toronto, 441-600 University Avenue, Toronto, Ontario, Canada M5G 1X5
| | - Benedicte A. Lie
- Department of Immunology, Oslo University Hospital, Rikshospitalet, 0027 Oslo 0372, Norway
| | - Marilynn Punaro
- Texas Scottish Rite Hospital for Children, Dallas, Texas 750219, USA
| | | | - David C. Wilson
- Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, Edinburgh and Child Life and Health, University of Edinburgh, Edinburgh EH9 1UW, UK
| | - Marla C. Dubinsky
- Departments of Pediatrics and Common Disease Genetics, Cedars Sinai Medical Center, Los Angeles, California 90048, USA
| | - Dimitri S. Monos
- Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Vito Annese
- Unit of Gastroenterology, Department of Medical and Surgical Specialties, Careggi University Hospital, Viale Pieraccini 18, Florence 50139, Italy
| | - Jane E. Munro
- Paediatric Rheumatology Unit, Royal Children's Hospital, Parkville, Victoria 3052, Australia
- Arthritis and Rheumatology Research, Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia
| | - Carol Wise
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas 750219, USA
| | - Helen Chapel
- Department of Clinical Immunology, Nuffield Department of Medicine, University of Oxford, OX1 1NF, UK
| | - Charlotte Cunningham-Rundles
- Institute of Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Hospital, New York, New York 10029, USA
| | - Jordan S. Orange
- Section of Immunology, Allergy, and Rheumatology, Department of Pediatric Medicine, Texas Children's Hospital, Houston, Texas 77030, USA
| | - Edward M. Behrens
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Rheumatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Kathleen E. Sullivan
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Subra Kugathasan
- Department of Pediatrics, Emory University School of Medicine and Children's Health Care of Atlanta, Atlanta, Georgia 30329, USA
| | - Anne M. Griffiths
- Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8
| | - Jack Satsangi
- Gastrointestinal Unit, Division of Medical Sciences, School of Molecular and Clinical Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Struan F. A. Grant
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Patrick M. A. Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Terri H. Finkel
- Department of Pediatrics, Nemours Children's Hospital, Orlando, Florida 32827, USA
| | - Constantin Polychronakos
- Departments of Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada H3H 1P3
| | - Robert N. Baldassano
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Gastroenterology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Eline T. Luning Prak
- Department of Pathology and Lab Medicine, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Justine A. Ellis
- Genes, Environment and Complex Disease, Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Hongzhe Li
- Department of Biostatistics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Brendan J. Keating
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
692
|
Abstract
Language is a defining characteristic of the human species, but its foundations remain mysterious. Heritable disorders offer a gateway into biological underpinnings, as illustrated by the discovery that FOXP2 disruptions cause a rare form of speech and language impairment. The genetic architecture underlying language-related disorders is complex, and although some progress has been made, it has proved challenging to pinpoint additional relevant genes with confidence. Next-generation sequencing and genome-wide association studies are revolutionizing understanding of the genetic bases of other neurodevelopmental disorders, like autism and schizophrenia, and providing fundamental insights into the molecular networks crucial for typical brain development. We discuss how a similar genomic perspective, brought to the investigation of language-related phenotypes, promises to yield equally informative discoveries. Moreover, we outline how follow-up studies of genetic findings using cellular systems and animal models can help to elucidate the biological mechanisms involved in the development of brain circuits supporting language.
Collapse
Affiliation(s)
- Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands;
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, 6525 XD Nijmegen, The Netherlands; .,Donders Institute for Brain, Cognition and Behavior, Radboud University, 6525 EN Nijmegen, The Netherlands;
| |
Collapse
|
693
|
Ananthakrishnan AN, Lieberman D. Patient Electronic Health Records as a Means to Approach Genetic Research in Gastroenterology. Gastroenterology 2015; 149:1134-7. [PMID: 26073373 PMCID: PMC4589451 DOI: 10.1053/j.gastro.2015.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 05/06/2015] [Accepted: 06/01/2015] [Indexed: 12/16/2022]
Abstract
Electronic health records (EHRs) are being increasingly utilized and form a unique source of extensive data gathered during routine clinical care. Through use of codified and free text concepts identified using clinical informatics tools, disease labels can be assigned with a high degree of accuracy. Analysis linking such EHR-assigned disease labels to a biospecimen repository has demonstrated that genetic associations identified in prospective cohorts can be replicated with adequate statistical power and novel phenotypic associations identified. In addition, genetic discovery research can be performed utilizing clinical, laboratory, and procedure data obtained during care. Challenges with such research include the need to tackle variability in quality and quantity of EHR data and importance of maintaining patient privacy and data security. With appropriate safeguards, this novel and emerging field of research offers considerable promise and potential to further scientific research in gastroenterology efficiently, cost-effectively, and with engagement of patients and communities.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | - David Lieberman
- Division of Gastroenterology and Hepatology, Oregon Health and Science University, Portland, OR
| |
Collapse
|
694
|
Philippakis AA, Azzariti DR, Beltran S, Brookes AJ, Brownstein CA, Brudno M, Brunner HG, Buske OJ, Carey K, Doll C, Dumitriu S, Dyke SO, den Dunnen JT, Firth HV, Gibbs RA, Girdea M, Gonzalez M, Haendel MA, Hamosh A, Holm IA, Huang L, Hurles ME, Hutton B, Krier JB, Misyura A, Mungall CJ, Paschall J, Paten B, Robinson PN, Schiettecatte F, Sobreira NL, Swaminathan GJ, Taschner PE, Terry SF, Washington NL, Züchner S, Boycott KM, Rehm HL. The Matchmaker Exchange: a platform for rare disease gene discovery. Hum Mutat 2015; 36:915-21. [PMID: 26295439 PMCID: PMC4610002 DOI: 10.1002/humu.22858] [Citation(s) in RCA: 365] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/21/2015] [Indexed: 12/21/2022]
Abstract
There are few better examples of the need for data sharing than in the rare disease community, where patients, physicians, and researchers must search for "the needle in a haystack" to uncover rare, novel causes of disease within the genome. Impeding the pace of discovery has been the existence of many small siloed datasets within individual research or clinical laboratory databases and/or disease-specific organizations, hoping for serendipitous occasions when two distant investigators happen to learn they have a rare phenotype in common and can "match" these cases to build evidence for causality. However, serendipity has never proven to be a reliable or scalable approach in science. As such, the Matchmaker Exchange (MME) was launched to provide a robust and systematic approach to rare disease gene discovery through the creation of a federated network connecting databases of genotypes and rare phenotypes using a common application programming interface (API). The core building blocks of the MME have been defined and assembled. Three MME services have now been connected through the API and are available for community use. Additional databases that support internal matching are anticipated to join the MME network as it continues to grow.
Collapse
Affiliation(s)
- Anthony A. Philippakis
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Cardiology, Brigham & Women's Hospital,
Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Danielle R. Azzariti
- Laboratory for Molecular Medicine, Partners Personalized
Medicine, Boston, MA USA
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico, Barcelona,
Spain
| | | | - Catherine A. Brownstein
- Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics and the Manton Center for
Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto,
Canada
- Genetics and Genome Biology Program, The Hospital for Sick
Children, Toronto, Canada
- Centre for Computational Medicine, The Hospital for Sick
Children, Toronto, Canada
| | - Han G. Brunner
- Radboud University Medical Center,Department of Human
Genetics, PO Box 9101, 6500HB Nijmegen, The Netherlands
- Maastricht University Medical Center, Department of Clinical
Genetics,PO Box 5800, 6202AZ Maastricht, The Netherlands
| | - Orion J. Buske
- Department of Computer Science, University of Toronto, Toronto,
Canada
- Genetics and Genome Biology Program, The Hospital for Sick
Children, Toronto, Canada
- Centre for Computational Medicine, The Hospital for Sick
Children, Toronto, Canada
| | | | | | - Sergiu Dumitriu
- Centre for Computational Medicine, The Hospital for Sick
Children, Toronto, Canada
| | - Stephanie O.M. Dyke
- Centre of Genomics and Policy, Faculty of Medicine, McGill
University, Canada
| | - Johan T. den Dunnen
- Human and Clinical Genetics, Leiden University Medical Center,
Leiden, Nederland
| | - Helen V. Firth
- East Anglian Medical Genetics Service, Box 134, Cambridge
University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ,
UK
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine,
Houston, Tx 77030, U.S.A
| | - Marta Girdea
- Department of Computer Science, University of Toronto, Toronto,
Canada
- Centre for Computational Medicine, The Hospital for Sick
Children, Toronto, Canada
| | | | - Melissa A. Haendel
- Department of Medical Informatics and Clinical Epidemiology,
Oregon Health & Science University, Portland, OR, USA
| | - Ada Hamosh
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins
University, Baltimore, MD, USA
| | - Ingrid A. Holm
- Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics and the Manton Center for
Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA
| | - Lijia Huang
- The Children's Hospital of Eastern Ontario Research Institute,
Ottawa, ON, Canada
| | - Matthew E. Hurles
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Hinxton CB10 1SA, U.K
| | - Ben Hutton
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Hinxton CB10 1SA, U.K
| | - Joel B. Krier
- Division of Genetics, Department of Medicine, Brigham and
Women's Hospital, 41 Avenue Louis Pasteur, Suite 301, Boston, MA 02115, USA
| | - Andriy Misyura
- Centre for Computational Medicine, The Hospital for Sick
Children, Toronto, Canada
| | | | - Justin Paschall
- European Molecular Biology Laboratory - European
Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SD,
UK
| | - Benedict Paten
- UC Santa Cruz Genomics Institute, 1156 High Street, Santa
Cruz, CA, USA
| | - Peter N. Robinson
- Institute for Medical Genetics and Human Genetics,
Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
- Max Planck Institute for Molecular Genetics, 14195 Berlin,
Germany
- Institute for Bioinformatics, Department of Mathematics and
Computer Science, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin Brandenburg Center for Regenerative Therapies, 13353
Berlin, Germany
| | | | - Nara L. Sobreira
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins
University, Baltimore, MD, USA
| | - Ganesh J. Swaminathan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus,
Hinxton CB10 1SA, U.K
| | - Peter E. Taschner
- Department of Medical Informatics and Clinical Epidemiology,
Oregon Health & Science University, Portland, OR, USA
- Division of Genetics, Department of Medicine, Brigham and
Women's Hospital, 41 Avenue Louis Pasteur, Suite 301, Boston, MA 02115, USA
| | | | | | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics
and John P. Hussman Institute for Human Genomics, University of Miami Miller School of
Medicine, Miami, FL, USA
| | - Kym M. Boycott
- Department of Genetics, Children's Hospital of Eastern
Ontario, Ottawa, Ontario, Canada
| | - Heidi L. Rehm
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Laboratory for Molecular Medicine, Partners Personalized
Medicine, Boston, MA USA
- Department of Pathology, Brigham & Women's Hospital, Boston,
MA, USA
| |
Collapse
|
695
|
Li Y, Ryan PB, Wei Y, Friedman C. A Method to Combine Signals from Spontaneous Reporting Systems and Observational Healthcare Data to Detect Adverse Drug Reactions. Drug Saf 2015; 38:895-908. [PMID: 26153397 PMCID: PMC4579260 DOI: 10.1007/s40264-015-0314-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Observational healthcare data contain information useful for hastening detection of adverse drug reactions (ADRs) that may be missed by using data in spontaneous reporting systems (SRSs) alone. There are only several papers describing methods that integrate evidence from healthcare databases and SRSs. We propose a methodology that combines ADR signals from these two sources. OBJECTIVES The aim of this study was to investigate whether the proposed method would result in more accurate ADR detection than methods using SRSs or healthcare data alone. RESEARCH DESIGN We applied the method to four clinically serious ADRs, and evaluated it using three experiments that involve combining an SRS with a single facility small-scale electronic health record (EHR), a larger scale network-based EHR, and a much larger scale healthcare claims database. The evaluation used a reference standard comprising 165 positive and 234 negative drug-ADR pairs. MEASURES Area under the receiver operator characteristics curve (AUC) was computed to measure performance. RESULTS There was no improvement in the AUC when the SRS and small-scale HER were combined. The AUC of the combined SRS and large-scale EHR was 0.82 whereas it was 0.76 for each of the individual systems. Similarly, the AUC of the combined SRS and claims system was 0.82 whereas it was 0.76 and 0.78, respectively, for the individual systems. CONCLUSIONS The proposed method resulted in a significant improvement in the accuracy of ADR detection when the resources used for combining had sufficient amounts of data, demonstrating that the method could integrate evidence from multiple sources and serve as a tool in actual pharmacovigilance practice.
Collapse
Affiliation(s)
- Ying Li
- Department of Biomedical Informatics, Columbia University Medical Center, 622 W. 168th Street, Presbyterian Building 20th Floor, New York, NY, 10032, USA.
| | - Patrick B Ryan
- Department of Biomedical Informatics, Columbia University Medical Center, 622 W. 168th Street, Presbyterian Building 20th Floor, New York, NY, 10032, USA
- Janssen Research and Development, 1125 Trenton Harbourton Rd, Titusville, NJ, 08560, USA
- Observational Health Data Sciences and Informatics (OHDSI), New York, NY, 10032, USA
| | - Ying Wei
- Department of Biostatistics, Columbia University, New York, NY, 10032, USA
| | - Carol Friedman
- Department of Biomedical Informatics, Columbia University Medical Center, 622 W. 168th Street, Presbyterian Building 20th Floor, New York, NY, 10032, USA
| |
Collapse
|
696
|
Oellrich A, Collier N, Groza T, Rebholz-Schuhmann D, Shah N, Bodenreider O, Boland MR, Georgiev I, Liu H, Livingston K, Luna A, Mallon AM, Manda P, Robinson PN, Rustici G, Simon M, Wang L, Winnenburg R, Dumontier M. The digital revolution in phenotyping. Brief Bioinform 2015; 17:819-30. [PMID: 26420780 PMCID: PMC5036847 DOI: 10.1093/bib/bbv083] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 12/22/2022] Open
Abstract
Phenotypes have gained increased notoriety in the clinical and biological domain owing to their application in numerous areas such as the discovery of disease genes and drug targets, phylogenetics and pharmacogenomics. Phenotypes, defined as observable characteristics of organisms, can be seen as one of the bridges that lead to a translation of experimental findings into clinical applications and thereby support 'bench to bedside' efforts. However, to build this translational bridge, a common and universal understanding of phenotypes is required that goes beyond domain-specific definitions. To achieve this ambitious goal, a digital revolution is ongoing that enables the encoding of data in computer-readable formats and the data storage in specialized repositories, ready for integration, enabling translational research. While phenome research is an ongoing endeavor, the true potential hidden in the currently available data still needs to be unlocked, offering exciting opportunities for the forthcoming years. Here, we provide insights into the state-of-the-art in digital phenotyping, by means of representing, acquiring and analyzing phenotype data. In addition, we provide visions of this field for future research work that could enable better applications of phenotype data.
Collapse
|
697
|
Srinivasan S, Clements JA, Batra J. Single nucleotide polymorphisms in clinics: Fantasy or reality for cancer? Crit Rev Clin Lab Sci 2015; 53:29-39. [DOI: 10.3109/10408363.2015.1075469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
698
|
Mo H, Thompson WK, Rasmussen LV, Pacheco JA, Jiang G, Kiefer R, Zhu Q, Xu J, Montague E, Carrell DS, Lingren T, Mentch FD, Ni Y, Wehbe FH, Peissig PL, Tromp G, Larson EB, Chute CG, Pathak J, Denny JC, Speltz P, Kho AN, Jarvik GP, Bejan CA, Williams MS, Borthwick K, Kitchner TE, Roden DM, Harris PA. Desiderata for computable representations of electronic health records-driven phenotype algorithms. J Am Med Inform Assoc 2015; 22:1220-30. [PMID: 26342218 PMCID: PMC4639716 DOI: 10.1093/jamia/ocv112] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/24/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Electronic health records (EHRs) are increasingly used for clinical and translational research through the creation of phenotype algorithms. Currently, phenotype algorithms are most commonly represented as noncomputable descriptive documents and knowledge artifacts that detail the protocols for querying diagnoses, symptoms, procedures, medications, and/or text-driven medical concepts, and are primarily meant for human comprehension. We present desiderata for developing a computable phenotype representation model (PheRM). METHODS A team of clinicians and informaticians reviewed common features for multisite phenotype algorithms published in PheKB.org and existing phenotype representation platforms. We also evaluated well-known diagnostic criteria and clinical decision-making guidelines to encompass a broader category of algorithms. RESULTS We propose 10 desired characteristics for a flexible, computable PheRM: (1) structure clinical data into queryable forms; (2) recommend use of a common data model, but also support customization for the variability and availability of EHR data among sites; (3) support both human-readable and computable representations of phenotype algorithms; (4) implement set operations and relational algebra for modeling phenotype algorithms; (5) represent phenotype criteria with structured rules; (6) support defining temporal relations between events; (7) use standardized terminologies and ontologies, and facilitate reuse of value sets; (8) define representations for text searching and natural language processing; (9) provide interfaces for external software algorithms; and (10) maintain backward compatibility. CONCLUSION A computable PheRM is needed for true phenotype portability and reliability across different EHR products and healthcare systems. These desiderata are a guide to inform the establishment and evolution of EHR phenotype algorithm authoring platforms and languages.
Collapse
Affiliation(s)
- Huan Mo
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - William K Thompson
- Center for Biomedical Research Informatics, NorthShore University HealthSystem, Evanston, IL, USA
| | - Luke V Rasmussen
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer A Pacheco
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Guoqian Jiang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Richard Kiefer
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Qian Zhu
- Department of Information Systems, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Jie Xu
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Enid Montague
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Todd Lingren
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Frank D Mentch
- Center for Applied Genomics, the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yizhao Ni
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Firas H Wehbe
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Peggy L Peissig
- Marshfield Clinic Research Foundation, Marshfield Clinic, Marshfield, WI, USA
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Cape Town, South Africa
| | | | - Christopher G Chute
- Division of General Internal Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jyotishman Pathak
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Peter Speltz
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Abel N Kho
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Gail P Jarvik
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA, USA Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Marc S Williams
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Kenneth Borthwick
- The Sigfried and Janet Weis Center for Research, Geisinger Health System, Danville, PA, USA
| | - Terrie E Kitchner
- Marshfield Clinic Research Foundation, Marshfield Clinic, Marshfield, WI, USA
| | - Dan M Roden
- Department of Medicine, Vanderbilt University, Nashville, TN, USA Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Paul A Harris
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
699
|
Li YR, Li J, Zhao SD, Bradfield JP, Mentch FD, Maggadottir SM, Hou C, Abrams DJ, Chang D, Gao F, Guo Y, Wei Z, Connolly JJ, Cardinale CJ, Bakay M, Glessner JT, Li D, Kao C, Thomas KA, Qiu H, Chiavacci RM, Kim CE, Wang F, Snyder J, Richie MD, Flatø B, Førre Ø, Denson LA, Thompson SD, Becker ML, Guthery SL, Latiano A, Perez E, Resnick E, Russell RK, Wilson DC, Silverberg MS, Annese V, Lie BA, Punaro M, Dubinsky MC, Monos DS, Strisciuglio C, Staiano A, Miele E, Kugathasan S, Ellis JA, Munro JE, Sullivan KE, Wise CA, Chapel H, Cunningham-Rundles C, Grant SFA, Orange JS, Sleiman PMA, Behrens EM, Griffiths AM, Satsangi J, Finkel TH, Keinan A, Prak ETL, Polychronakos C, Baldassano RN, Li H, Keating BJ, Hakonarson H. Meta-analysis of shared genetic architecture across ten pediatric autoimmune diseases. Nat Med 2015; 21:1018-1027. [PMID: 26301688 PMCID: PMC4863040 DOI: 10.1038/nm.3933] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Accepted: 07/23/2015] [Indexed: 12/15/2022]
Abstract
Genome-wide association studies (GWASs) have identified hundreds of susceptibility genes, including shared associations across clinically distinct autoimmune diseases. We performed an inverse χ(2) meta-analysis across ten pediatric-age-of-onset autoimmune diseases (pAIDs) in a case-control study including more than 6,035 cases and 10,718 shared population-based controls. We identified 27 genome-wide significant loci associated with one or more pAIDs, mapping to in silico-replicated autoimmune-associated genes (including IL2RA) and new candidate loci with established immunoregulatory functions such as ADGRL2, TENM3, ANKRD30A, ADCY7 and CD40LG. The pAID-associated single-nucleotide polymorphisms (SNPs) were functionally enriched for deoxyribonuclease (DNase)-hypersensitivity sites, expression quantitative trait loci (eQTLs), microRNA (miRNA)-binding sites and coding variants. We also identified biologically correlated, pAID-associated candidate gene sets on the basis of immune cell expression profiling and found evidence of genetic sharing. Network and protein-interaction analyses demonstrated converging roles for the signaling pathways of type 1, 2 and 17 helper T cells (TH1, TH2 and TH17), JAK-STAT, interferon and interleukin in multiple autoimmune diseases.
Collapse
Affiliation(s)
- Yun R Li
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Medical Scientist Training Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jin Li
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sihai D Zhao
- Department of Biostatistics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan P Bradfield
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Frank D Mentch
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - S Melkorka Maggadottir
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cuiping Hou
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Debra J Abrams
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Diana Chang
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA
- Program in Computational Biology and Medicine, Cornell University, Ithaca, New York, USA
| | - Feng Gao
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA
| | - Yiran Guo
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - John J Connolly
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Christopher J Cardinale
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marina Bakay
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph T Glessner
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dong Li
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Charlly Kao
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kelly A Thomas
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Haijun Qiu
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rosetta M Chiavacci
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cecilia E Kim
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Fengxiang Wang
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - James Snyder
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Marylyn D Richie
- Department of Biochemistry and Molecular Biology, Eberly College of Science, The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Berit Flatø
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øystein Førre
- Department of Rheumatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Lee A Denson
- Division of Gastroenterology, The Center for Inflammatory Bowel Disease, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Susan D Thompson
- Divison of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Mara L Becker
- Division of Rheumatology, Children's Mercy Hospitals and Clinics, Kansas City, Missouri, USA
| | - Stephen L Guthery
- Department of Pediatrics, University of Utah School of Medicine and Primary Children's Medical Center, Salt Lake City, Utah, USA
| | - Anna Latiano
- Division of Gastroenterology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Elena Perez
- Division of Pediatric Allergy and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Elena Resnick
- Institute of Immunology and Department of Medicine, Mount Sinai School of Medicine, New York, New York, USA
| | - Richard K Russell
- Department of Paediatric Gastroenterology, Yorkhill Hospital for Sick Children, Glasgow, Scotland, UK
| | - David C Wilson
- Paediatric Gastroenterology and Nutrition, Royal Hospital for Sick Children, University of Edinburgh, Ediburgh, UK
| | - Mark S Silverberg
- Mount Sinai Hospital IBD Centre, University of Toronto, Toronto, Ontario, Canada
| | - Vito Annese
- Unit of Gastroenterology, Department of Medical and Surgical Specialties, Careggi University Hospital, Florence, Italy
| | - Benedicte A Lie
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Marilynn Punaro
- Department of Rheumatology, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
| | - Marla C Dubinsky
- Department of Pediatrics, Pediatric IBD Center, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Dimitri S Monos
- Department of Pathology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Caterina Strisciuglio
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, Naples, Italy
| | - Subra Kugathasan
- Department of Pediatrics, Emory University School of Medicine and Children's Health Care of Atlanta, Atlanta, Georgia, USA
| | - Justine A Ellis
- Genes, Environment and Complex Disease, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Pediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Munro
- Pediatric Rheumatology Unit, Royal Children's Hospital, Parkville, Victoria, Australia
- Arthritis and Rheumatology Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carol A Wise
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
| | - Helen Chapel
- Department of Clinical Immunology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Struan F A Grant
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jordan S Orange
- Section of Immunology, Allergy, and Rheumatology, Department of Pediatric Medicine, Texas Children's Hospital, Houston, Texas, USA
| | - Patrick M A Sleiman
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward M Behrens
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Anne M Griffiths
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jack Satsangi
- Gastrointestinal Unit, Division of Medical Sciences, School of Molecular and Clinical Medicine, University of Edinburgh, Edinburgh, UK
| | - Terri H Finkel
- Department of Pediatrics, Nemours Children's Hospital, Orlando, Florida, USA
| | - Alon Keinan
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, USA
- Program in Computational Biology and Medicine, Cornell University, Ithaca, New York, USA
| | - Eline T Luning Prak
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Constantin Polychronakos
- Departments of Pediatrics and Human Genetics, McGill University Health Centre Research Institute, Montréal, Québec, Canada
| | - Robert N Baldassano
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Gastroenterology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hongzhe Li
- Department of Pathology and Lab Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brendan J Keating
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
700
|
Genetics of glucocorticoid-associated osteonecrosis in children with acute lymphoblastic leukemia. Blood 2015; 126:1770-6. [PMID: 26265699 DOI: 10.1182/blood-2015-05-643601] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/29/2015] [Indexed: 11/20/2022] Open
Abstract
Glucocorticoids are important therapy for acute lymphoblastic leukemia (ALL) and their major adverse effect is osteonecrosis. Our goal was to identify genetic and nongenetic risk factors for osteonecrosis. We performed a genome-wide association study of single nucleotide polymorphisms (SNPs) in a discovery cohort comprising 2285 children with ALL, treated on the Children's Oncology Group AALL0232 protocol (NCT00075725), adjusting for covariates. The minor allele at SNP rs10989692 (near the glutamate receptor GRIN3A locus) was associated with osteonecrosis (hazard ratio = 2.03; P = 3.59 × 10(-7)). The association was supported by 2 replication cohorts, including 361 children with ALL on St. Jude's Total XV protocol (NCT00137111) and 309 non-ALL patients from Vanderbilt University's BioVU repository treated with glucocorticoids (odds ratio [OR] = 1.87 and 2.26; P = .063 and .0074, respectively). In a meta-analysis, rs10989692 was also highest ranked (P = 2.68 × 10(-8)), and the glutamate pathway was the top ranked pathway (P = 9.8 × 10(-4)). Osteonecrosis-associated glutamate receptor variants were also associated with other vascular phenotypes including cerebral ischemia (OR = 1.64; P = 2.5 × 10(-3)), and arterial embolism and thrombosis (OR = 1.88; P = 4.2 × 10(-3)). In conclusion, osteonecrosis was associated with inherited variations near glutamate receptor genes. Further understanding this association may allow interventions to decrease osteonecrosis. These trials are registered at www.clinicaltrials.gov as #NCT00075725 and #NCT00137111.
Collapse
|