Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Benjamin S. Glicksberg is active.

Publication


Featured researches published by Benjamin S. Glicksberg.


Science Translational Medicine | 2015

Identification of type 2 diabetes subgroups through topological analysis of patient similarity

Li Li; Wei-Yi Cheng; Benjamin S. Glicksberg; Omri Gottesman; Ronald Tamler; Rong Chen; Erwin P. Bottinger; Joel T. Dudley

Patient networks constructed from genotype data and electronic medical records pinpointed three type 2 diabetes subtypes. Networks work for diabetes Big problems require big solutions, and for complex diseases such as cancer or diabetes, the big solution is big data. One long-term goal of U.S. President Barack Obama’s Precision Medicine Initiative is to assemble medical and genetic data from at least one million volunteers. But how might researchers use all those data? Li et al. provide one answer by using patient electronic medical records (EMRs) and genotype data from Mount Sinai Medical Center in New York to characterize new subtypes of type 2 diabetes (T2D). The group first clustered EMR data to identify T2D patients within the larger group. Topological analysis of the T2D group identified three new T2D subtypes on the basis of distinct patterns of clinical characteristics and disease comorbidities. Genetic association analysis identified more than 300 single nucleotide polymorphisms (SNPs) specific to each subtype. The authors found that classical T2D features such as obesity, high blood sugar, kidney disease, and eye disease, were limited to subtype 1, whereas other comorbidities such as cancer and neurological diseases were specific to subtypes 2 and 3, respectively. These distinctions might call for tailored treatment regimens rather than a one-size-fits-all approach for T2D. Although a larger sample size is needed to determine causal relationships, this study demonstrates the potential of precision medicine. Type 2 diabetes (T2D) is a heterogeneous complex disease affecting more than 29 million Americans alone with a rising prevalence trending toward steady increases in the coming decades. Thus, there is a pressing clinical need to improve early prevention and clinical management of T2D and its complications. Clinicians have understood that patients who carry the T2D diagnosis have a variety of phenotypes and susceptibilities to diabetes-related complications. We used a precision medicine approach to characterize the complexity of T2D patient populations based on high-dimensional electronic medical records (EMRs) and genotype data from 11,210 individuals. We successfully identified three distinct subgroups of T2D from topology-based patient-patient networks. Subtype 1 was characterized by T2D complications diabetic nephropathy and diabetic retinopathy; subtype 2 was enriched for cancer malignancy and cardiovascular diseases; and subtype 3 was associated most strongly with cardiovascular diseases, neurological diseases, allergies, and HIV infections. We performed a genetic association analysis of the emergent T2D subtypes to identify subtype-specific genetic markers and identified 1279, 1227, and 1338 single-nucleotide polymorphisms (SNPs) that mapped to 425, 322, and 437 unique genes specific to subtypes 1, 2, and 3, respectively. By assessing the human disease–SNP association for each subtype, the enriched phenotypes and biological functions at the gene level for each subtype matched with the disease comorbidities and clinical differences that we identified through EMRs. Our approach demonstrates the utility of applying the precision medicine paradigm in T2D and the promise of extending the approach to the study of other complex, multifactorial diseases.


Briefings in Bioinformatics | 2017

Translational bioinformatics in the era of real-time biomedical, health care and wellness data streams

Khader Shameer; Marcus A. Badgeley; Riccardo Miotto; Benjamin S. Glicksberg; Joseph W. Morgan; Joel T. Dudley

Abstract Monitoring and modeling biomedical, health care and wellness data from individuals and converging data on a population scale have tremendous potential to improve understanding of the transition to the healthy state of human physiology to disease setting. Wellness monitoring devices and companion software applications capable of generating alerts and sharing data with health care providers or social networks are now available. The accessibility and clinical utility of such data for disease or wellness research are currently limited. Designing methods for streaming data capture, real‐time data aggregation, machine learning, predictive analytics and visualization solutions to integrate wellness or health monitoring data elements with the electronic medical records (EMRs) maintained by health care providers permits better utilization. Integration of population‐scale biomedical, health care and wellness data would help to stratify patients for active health management and to understand clinically asymptomatic patients and underlying illness trajectories. In this article, we discuss various health‐monitoring devices, their ability to capture the unique state of health represented in a patient and their application in individualized diagnostics, prognosis, clinical or wellness intervention. We also discuss examples of translational bioinformatics approaches to integrating patient‐generated data with existing EMRs, personal health records, patient portals and clinical data repositories. Briefly, translational bioinformatics methods, tools and resources are at the center of these advances in implementing real‐time biomedical and health care analytics in the clinical setting. Furthermore, these advances are poised to play a significant role in clinical decision‐making and implementation of data‐driven medicine and wellness care.


BMC Genomics | 2015

Disease-associated variants in different categories of disease located in distinct regulatory elements

Meng Ma; Ying Ru; Ling-Shiang Chuang; Nai-Yun Hsu; Lisong Shi; Jörg Hakenberg; Wei-Yi Cheng; Andrew V. Uzilov; Wei Ding; Benjamin S. Glicksberg; Rong Chen

BackgroundThe invention of high throughput sequencing technologies has led to the discoveries of hundreds of thousands of genetic variants associated with thousands of human diseases. Many of these genetic variants are located outside the protein coding regions, and as such, it is challenging to interpret the function of these genetic variants by traditional genetic approaches. Recent genome-wide functional genomics studies, such as FANTOM5 and ENCODE have uncovered a large number of regulatory elements across hundreds of different tissues or cell lines in the human genome. These findings provide an opportunity to study the interaction between regulatory elements and disease-associated genetic variants. Identifying these diseased-related regulatory elements will shed light on understanding the mechanisms of how these variants regulate gene expression and ultimately result in disease formation and progression.ResultsIn this study, we curated and categorized 27,558 Mendelian disease variants, 20,964 complex disease variants, 5,809 cancer predisposing germline variants, and 43,364 recurrent cancer somatic mutations. Compared against nine different types of regulatory regions from FANTOM5 and ENCODE projects, we found that different types of disease variants show distinctive propensity for particular regulatory elements. Mendelian disease variants and recurrent cancer somatic mutations are 22-fold and 10- fold significantly enriched in promoter regions respectively (q<0.001), compared with allele-frequency-matched genomic background. Separate from these two categories, cancer predisposing germline variants are 27-fold enriched in histone modification regions (q<0.001), 10-fold enriched in chromatin physical interaction regions (q<0.001), and 6-fold enriched in transcription promoters (q<0.001). Furthermore, Mendelian disease variants and recurrent cancer somatic mutations share very similar distribution across types of functional effects.We further found that regulatory regions are located within over 50% coding exon regions. Transcription promoters, methylation regions, and transcription insulators have the highest density of disease variants, with 472, 239, and 72 disease variants per one million base pairs, respectively.ConclusionsDisease-associated variants in different disease categories are preferentially located in particular regulatory elements. These results will be useful for an overall understanding about the differences among the pathogenic mechanisms of various disease-associated variants.


BMJ Open | 2016

EHDViz: clinical dashboard development using open-source technologies

Marcus A. Badgeley; Khader Shameer; Benjamin S. Glicksberg; Max S Tomlinson; Patrick J. McCormick; Andrew Kasarskis; David L. Reich; Joel T. Dudley

Objective To design, develop and prototype clinical dashboards to integrate high-frequency health and wellness data streams using interactive and real-time data visualisation and analytics modalities. Materials and methods We developed a clinical dashboard development framework called electronic healthcare data visualization (EHDViz) toolkit for generating web-based, real-time clinical dashboards for visualising heterogeneous biomedical, healthcare and wellness data. The EHDViz is an extensible toolkit that uses R packages for data management, normalisation and producing high-quality visualisations over the web using R/Shiny web server architecture. We have developed use cases to illustrate utility of EHDViz in different scenarios of clinical and wellness setting as a visualisation aid for improving healthcare delivery. Results Using EHDViz, we prototyped clinical dashboards to demonstrate the contextual versatility of EHDViz toolkit. An outpatient cohort was used to visualise population health management tasks (n=14 221), and an inpatient cohort was used to visualise real-time acuity risk in a clinical unit (n=445), and a quantified-self example using wellness data from a fitness activity monitor worn by a single individual was also discussed (n-of-1). The back-end system retrieves relevant data from data source, populates the main panel of the application and integrates user-defined data features in real-time and renders output using modern web browsers. The visualisation elements can be customised using health features, disease names, procedure names or medical codes to populate the visualisations. The source code of EHDViz and various prototypes developed using EHDViz are available in the public domain at http://ehdviz.dudleylab.org. Conclusions Collaborative data visualisations, wellness trend predictions, risk estimation, proactive acuity status monitoring and knowledge of complex disease indicators are essential components of implementing data-driven precision medicine. As an open-source visualisation framework capable of integrating health assessment, EHDViz aims to be a valuable toolkit for rapid design, development and implementation of scalable clinical data visualisation dashboards.


Heart | 2018

Machine learning in cardiovascular medicine: are we there yet?

Khader Shameer; Kipp W. Johnson; Benjamin S. Glicksberg; Joel T. Dudley; Partho P. Sengupta

Artificial intelligence (AI) broadly refers to analytical algorithms that iteratively learn from data, allowing computers to find hidden insights without being explicitly programmed where to look. These include a family of operations encompassing several terms like machine learning, cognitive learning, deep learning and reinforcement learning-based methods that can be used to integrate and interpret complex biomedical and healthcare data in scenarios where traditional statistical methods may not be able to perform. In this review article, we discuss the basics of machine learning algorithms and what potential data sources exist; evaluate the need for machine learning; and examine the potential limitations and challenges of implementing machine in the context of cardiovascular medicine. The most promising avenues for AI in medicine are the development of automated risk prediction algorithms which can be used to guide clinical care; use of unsupervised learning techniques to more precisely phenotype complex disease; and the implementation of reinforcement learning algorithms to intelligently augment healthcare providers. The utility of a machine learning-based predictive model will depend on factors including data heterogeneity, data depth, data breadth, nature of modelling task, choice of machine learning and feature selection algorithms, and orthogonal evidence. A critical understanding of the strength and limitations of various methods and tasks amenable to machine learning is vital. By leveraging the growing corpus of big data in medicine, we detail pathways by which machine learning may facilitate optimal development of patient-specific models for improving diagnoses, intervention and outcome in cardiovascular medicine.


Bioinformatics | 2016

Comparative analyses of population-scale phenomic data in electronic medical records reveal race-specific disease networks

Benjamin S. Glicksberg; Li Li; Marcus A. Badgeley; Khader Shameer; Roman Kosoy; Noam D. Beckmann; Nam H. Pho; Jörg Hakenberg; Meng Ma; Kristin L. Ayers; Gabriel E. Hoffman; Shuyu Dan Li; Eric E. Schadt; Chirag Patel; Rong Chen; Joel T. Dudley

Motivation: Underrepresentation of racial groups represents an important challenge and major gap in phenomics research. Most of the current human phenomics research is based primarily on European populations; hence it is an important challenge to expand it to consider other population groups. One approach is to utilize data from EMR databases that contain patient data from diverse demographics and ancestries. The implications of this racial underrepresentation of data can be profound regarding effects on the healthcare delivery and actionability. To the best of our knowledge, our work is the first attempt to perform comparative, population-scale analyses of disease networks across three different populations, namely Caucasian (EA), African American (AA) and Hispanic/Latino (HL). Results: We compared susceptibility profiles and temporal connectivity patterns for 1988 diseases and 37 282 disease pairs represented in a clinical population of 1 025 573 patients. Accordingly, we revealed appreciable differences in disease susceptibility, temporal patterns, network structure and underlying disease connections between EA, AA and HL populations. We found 2158 significantly comorbid diseases for the EA cohort, 3265 for AA and 672 for HL. We further outlined key disease pair associations unique to each population as well as categorical enrichments of these pairs. Finally, we identified 51 key ‘hub’ diseases that are the focal points in the race-centric networks and of particular clinical importance. Incorporating race-specific disease comorbidity patterns will produce a more accurate and complete picture of the disease landscape overall and could support more precise understanding of disease relationships and patient management towards improved clinical outcomes. Contacts: [email protected] or [email protected] Supplementary information: Supplementary data are available at Bioinformatics online.


Briefings in Bioinformatics | 2018

Systematic analyses of drugs and disease indications in RepurposeDB reveal pharmacological, biological and epidemiological factors influencing drug repositioning

Khader Shameer; Benjamin S. Glicksberg; Rachel Hodos; Kipp W. Johnson; Marcus A. Badgeley; Ben Readhead; Max S Tomlinson; Timothy O’Connor; Riccardo Miotto; Brian Kidd; Rong Chen; Avi Ma’ayan; Joel T. Dudley

&NA; Increase in global population and growing disease burden due to the emergence of infectious diseases (Zika virus), multidrug‐resistant pathogens, drug‐resistant cancers (cisplatin‐resistant ovarian cancer) and chronic diseases (arterial hypertension) necessitate effective therapies to improve health outcomes. However, the rapid increase in drug development cost demands innovative and sustainable drug discovery approaches. Drug repositioning, the discovery of new or improved therapies by reevaluation of approved or investigational compounds, solves a significant gap in the public health setting and improves the productivity of drug development. As the number of drug repurposing investigations increases, a new opportunity has emerged to understand factors driving drug repositioning through systematic analyses of drugs, drug targets and associated disease indications. However, such analyses have so far been hampered by the lack of a centralized knowledgebase, benchmarking data sets and reporting standards. To address these knowledge and clinical needs, here, we present RepurposeDB, a collection of repurposed drugs, drug targets and diseases, which was assembled, indexed and annotated from public data. RepurposeDB combines information on 253 drugs [small molecules (74.30%) and protein drugs (25.29%)] and 1125 diseases. Using RepurposeDB data, we identified pharmacological (chemical descriptors, physicochemical features and absorption, distribution, metabolism, excretion and toxicity properties), biological (protein domains, functional process, molecular mechanisms and pathway cross talks) and epidemiological (shared genetic architectures, disease comorbidities and clinical phenotype similarities) factors mediating drug repositioning. Collectively, RepurposeDB is developed as the reference database for drug repositioning investigations. The pharmacological, biological and epidemiological principles of drug repositioning identified from the meta‐analyses could augment therapeutic development.


PLOS ONE | 2016

Data-Driven Identification of Risk Factors of Patient Satisfaction at a Large Urban Academic Medical Center

Li Li; Nathan J. Lee; Benjamin S. Glicksberg; Brian Radbill; Joel T. Dudley

Background The Hospital Consumer Assessment of Healthcare Providers and Systems (HCAHPS) survey is the first publicly reported nationwide survey to evaluate and compare hospitals. Increasing patient satisfaction is an important goal as it aims to achieve a more effective and efficient healthcare delivery system. In this study, we develop and apply an integrative, data-driven approach to identify clinical risk factors that associate with patient satisfaction outcomes. Methods We included 1,771 unique adult patients who completed the HCAHPS survey and were discharged from the inpatient Medicine service from 2010 to 2012. We collected 266 clinical features including patient demographics, lab measurements, medications, disease categories, and procedures. We developed and applied a data-driven approach to identify risk factors that associate with patient satisfaction outcomes. Findings We identify 102 significant risk factors associating with 18 surveyed questions. The most significantly recurrent clinical risk factors were: self-evaluation of health, education level, Asian, White, treatment in BMT oncology division, being prescribed a new medication. Patients who were prescribed pregabalin were less satisfied particularly in relation to communication with nurses and pain management. Explanation of medication usage was associated with communication with nurses (q = 0.001); however, explanation of medication side effects was associated with communication with doctors (q = 0.003). Overall hospital rating was associated with hospital environment, communication with doctors, and communication about medicines. However, patient likelihood to recommend hospital was associated with hospital environment, communication about medicines, pain management, and communication with nurse. Conclusions Our study identified a number of putatively novel clinical risk factors for patient satisfaction that suggest new opportunities to better understand and manage patient satisfaction. Hospitals can use a data-driven approach to identify clinical risk factors for poor patient satisfaction to support development of specific interventions to improve patients’ experience of care.


JACC: Basic to Translational Science | 2017

Enabling Precision Cardiology Through Multiscale Biology and Systems Medicine

Kipp W. Johnson; Khader Shameer; Benjamin S. Glicksberg; Ben Readhead; Partho P. Sengupta; Johan Björkegren; Jason C. Kovacic; Joel T. Dudley

Summary The traditional paradigm of cardiovascular disease research derives insight from large-scale, broadly inclusive clinical studies of well-characterized pathologies. These insights are then put into practice according to standardized clinical guidelines. However, stagnation in the development of new cardiovascular therapies and variability in therapeutic response implies that this paradigm is insufficient for reducing the cardiovascular disease burden. In this state-of-the-art review, we examine 3 interconnected ideas we put forth as key concepts for enabling a transition to precision cardiology: 1) precision characterization of cardiovascular disease with machine learning methods; 2) the application of network models of disease to embrace disease complexity; and 3) using insights from the previous 2 ideas to enable pharmacology and polypharmacology systems for more precise drug-to-patient matching and patient-disease stratification. We conclude by exploring the challenges of applying a precision approach to cardiology, which arise from a deficit of the required resources and infrastructure, and emerging evidence for the clinical effectiveness of this nascent approach.


eLife | 2017

Genetic identification of a common collagen disease in puerto ricans via identity-by-descent mapping in a health system

Gillian M Belbin; Jacqueline Odgis; Elena P. Sorokin; Muh Ching Yee; Sumita Kohli; Benjamin S. Glicksberg; Christopher R. Gignoux; Genevieve L Wojcik; Tielman Van Vleck; Janina M. Jeff; Michael D. Linderman; Douglas M. Ruderfer; Xiaoqiang Cai; Amanda Merkelson; Anne E. Justice; Kristin L. Young; Misa Graff; Kari E. North; Ulrike Peters; Regina James; Lucia A. Hindorff; Ruth Kornreich; Lisa Edelmann; Omri Gottesman; Eli A. Stahl; Judy H. Cho; Ruth J. F. Loos; Erwin P. Bottinger; Girish N. Nadkarni; Noura S. Abul-Husn

Achieving confidence in the causality of a disease locus is a complex task that often requires supporting data from both statistical genetics and clinical genomics. Here we describe a combined approach to identify and characterize a genetic disorder that leverages distantly related patients in a health system and population-scale mapping. We utilize genomic data to uncover components of distant pedigrees, in the absence of recorded pedigree information, in the multi-ethnic BioMe biobank in New York City. By linking to medical records, we discover a locus associated with both elevated genetic relatedness and extreme short stature. We link the gene, COL27A1, with a little-known genetic disease, previously thought to be rare and recessive. We demonstrate that disease manifests in both heterozygotes and homozygotes, indicating a common collagen disorder impacting up to 2% of individuals of Puerto Rican ancestry, leading to a better understanding of the continuum of complex and Mendelian disease.

Collaboration


Dive into the Benjamin S. Glicksberg's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Khader Shameer

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kipp W. Johnson

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Li Li

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ben Readhead

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Marcus A. Badgeley

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Andrew Kasarskis

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Bin Chen

Michigan State University

View shared research outputs
Top Co-Authors

Avatar

Meng Ma

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge