Network


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

Hotspot


Dive into the research topics where Stuart A. Scott is active.

Publication


Featured researches published by Stuart A. Scott.


The Lancet Psychiatry | 2016

Polygenic overlap between schizophrenia risk and antipsychotic response: a genomic medicine approach

Douglas M. Ruderfer; Alexander Charney; Ben Readhead; Brian A. Kidd; Anna K. Kähler; Paul J Kenny; Michael J. Keiser; Jennifer L. Moran; Christina M. Hultman; Stuart A. Scott; Patrick F. Sullivan; Shaun Purcell; Joel T. Dudley; Pamela Sklar

BACKGROUNDnTherapeutic treatments for schizophrenia do not alleviate symptoms for all patients and efficacy is limited by common, often severe, side-effects. Genetic studies of disease can identify novel drug targets, and drugs for which the mechanism has direct genetic support have increased likelihood of clinical success. Large-scale genetic studies of schizophrenia have increased the number of genes and gene sets associated with risk. We aimed to examine the overlap between schizophrenia risk loci and gene targets of a comprehensive set of medications to potentially inform and improve treatment of schizophrenia.nnnMETHODSnWe defined schizophrenia risk loci as genomic regions reaching genome-wide significance in the latest Psychiatric Genomics Consortium schizophrenia genome-wide association study (GWAS) of 36u2008989 cases and 113u2008075 controls and loss of function variants observed only once among 5079 individuals in an exome-sequencing study of 2536 schizophrenia cases and 2543 controls (Swedish Schizophrenia Study). Using two large and orthogonally created databases, we collated drug targets into 167 gene sets targeted by pharmacologically similar drugs and examined enrichment of schizophrenia risk loci in these sets. We further linked the exome-sequenced data with a national drug registry (the Swedish Prescribed Drug Register) to assess the contribution of rare variants to treatment response, using clozapine prescription as a proxy for treatment resistance.nnnFINDINGSnWe combined results from testing rare and common variation and, after correction for multiple testing, two gene sets were associated with schizophrenia risk: agents against amoebiasis and other protozoal diseases (106 genes, p=0·00046, pcorrected =0·024) and antipsychotics (347 genes, p=0·00078, pcorrected=0·046). Further analysis pointed to antipsychotics as having independent enrichment after removing genes that overlapped these two target sets. We noted significant enrichment both in known targets of antipsychotics (70 genes, p=0·0078) and novel predicted targets (277 genes, p=0·019). Patients with treatment-resistant schizophrenia had an excess of rare disruptive variants in gene targets of antipsychotics (347 genes, p=0·0067) and in genes with evidence for a role in antipsychotic efficacy (91 genes, p=0·0029).nnnINTERPRETATIONnOur results support genetic overlap between schizophrenia pathogenesis and antipsychotic mechanism of action. This finding is consistent with treatment efficacy being polygenic and suggests that single-target therapeutics might be insufficient. We provide evidence of a role for rare functional variants in antipsychotic treatment response, pointing to a subset of patients where their genetic information could inform treatment. Finally, we present a novel framework for identifying treatments from genetic data and improving our understanding of therapeutic mechanism.nnnFUNDINGnUS National Institutes of Health.


Clinical Pharmacology & Therapeutics | 2018

Multisite Investigation of Strategies for the Implementation of CYP2C19 Genotype‐Guided Antiplatelet Therapy

Philip E. Empey; James M. Stevenson; Sony Tuteja; Kristin Weitzel; Dominick J. Angiolillo; Amber L. Beitelshees; James C. Coons; Julio D. Duarte; Francesco Franchi; Linda Jo Bone Jeng; Julie A. Johnson; Rolf P. Kreutz; Nita A. Limdi; Kristin A. Maloney; Aniwaa Owusu Obeng; Josh F. Peterson; Natasha Petry; Victoria M. Pratt; Fabiana Rollini; Stuart A. Scott; Todd C. Skaar; Mark R. Vesely; George A. Stouffer; Russell A. Wilke; Larisa H. Cavallari; Craig R. Lee

CYP2C19 genotype‐guided antiplatelet therapy following percutaneous coronary intervention is increasingly implemented in clinical practice. However, challenges such as selecting a testing platform, communicating test results, building clinical decision support processes, providing patient and provider education, and integrating methods to support the translation of emerging evidence to clinical practice are barriers to broad adoption. In this report, we compare and contrast implementation strategies of 12 early adopters, describing solutions to common problems and initial performance metrics for each program. Key differences between programs included the test result turnaround time and timing of therapy changes, which are both related to the CYP2C19 testing model and platform used. Sites reported the need for new informatics infrastructure, expert clinicians such as pharmacists to interpret results, physician champions, and ongoing education. Consensus lessons learned are presented to provide a path forward for those seeking to implement similar clinical pharmacogenomics programs within their institutions.


Pharmacogenomics | 2017

Institutional profile: translational pharmacogenomics at the Icahn School of Medicine at Mount Sinai

Stuart A. Scott; Aniwaa Owusu Obeng; Mariana R Botton; Yao Yang; Erick R. Scott; Stephen Ellis; Richard Wallsten; Tom Kaszemacher; Xiang Zhou; Rong Chen; Paola Nicoletti; Hetanshi Naik; Eimear E. Kenny; Aida Vega; Eva Waite; George A. Diaz; Joel T. Dudley; Jonathan L. Halperin; Lisa Edelmann; Andrew Kasarskis; Jean-Sébastien Hulot; Inga Peter; Erwin P. Bottinger; Kurt Hirschhorn; Pamela Sklar; Judy H. Cho; Robert J. Desnick; Eric E. Schadt

For almost 50xa0years, the Icahn School of Medicine at Mount Sinai has continually invested in genetics and genomics, facilitating a healthy ecosystem that provides widespread support for the ongoing programs in translational pharmacogenomics. These programs can be broadly cataloged into discovery, education, clinical implementation and testing, which are collaboratively accomplished by multiple departments, institutes, laboratories, companies and colleagues. Focus areas have included drug response association studies and allele discovery, multiethnic pharmacogenomics, personalized genotyping and survey-based education programs, pre-emptive clinical testing implementation and novel assay development. This overview summarizes the current state of translational pharmacogenomics at Mount Sinai, including a future outlook on the forthcoming expansions in overall support, research and clinical programs, genomic technology infrastructure and the participating faculty.


The Journal of Molecular Diagnostics | 2018

Recommendations for Clinical CYP2C19 Genotyping Allele Selection: A Report of the Association for Molecular Pathology

Victoria M. Pratt; Andria L. Del Tredici; Houda Hachad; Yuan Ji; Lisa Kalman; Stuart A. Scott; Karen E. Weck

This document was developed by the Pharmacogenomics (PGx) Working Group of the Association for Molecular Pathology Clinical Practice Committee, whose aim is to recommend variants for inclusion in clinical pharmacogenomic testing panels. The goals of the Association for Molecular Pathology PGx Working Group are to define the key attributes of PGx alleles recommended for clinical testing and to define a minimum set of variants that should be included in clinical PGx genotyping assays. These recommendations include a minimum panel of variant alleles (tier 1) and an extended panel of variant alleles (tier 2) that will aid clinical laboratories when designing PGx assays. The Working Group considered variant allele frequencies in different populations and ethnicities, the availability of reference materials, and other technical considerations for PGx testing when developing these recommendations. These CYP2C19 genotyping recommendations are the first of a series of recommendations for PGx testing. These recommendations are not to be interpreted as restrictive, but they are meant to provide a helpful guide.


npj Genomic Medicine | 2016

A de novo 2.78-Mb duplication on chromosome 21q22.11 implicates candidate genes in the partial trisomy 21 phenotype

James D Weisfeld-Adams; Amanda K Tkachuk; Kenneth N Maclean; Naomi L Meeks; Stuart A. Scott

Down syndrome (DS) is the most common genetic cause of intellectual disability (ID) and in the majority of cases is the result of complete trisomy 21. The hypothesis that the characteristic DS clinical features are due to a single DS critical region (DSCR) at distal chromosome 21q has been refuted by recently reported segmental trisomy 21 cases characterised by microarray-based comparative genomic hybridisation (aCGH). These rare cases have implicated multiple regions on chromosome 21 in the aetiology of distinct features of DS; however, the map of chromosome 21 copy-number aberrations and their associated phenotypes remains incompletely defined. We report a child with ID who was deemed very high risk for DS on antenatal screening (1 in 13) and has partial, but distinct, dysmorphologic features of DS without congenital heart disease (CHD). Oligonucleotide aCGH testing of the proband detected a previously unreported de novo 2.78-Mb duplication on chromosome 21q22.11 that includes 16 genes; however, this aberration does not harbour any of the historical DSCR genes (APP, DSCR1, DYRK1A and DSCAM). This informative case implicates previously under-recognised candidate genes (SOD1, SYNJ1 and ITSN1) in the pathogenesis of specific DS clinical features and supports a critical region for CHD located more distal on chromosome 21q. In addition, this unique case illustrates how the increasing resolution of microarray and high-throughput sequencing technologies can continue to reveal new biology and enhance understanding of widely studied genetic diseases that were originally described over 50 years ago.


npj Genomic Medicine | 2018

Cytogenomic identification and long-read single molecule real-time (SMRT) sequencing of a Bardet–Biedl Syndrome 9 ( BBS9 ) deletion

Jennifer Reiner; Laura Pisani; Wanqiong Qiao; Ram Singh; Yao Yang; Lisong Shi; Wahab A. Khan; Robert Sebra; Ninette Cohen; Arvind Babu; Lisa Edelmann; Ethylin Wang Jabs; Stuart A. Scott

Bardet–Biedl syndrome (BBS) is a recessive disorder characterized by heterogeneous clinical manifestations, including truncal obesity, rod-cone dystrophy, renal anomalies, postaxial polydactyly, and variable developmental delays. At least 20 genes have been implicated in BBS, and all are involved in primary cilia function. We report a 1-year-old male child from Guyana with obesity, postaxial polydactyly on his right foot, hypotonia, ophthalmologic abnormalities, and developmental delay, which together indicated a clinical diagnosis of BBS. Clinical chromosomal microarray (CMA) testing and high-throughput BBS gene panel sequencing detected a homozygous 7p14.3 deletion of exons 1–4 of BBS9 that was encompassed by a 17.5u2009Mb region of homozygosity at chromosome 7p14.2–p21.1. The precise breakpoints of the deletion were delineated to a 72.8u2009kb region in the proband and carrier parents by third-generation long-read single molecule real-time (SMRT) sequencing (Pacific Biosciences), which suggested non-homologous end joining as a likely mechanism of formation. Long-read SMRT sequencing of the deletion breakpoints also determined that the aberration included the neighboring RP9 gene implicated in retinitis pigmentosa; however, the clinical significance of this was considered uncertain given the paucity of reported cases with unambiguous RP9 mutations. Taken together, our study characterized a BBS9 deletion, and the identification of this shared haplotype in the parents suggests that this pathogenic aberration may be a BBS founder mutation in the Guyanese population. Importantly, this informative case also highlights the utility of long-read SMRT sequencing to map nucleotide breakpoints of clinically relevant structural variants.


Pharmacogenomics | 2018

Multi-ethnic SULT1A1 copy number profiling with multiplex ligation-dependent probe amplification

Raymon Vijzelaar; Mariana R Botton; Lisette Stolk; Suparna Martis; Robert J. Desnick; Stuart A. Scott

AIMnTo develop a SULT1A1 multiplex ligation-dependent probe amplification assay and to investigate multi-ethnic copy number variant frequencies.nnnMETHODSnA novel multiplex ligation-dependent probe amplification assay was developed and tested on 472 African-American, Asian, Caucasian, Hispanic and Ashkenazi Jewish individuals.nnnRESULTSnThe frequencies of atypical total copy number (i.e., greater or less than two) were 38.7% for Hispanics, 38.9% for Ashkenazi Jewish, 43.2% for Caucasians, 53.6% for Asians and 64.1% for African-Americans. Heterozygous SULT1A1 deletion carriers (slow sulfators) were most common among Caucasians (8.4%), whereas African-Americans had the highest frequencies of three or more copies (rapid sulfators; 60.9%).nnnCONCLUSIONnDifferent ethnic and racial populations have varying degrees of SULT1A1-mediated sulfation activity, which warrants further research and that may have utility for drug response prediction among SULT1A1-metabolized medications.


Archive | 2008

Molecular Medical Genetics

Lisa Edelmann; Stuart A. Scott; Ruth Kornreich

Mode of inheritance is important for predicting clinical status of individuals carrying mutations and for risk assessment of family members of a patient affected with a genetic disease. Modes include: n n nAutosomal recessive (AR) n n nAutosomal dominant (AD) n n nX-linked n n nMitochondrial


PMC | 2016

Characterization of 137 Genomic DNA Reference Materials for 28 Pharmacogenetic Genes: A GeT-RM Collaborative Project

Victoria M. Pratt; Robin E. Everts; Praful Aggarwal; Brittany N. Beyer; Ulrich Broeckel; Ruth Epstein-Baak; Paul Hujsak; Ruth Kornreich; Jun Liao; Rachel Lorier; Stuart A. Scott; Chingying Smith; Lorraine Toji; Amy Turner; Lisa Kalman


CRI | 2016

The Clinical Pharmacogenetics Implementation Consortium (CPIC) Term Standardization for Clinical Pharmacogenetic Test Results: Alleles and Phenotypes.

James M. Hoffman; Henry M. Dunnenberger; Jonathan D. Burlison; Michelle Whirl Carrillo; Robert R. Freimuth; Marc S. Williams; Stuart A. Scott; Josh F. Peterson; Heidi L. Rehm; Teri E. Klein; Mary V. Relling; Kelly E. Caudle

Collaboration


Dive into the Stuart A. Scott's collaboration.

Top Co-Authors

Avatar

Lisa Edelmann

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joel T. Dudley

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa Kalman

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Pamela Sklar

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Yao Yang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aida Vega

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge