Assessment of Variability in the SOMAscan Assay

SOMAscan is an aptamer-based proteomics assay capable of measuring 1,305 human protein analytes in serum, plasma, and other biological matrices with high sensitivity and specificity. In this work, we present a comprehensive meta-analysis of performance based on multiple serum and plasma runs using the current 1.3 k assay, as well as the previous 1.1 k version. We discuss normalization procedures and examine different strategies to minimize intra- and interplate nuisance effects. We implement a meta-analysis based on calibrator samples to characterize the coefficient of variation and signal-over-background intensity of each protein analyte. By incorporating coefficient of variation estimates into a theoretical model of statistical variability, we also provide a framework to enable rigorous statistical tests of significance in intervention studies and clinical trials, as well as quality control within and across laboratories. Furthermore, we investigate the stability of healthy subject baselines and determine the set of analytes that exhibit biologically stable baselines after technical variability is factored in. This work is accompanied by an interactive web-based tool, an initiative with the potential to become the cornerstone of a regularly updated, high quality repository with data sharing, reproducibility, and reusability as ultimate goals.

[1]  M. Croft The role of TNF superfamily members in T-cell function and diseases , 2009, Nature Reviews Immunology.

[2]  Sandra Romero-Steiner,et al.  Molecular signatures of antibody responses derived from a systems biological study of 5 human vaccines , 2013, Nature Immunology.

[3]  K. Davies,et al.  Identification of serum protein biomarkers for utrophin based DMD therapy , 2017, Scientific Reports.

[4]  Yuri Kotliarov,et al.  Global Analyses of Human Immune Variation Reveal Baseline Predictors of Postvaccination Responses , 2014, Cell.

[5]  B. Aggarwal Signalling pathways of the TNF superfamily: a double-edged sword , 2003, Nature Reviews Immunology.

[6]  Sheri K. Wilcox,et al.  Protein Signature of Lung Cancer Tissues , 2012, PloS one.

[7]  Magda Tsolaki,et al.  Candidate blood proteome markers of Alzheimer's disease onset and progression: a systematic review and replication study. , 2013, Journal of Alzheimer's disease : JAD.

[8]  Mark R Segal,et al.  Development and Validation of a Protein-Based Risk Score for Cardiovascular Outcomes Among Patients With Stable Coronary Heart Disease. , 2016, JAMA.

[9]  Hugues Bersini,et al.  Batch effect removal methods for microarray gene expression data integration: a survey , 2013, Briefings Bioinform..

[10]  Alternative ways of estimating serological titer reproducibility , 1981, Journal of clinical microbiology.

[11]  Stephen A. Williams,et al.  Unlocking Biomarker Discovery: Large Scale Application of Aptamer Proteomic Technology for Early Detection of Lung Cancer , 2010, PloS one.

[12]  Yudi Pawitan,et al.  Multidimensional Normalization to Minimize Plate Effects of Suspension Bead Array Data. , 2016, Journal of proteome research.

[13]  Larry Gold,et al.  Nucleic Acid Ligands With Protein-like Side Chains: Modified Aptamers and Their Use as Diagnostic and Therapeutic Agents , 2014, Molecular therapy. Nucleic acids.

[14]  M. Ruhwald,et al.  IP-10 measured by Dry Plasma Spots as biomarker for therapy responses in Mycobacterium Tuberculosis infection , 2015, Scientific Reports.

[15]  Xu Shi,et al.  Aptamer-Based Proteomic Profiling Reveals Novel Candidate Biomarkers and Pathways in Cardiovascular Disease , 2016, Circulation.

[16]  Richard T. Lee,et al.  Growth Differentiation Factor 11 Is a Circulating Factor that Reverses Age-Related Cardiac Hypertrophy , 2013, Cell.

[17]  George F. Reed,et al.  Use of Coefficient of Variation in Assessing Variability of Quantitative Assays , 2002, Clinical and Vaccine Immunology.

[18]  Christian Gieger,et al.  Connecting genetic risk to disease end points through the human blood plasma proteome , 2016, Nature Communications.

[19]  Terence P. Speed,et al.  A comparison of normalization methods for high density oligonucleotide array data based on variance and bias , 2003, Bioinform..

[20]  Fredrik Levander,et al.  Normalyzer: A Tool for Rapid Evaluation of Normalization Methods for Omics Data Sets , 2014, Journal of proteome research.

[21]  R. J. Wood,et al.  Reproducibility of serological titers , 1980, Journal of clinical microbiology.

[22]  Joel Dudley,et al.  High-Throughput Characterization of Blood Serum Proteomics of IBD Patients with Respect to Aging and Genetic Factors , 2017, PLoS genetics.

[23]  M. Sabatine Using Aptamer-Based Technology to Probe the Plasma Proteome for Cardiovascular Disease Prediction. , 2016, JAMA.

[24]  Stephen A. Williams,et al.  Validation of a blood protein signature for non-small cell lung cancer , 2014, Clinical Proteomics.

[25]  Jean-Pierre A. Kocher,et al.  A new statistic for identifying batch effects in high-throughput genomic data that uses guided principal component analysis , 2013, Bioinform..

[26]  Virginia Pascual,et al.  A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. , 2008, Immunity.

[27]  Magda Tsolaki,et al.  Alzheimer's disease biomarker discovery using SOMAscan multiplexed protein technology , 2014, Alzheimer's & Dementia.

[28]  Chunyu Liu,et al.  Removing Batch Effects in Analysis of Expression Microarray Data: An Evaluation of Six Batch Adjustment Methods , 2011, PloS one.

[29]  Tracy R. Keeney,et al.  Aptamer-based multiplexed proteomic technology for biomarker discovery , 2010, Nature Precedings.

[30]  Yuri Kotliarov,et al.  Web Tool for Navigating and Plotting SomaLogic ADAT Files , 2017, Journal of open research software.