PT - JOURNAL ARTICLE AU - Hur, Benjamin AU - Gupta, Vinod K. AU - Huang, Harvey AU - Wright, Kerry A. AU - Warrington, Kenneth J. AU - Taneja, Veena AU - Davis, John M. AU - Sung, Jaeyun TI - Plasma Metabolomic Profiling in Patients with Rheumatoid Arthritis Identifies Biochemical Features Predictive of Quantitative Disease Activity AID - 10.1101/2020.09.13.20193664 DP - 2021 Jan 01 TA - medRxiv PG - 2020.09.13.20193664 4099 - http://medrxiv.org/content/early/2021/03/25/2020.09.13.20193664.short 4100 - http://medrxiv.org/content/early/2021/03/25/2020.09.13.20193664.full AB - Background Rheumatoid arthritis (RA) is a chronic, autoimmune disorder characterized by joint inflammation and pain. In patients with RA, metabolomic approaches, i.e., high-throughput profiling of small-molecule metabolites, on plasma or serum has thus far enabled the discovery of biomarkers for clinical subgroups, risk factors, and predictors of treatment response. Despite these recent advancements, the identification of blood metabolites that reflect quantitative disease activity remains an important challenge in precision medicine for RA. Herein, we use global plasma metabolomic profiling analyses to detect metabolites associated with, and predictive of, quantitative disease activity in patients with RA.Methods Ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was performed on a discovery cohort consisting of 128 plasma samples from 64 RA patients, and on a validation cohort of 12 samples from 12 patients. The resulting metabolomic profiles were analyzed with two different strategies to find metabolites associated with RA disease activity defined by the Disease Activity Score-28 using C-reactive protein (DAS28-CRP). More specifically, mixed-effects regression models were used to identify metabolites differentially abundant between two disease activity groups (‘lower’, DAS28-CRP ≤ 3.2; and ‘higher’, DAS28-CRP > 3.2); and to identify metabolites significantly associated with DAS28-CRP scores. A generalized linear model (GLM) was then constructed for estimating DAS28-CRP using plasma metabolite abundances. Finally, for associating metabolites with CRP (an indicator of inflammation), metabolites differentially abundant between two patient groups (‘low-CRP’, CRP ≤ 3.0 mg/L; ‘high-CRP’, CRP > 3.0 mg/L) were investigated.Results We identified 33 metabolites differentially abundant between lower and higher disease activity groups (P < 0.05). Additionally, we identified 51 metabolites associated with DAS28-CRP (P < 0.05). A GLM based upon these 51 metabolites resulted in higher prediction accuracy (mean absolute error [MAE]±SD: 1.51±1.77) compared to a GLM without feature selection (MAE±SD: 2.02±2.21). The predictive value of this feature set was further demonstrated on a validation cohort of twelve plasma samples, wherein we observed a stronger correlation between predicted vs. actual DAS28-CRP (with feature selection: Spearman’s ρ = 0.69, 95% CI: [0.18, 0.90]; without feature selection: Spearman’s ρ = 0.18, 95% CI: [-0.44, 0.68]). Lastly, among all identified metabolites, the abundances of eight were significantly associated with CRP patient groups while controlling for potential confounders (P < 0.05).Conclusions We demonstrate for the first time the prediction of quantitative disease activity in RA using plasma metabolomes. The metabolites identified herein provide insight into circulating pro-/anti-inflammatory metabolic signatures that reflect disease activity and inflammatory status in RA patients.Competing Interest StatementThe authors have declared no competing interest.Funding StatementThis work was supported in part by the Mayo Clinic Center for Individualized Medicine (to B.H., V.K.G., and J.S.), and Mark E. and Mary A. Davis to Mayo Clinic Center for Individualized Medicine (J.M.D. and J.S.).Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:The study was approved by the Mayo Clinic institutional review board (no. 14-000616 and no. 14-000680).All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesRaw metabolomic datasets, as well as source codes used to reproduce the results in this study, are available at https://github.com/jaeyunsung/RA_plasma_metabolomics_2020. https://github.com/jaeyunsung/RA_plasma_metabolomics_2020 RARheumatoid arthritisDMARDsDisease-modifying anti-rheumatic drugsbDMARDsBiologic disease-modifying anti-rheumatic drugscsDMARDsConventional synthetic disease-modifying anti-rheumatic drugsUPLC-MS/MSUltra-high performance liquid chromatography-tandem mass spectrometryDAS28-CRPDisease Activity Score-28 using C-reactive proteinBMIBody mass indexGLMGeneralized linear modelMAEMean absolute errorSDStandard deviationESRErythrocyte sedimentation rateCRPC-reactive proteinAnti-CCPanti-cyclic citrullinated peptide antibodiesHMDBHuman metabolome databaseCIACollagen-induced arthritisCDAIClinical disease activity indexSDAISimple disease activity index