67
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Towards Personalized Medicine in Rheumatoid Arthritis

& ORCID Icon
Pages 89-114 | Received 04 Jan 2024, Accepted 03 May 2024, Published online: 17 May 2024

References

  • Symmons D, Turner G, Webb R, et al. The prevalence of rheumatoid arthritis in the United Kingdom: new estimates for a new century. Rheumatology. 2002;41(7):793–800. doi:10.1093/rheumatology/41.7.793
  • Chiu YM, Lu YP, Lan JL, Chen DY, Wang JD. Lifetime risks, life expectancy, and health care expenditures for rheumatoid arthritis: a nationwide cohort followed up from 2003 to 2016. Arthritis Rheumatol. 2021;73(5):750–758. doi:10.1002/art.41597
  • Smolen JS, Landewé R, Breedveld FC, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs. Ann Rheum Dis. 2010;69(6):964–975. doi:10.1136/ard.2009.126532
  • Nam JL, Takase-Minegishi K, Ramiro S, et al. Efficacy of biological disease-modifying antirheumatic drugs: a systematic literature review informing the 2016 update of the EULAR recommendations for the management of rheumatoid arthritis. Ann Rheum Dis. 2017;76(6):1113–1136. doi:10.1136/annrheumdis-2016-210713
  • guideline NG100 N. Rheumatoid Arthritis in Adults: Diagnosis and Management. London, UK: National Institute for Health and Care Excellence; 2018.
  • England NH. Improving Outcomes through Personalised Medicine; 2016.
  • Abrahams E. Right drug-right patient-right time: personalized medicine coalition. Clin Transl Sci. 2008;1(1):11–12. doi:10.1111/j.1752-8062.2008.00003.x
  • MacGregor AJ, Snieder H, Rigby AS, et al. Characterizing the quantitative genetic contribution to rheumatoid arthritis using data from twins. Arthritis Rheum. 2000;43(1):30–37. doi:10.1002/1529-0131(200001)43:1<30::Aid-anr5>3.0.Co;2-b
  • Yarwood A, Huizinga TW, Worthington J. The genetics of rheumatoid arthritis: risk and protection in different stages of the evolution of RA. Rheumatology. 2016;55(2):199–209. doi:10.1093/rheumatology/keu323
  • van der Woude D, Houwing-Duistermaat JJ, Toes RE, et al. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum. 2009;60(4):916–923. doi:10.1002/art.24385
  • Doran MF, Pond GR, Crowson CS, O’Fallon WM, Gabriel SE. Trends in incidence and mortality in rheumatoid arthritis in Rochester, Minnesota, over a forty-year period. Arthritis Rheum. 2002;46(3):625–631. doi:10.1002/art.509
  • Symmons DP, Barrett EM, Bankhead CR, Scott DG, Silman AJ. The incidence of rheumatoid arthritis in the United Kingdom: results from the Norfolk Arthritis Register. Br J Rheumatol. 1994;33(8):735–739. doi:10.1093/rheumatology/33.8.735
  • Ma MH, Ibrahim F, Walker D, et al. Remission in early rheumatoid arthritis: predicting treatment response. J Rheumatol. 2012;39(3):470–475. doi:10.3899/jrheum.110169
  • Coulthard LR, Taylor JC, Eyre S, et al. Genetic variants within the MAP kinase signalling network and anti-TNF treatment response in rheumatoid arthritis patients. Ann Rheumatic Dis. 2011;70(1):98–103. doi:10.1136/ard.2010.133249
  • Atzeni F, Bongiovanni S, Marchesoni A, et al. Predictors of response to anti-TNF therapy in RA patients with moderate or high DAS28 scores. Joint Bone Spine. 2014;81(1):37–40. doi:10.1016/j.jbspin.2013.04.005
  • Yamanaka H, Harigai M, Ishiguro N, et al. Trend of patient characteristics and its impact on the response to Adalimumab in patients with rheumatoid arthritis: post hoc time-course analysis of an all-case PMS in Japan. Mod Rheumatol. 2015;25(4):495–502. doi:10.3109/14397595.2014.994263
  • Mancarella L, Bobbio-Pallavicini F, Ceccarelli F, et al. Good clinical response, remission, and predictors of remission in rheumatoid arthritis patients treated with tumor necrosis factor-alpha blockers: the GISEA study. J Rheumatol. 2007;34(8):1670–1673.
  • Levitsky A, Brismar K, Hafström I, et al. Obesity is a strong predictor of worse clinical outcomes and treatment responses in early rheumatoid arthritis: results from the SWEFOT trial. RMD Open. 2017;3(2):e000458. doi:10.1136/rmdopen-2017-000458
  • Hyrich KL, Watson KD, Silman AJ, Symmons DP. Predictors of response to anti-TNF-alpha therapy among patients with rheumatoid arthritis: results from the British society for rheumatology biologics register. Rheumatology. 2006;45(12):1558–1565. doi:10.1093/rheumatology/kel149
  • Fernández-Nebro A, Irigoyen MV, Ureña I, et al. Effectiveness, predictive response factors, and safety of anti-tumor necrosis factor (TNF) therapies in anti-TNF-naive rheumatoid arthritis. J Rheumatol. 2007;34(12):2334–2342.
  • Kleinert S, Tony HP, Krause A, et al. Impact of patient and disease characteristics on therapeutic success during Adalimumab treatment of patients with rheumatoid arthritis: data from a German noninterventional observational study. Rheumatol Int. 2012;32(9):2759–2767. doi:10.1007/s00296-011-2033-5
  • Sugiyama D, Nishimura K, Tamaki K, et al. Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis. 2010;69(1):70–81. doi:10.1136/ard.2008.096487
  • Saevarsdottir S, Wedrén S, Seddighzadeh M, et al. Patients with early rheumatoid arthritis who smoke are less likely to respond to treatment with methotrexate and tumor necrosis factor inhibitors: observations from the epidemiological investigation of rheumatoid arthritis and the Swedish rheumatology register cohorts. Arthritis Rheum. 2011;63(1):26–36. doi:10.1002/art.27758
  • Chatzidionysiou K, Lukina G, Gabay C, et al. Smoking and response to rituximab in rheumatoid arthritis: results from an international European collaboration. Scand J Rheumatol. 2019;48(1):17–23. doi:10.1080/03009742.2018.1466363
  • Sokolove J, Wagner CA, Lahey LJ, et al. Increased inflammation and disease activity among current cigarette smokers with rheumatoid arthritis: a cross-sectional analysis of US veterans. Rheumatology. 2016;55(11):1969–1977. doi:10.1093/rheumatology/kew285
  • Adami G, Fassio A, Pistillo F, et al. Factors associated with radiographic progression in rheumatoid arthritis starting biological diseases modifying anti-rheumatic drugs (bDMARDs). Ther Adv Musculoskelet Dis. 2023;15:1759720x231174534. doi:10.1177/1759720x231174534
  • Kristensen LE, Bliddal H, Christensen R, et al. Is swollen to tender joint count ratio a new and useful clinical marker for biologic drug response in rheumatoid arthritis? Results from a Swedish cohort. Arthritis Care Res. 2014;66(2):173–179. doi:10.1002/acr.22107
  • Maldonado-Montoro M, Cañadas-Garre M, González-Utrilla A, Plaza-Plaza JC, Calleja-Hernández M. Genetic and clinical biomarkers of tocilizumab response in patients with rheumatoid arthritis. Pharmacol Res. 2016;111:264–271. doi:10.1016/j.phrs.2016.06.016
  • Gremese E, Carletto A, Padovan M, et al. Obesity and reduction of the response rate to anti-tumor necrosis factor α in rheumatoid arthritis: an approach to a personalized medicine. Arthritis Care Res. 2013;65(1):94–100. doi:10.1002/acr.21768
  • de Heredia FP, Gómez-Martínez S, Marcos A. Obesity, inflammation and the immune system. Proc Nutr Soc. 2012;71(2):332–338. doi:10.1017/s0029665112000092
  • Jani M, Chinoy H, Warren RB, et al. Clinical utility of random anti-tumor necrosis factor drug-level testing and measurement of antidrug antibodies on the long-term treatment response in rheumatoid arthritis. Arthritis Rheumatol. 2015;67(8):2011–2019. doi:10.1002/art.39169
  • Mould DR. The Pharmacokinetics of Biologics: a Primer. Dig Dis. 2015;33(Suppl 1):61–69. doi:10.1159/000437077
  • Ogata A, Atsumi T, Fukuda T, et al. Sustainable efficacy of switching from intravenous to subcutaneous tocilizumab monotherapy in patients with rheumatoid arthritis. Arthritis Care Res. 2015;67(10):1354–1362. doi:10.1002/acr.22598
  • Hope HF, Bluett J, Barton A, Hyrich KL, Cordingley L, Verstappen SM. Psychological factors predict adherence to methotrexate in rheumatoid arthritis; findings from a systematic review of rates, predictors and associations with patient-reported and clinical outcomes. RMD Open. 2016;2(1):e000171. doi:10.1136/rmdopen-2015-000171
  • Bluett J, Morgan C, Thurston L, et al. Impact of inadequate adherence on response to subcutaneously administered anti-tumour necrosis factor drugs: results from the Biologics in rheumatoid arthritis genetics and genomics study syndicate cohort. Rheumatology. 2015;54(3):494–499. doi:10.1093/rheumatology/keu358
  • National Institute for Health and Care Excellence. NICE guideline [NG33]: tuberculosis; 2023. Available from: https://www.nice.org.uk/guidance/ng33/chapter/Recommendations. Accessed May 4, 2024.
  • Drozdzik M, Rudas T, Pawlik A, Gornik W, Kurzawski M, Herczynska M. Reduced folate carrier-1 80G> A polymorphism affects methotrexate treatment outcome in rheumatoid arthritis. Pharmacogeno J. 2007;7(6):404–407. doi:10.1038/sj.tpj.6500438
  • Li X, Hu M, Li W, et al. The association between reduced folate carrier-1 gene 80G/A polymorphism and methotrexate efficacy or methotrexate related-toxicity in rheumatoid arthritis: a meta-analysis. Int Immunopharmacol. 2016;38:8–15. doi:10.1016/j.intimp.2016.05.012
  • Kung TN, Dennis J, Ma Y, et al. RFC1 80G> A is a genetic determinant of methotrexate efficacy in rheumatoid arthritis: a human genome epidemiologic review and meta‐analysis of observational studies. Arthritis Rheumatol. 2014;66(5):1111–1120. doi:10.1002/art.38331
  • Liu C, Batliwalla F, Li W, et al. Genome-wide association scan identifies candidate polymorphisms associated with differential response to anti-TNF treatment in rheumatoid arthritis. Mol Med. 2008;14(9–10):575–581. doi:10.2119/2008-00056.Liu
  • Padyukov L, Lampa J, Heimburger M, et al. Genetic markers for the efficacy of tumour necrosis factor blocking therapy in rheumatoid arthritis. Ann Rheumatic Dis. 2003;62(6):526. doi:10.1136/ard.62.6.526
  • Krintel SB, Palermo G, Johansen JS, et al. Investigation of single nucleotide polymorphisms and biological pathways associated with response to TNFα inhibitors in patients with rheumatoid arthritis. Pharmacogenet Genomics. 2012;22(8):577–589. doi:10.1097/FPC.0b013e3283544043
  • Acosta-Colman I, Palau N, Tornero J, et al. GWAS replication study confirms the association of PDE3A–SLCO1C1 with anti-TNF therapy response in rheumatoid arthritis. Pharmacogenomics. 2013;14(7):727–734. doi:10.2217/pgs.13.60
  • Plant D, Bowes J, Potter C, et al. Genome-wide association study of genetic predictors of anti-tumor necrosis factor treatment efficacy in rheumatoid arthritis identifies associations with polymorphisms at seven loci. Arthritis Rheum. 2011;63(3):645–653. doi:10.1002/art.30130
  • Márquez A, Ferreiro-Iglesias A, Dávila-Fajardo CL, et al. Lack of validation of genetic variants associated with anti–tumor necrosis factor therapy response in rheumatoid arthritis: a genome-wide association study replication and meta-analysis. Arthritis Res Therapy. 2014;16(2):1–7. doi:10.1186/ar4504
  • Cui J, Saevarsdottir S, Thomson B, et al. Rheumatoid arthritis risk allele PTPRC is also associated with response to anti–tumor necrosis factor α therapy. Arthritis Rheum. 2010;62(7):1849–1861. doi:10.1002/art.27457
  • Plant D, Prajapati R, Hyrich KL, et al. Replication of association of the PTPRC gene with response to anti–tumor necrosis factor therapy in a large UK cohort. Arthritis Rheum. 2012;64(3):665–670. doi:10.1002/art.33381
  • Ferreiro-Iglesias A, Montes A, Perez-Pampin E, et al. Replication of PTPRC as genetic biomarker of response to TNF inhibitors in patients with rheumatoid arthritis. Pharmacogeno J. 2016;16(2):137–140. doi:10.1038/tpj.2015.29
  • Lee YH, Bae SC. Associations between PTPRC rs10919563 A/G and FCGR2A R131H polymorphisms and responsiveness to TNF blockers in rheumatoid arthritis: a meta-analysis. Rheumatol Int. 2016;36(6):837–844. doi:10.1007/s00296-016-3476-5
  • Cui J, Stahl EA, Saevarsdottir S, et al. Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis. PLoS Genet. 2013;9(3):e1003394. doi:10.1371/journal.pgen.1003394
  • Ferreiro-Iglesias A, Montes A, Perez-Pampin E, et al. Evaluation of 12 GWAS-drawn SNPs as biomarkers of rheumatoid arthritis response to TNF inhibitors. A potential SNP association with response to etanercept. PLoS One. 2019;14(2):e0213073. doi:10.1371/journal.pone.0213073
  • Wang J, Bansal A, Martin M, et al. Genome-wide association analysis implicates the involvement of eight loci with response to tocilizumab for the treatment of rheumatoid arthritis. Pharmacogeno J. 2013;13(3):235–241. doi:10.1038/tpj.2012.8
  • International Conference on Harmonisation. Guidance on E15 pharmacogenomics definitions and sample coding; availability. Notice. Fed Regist. 2008;73(68):19074–19076.
  • Massey J, Plant D, Hyrich K, et al. Genome-wide association study of response to tumour necrosis factor inhibitor therapy in rheumatoid arthritis. Pharmacogeno J. 2018;18(5):657–664. doi:10.1038/s41397-018-0040-6
  • Cordingley L, Prajapati R, Plant D, et al. Impact of psychological factors on subjective disease activity assessments in patients with severe rheumatoid arthritis. Arthritis Care Res. 2014;66(6):861–868. doi:10.1002/acr.22249
  • Hensor EMA, McKeigue P, Ling SF, et al. Validity of a two-component imaging-derived disease activity score for improved assessment of synovitis in early rheumatoid arthritis. Rheumatology. 2019;58(8):1400–1409. doi:10.1093/rheumatology/kez049
  • Gilani SS, Nair N, Plant D, et al. Pharmacogenetics of TNF inhibitor response in rheumatoid arthritis utilizing the two-component disease activity score. Pharmacogenomics. 2020;21(16):1151–1156. doi:10.2217/pgs-2020-0043
  • Lopez-Lasanta M, Julià A, Maymó J, et al. Variation at interleukin-6 receptor gene is associated to joint damage in rheumatoid arthritis. Arthritis Res Therapy. 2015;17(1):242. doi:10.1186/s13075-015-0737-8
  • Kastbom A, Cöster L, Arlestig L, et al. Influence of FCGR3A genotype on the therapeutic response to rituximab in rheumatoid arthritis: an observational cohort study. BMJ Open. 2012;2(5):e001524. doi:10.1136/bmjopen-2012-001524
  • Hamada M, Tsunakawa Y, Jeon H, Yadav MK, Takahashi S. Role of MafB in macrophages. Exp Anim. 2020;69(1):1–10. doi:10.1538/expanim.19-0076
  • Lewis CM, Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Med. 2020;12(1):44. doi:10.1186/s13073-020-00742-5
  • Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape. Cell. 2007;128(4):635–638. doi:10.1016/j.cell.2007.02.006
  • Jaenisch R, Bird A. Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nature Genet. 2003;33(3):245–254. doi:10.1038/ng1089
  • Nair N, Plant D, Verstappen SM, et al. Differential DNA methylation correlates with response to methotrexate in rheumatoid arthritis. Rheumatology. 2020;59(6):1364–1371. doi:10.1093/rheumatology/kez411
  • Gosselt HR, van Zelst BD, de Rotte MC, Hazes JM, de Jonge R, Heil SG. Higher baseline global leukocyte DNA methylation is associated with MTX non-response in early RA patients. Arthritis Res Ther. 2019;21(1):1–8. doi:10.1186/s13075-019-1936-5
  • Gosselt HR, Vallerga CL, Mandaviya PR, et al. Epigenome wide association study of response to methotrexate in early rheumatoid arthritis patients. PLoS One. 2021;16(3):e0247709. doi:10.1371/journal.pone.0247709
  • Glossop JR, Nixon NB, Emes RD, et al. DNA methylation at diagnosis is associated with response to disease-modifying drugs in early rheumatoid arthritis. Epigenomics. 2017;9(4):419–428. doi:10.2217/epi-2016-0042
  • Plant D, Webster A, Nair N, et al. Differential methylation as a biomarker of response to etanercept in patients with rheumatoid arthritis. Arthritis Rheumatol. 2016;68(6):1353–1360. doi:10.1002/art.39590
  • Sode J, Krintel SB, Carlsen AL, et al. Plasma microRNA profiles in patients with early rheumatoid arthritis responding to Adalimumab plus methotrexate vs methotrexate alone: a placebo-controlled clinical trial. J Rheumatol. 2018;45(1):53–61. doi:10.3899/jrheum.170266
  • Krintel SB, Dehlendorff C, Hetland ML, et al. Prediction of treatment response to Adalimumab: a double-blind placebo-controlled study of circulating microRNA in patients with early rheumatoid arthritis. Pharmacogeno J. 2016;16(2):141–146. doi:10.1038/tpj.2015.30
  • Cheng P, Wang J. The potential of circulating microRNA‐125a and microRNA‐125b as markers for inflammation and clinical response to infliximab in rheumatoid arthritis patients. J Clin Lab Analysis. 2020;34(8):e23329. doi:10.1002/jcla.23329
  • Duroux-Richard I, Pers Y-M, Fabre S, et al. Circulating miRNA-125b is a potential biomarker predicting response to rituximab in rheumatoid arthritis. Med Inflam. 2014;2014:2. doi:10.1155/2014/342524
  • Liu Y, Han Y, Qu H, Fang J, Ye M, Yin W. Correlation of microRNA expression profile with clinical response to tumor necrosis factor inhibitor in treating rheumatoid arthritis patients: a prospective cohort study. J Clin Lab Analysis. 2019;33(7):e22953. doi:10.1002/jcla.22953
  • Castro-Villegas C, Pérez-Sánchez C, Escudero A, et al. Circulating miRNAs as potential biomarkers of therapy effectiveness in rheumatoid arthritis patients treated with anti-TNFα. Arthritis Res Ther. 2015;17(1):1–15. doi:10.1186/s13075-015-0555-z
  • Cavalli G, Heard E. Advances in epigenetics link genetics to the environment and disease. Nature. 2019;571(7766):489–499. doi:10.1038/s41586-019-1411-0
  • Tessarz P, Kouzarides T. Histone core modifications regulating nucleosome structure and dynamics. Nat Rev Mol Cell Biol. 2014;15(11):703–708. doi:10.1038/nrm3890
  • Nair N, Wilson AG. Assessing the potential of epigenetic targets as biomarkers in the diagnosis and treatment of rheumatoid arthritis. Expert Rev Clin Immunol. 2023;19(5):483–488. doi:10.1080/1744666x.2023.2193686
  • Araki Y, Mimura T. The mechanisms underlying chronic inflammation in rheumatoid arthritis from the perspective of the epigenetic landscape. J Immunol Res. 2016;2016:1. doi:10.1155/2016/6290682
  • Wada TT, Araki Y, Sato K, et al. Aberrant histone acetylation contributes to elevated interleukin-6 production in rheumatoid arthritis synovial fibroblasts. Biochem Biophys Res Commun. 2014;444(4):682–686. doi:10.1016/j.bbrc.2014.01.195
  • Bogunia-Kubik K, Wysoczańska B, Piątek D, Iwaszko M, Ciechomska M, Świerkot J. Significance of polymorphism and expression of miR-146a and NFkB1 genetic variants in patients with rheumatoid arthritis. Archivum Immunol Et Ther Experiment. 2016;64(S1):131–136. doi:10.1007/s00005-016-0443-5
  • Evangelatos G, Fragoulis GE, Koulouri V, Lambrou GI. MicroRNAs in rheumatoid arthritis: from pathogenesis to clinical impact. Autoimmunity Rev. 2019;18(11):102391. doi:10.1016/j.autrev.2019.102391
  • Liu T, Zhang L, Joo D, Sun S-C. NF-κB signaling in inflammation. Signal Transd Target Ther. 2017;2(1):1–9. doi:10.1038/sigtrans.2017.23
  • Lequerré T, Gauthier-Jauneau A-C, Bansard C, et al. Gene profiling in white blood cells predicts infliximab responsiveness in rheumatoid arthritis. Arthritis Res Ther. 2006;8(4):1–11. doi:10.1186/ar1990
  • Tanino M, Matoba R, Nakamura S, et al. Prediction of efficacy of anti-TNF biologic agent, infliximab, for rheumatoid arthritis patients using a comprehensive transcriptome analysis of white blood cells. Biochem Biophys Res Commun. 2009;387(2):261–265. doi:10.1016/j.bbrc.2009.06.149
  • Julià A, Erra A, Palacio C, et al. An eight-gene blood expression profile predicts the response to infliximab in rheumatoid arthritis. PLoS One. 2009;4(10):e7556. doi:10.1371/journal.pone.0007556
  • Stuhlmüller B, Häupl T, Hernandez MM, et al. CD11c as a transcriptional biomarker to predict response to anti-TNF monotherapy with Adalimumab in patients with rheumatoid arthritis. Clin Pharmacol Ther. 2010;87(3):311–321. doi:10.1038/clpt.2009.244
  • van Baarsen LG, Wijbrandts CA, Rustenburg F, et al. Regulation of IFN response gene activity during infliximab treatment in rheumatoid arthritis is associated with clinical response to treatment. Arthritis Res Ther. 2010;12(1):1–10. doi:10.1186/ar2912
  • Toonen EJ, Gilissen C, Franke B, et al. Validation study of existing gene expression signatures for anti-TNF treatment in patients with rheumatoid arthritis. PLoS One. 2012;7(3):e33199. doi:10.1371/journal.pone.0033199
  • Dennis G, Holweg CT, Kummerfeld SK, et al. Synovial phenotypes in rheumatoid arthritis correlate with response to biologic therapeutics. Arthritis Res Ther. 2014;16(2):1–18. doi:10.1186/ar4555
  • Wright HL, Thomas HB, Moots RJ, Edwards SW. Interferon gene expression signature in rheumatoid arthritis neutrophils correlates with a good response to TNFi therapy. Rheumatology. 2015;54(1):188–193. doi:10.1093/rheumatology/keu299
  • Smith SL, Eyre S, Yarwood A, et al. Investigating CD11c expression as a potential genomic biomarker of response to TNF inhibitor biologics in whole blood rheumatoid arthritis samples. Arthritis Res Ther. 2015;17(1):1–7. doi:10.1186/s13075-015-0868-y
  • Oswald M, Curran ME, Lamberth SL, et al. Modular analysis of peripheral blood gene expression in rheumatoid arthritis captures reproducible gene expression changes in tumor necrosis factor responders. Arthritis Rheumatol. 2015;67(2):344–351. doi:10.1002/art.38947
  • Spiliopoulou A, Colombo M, Plant D, et al. Association of response to TNF inhibitors in rheumatoid arthritis with quantitative trait loci for CD40 and CD39. Ann Rheumatic Dis. 2019;78(8):1055–1061. doi:10.1136/annrheumdis-2018-214877
  • Oliver J, Nair N, Orozco G, et al. Transcriptome-wide study of TNF-inhibitor therapy in rheumatoid arthritis reveals early signature of successful treatment. Arthritis Res Therapy. 2021;23(1):80. doi:10.1186/s13075-021-02451-9
  • Cai Y, Xu K, Aihaiti Y, et al. Derlin-1, as a potential early predictive biomarker for nonresponse to infliximab treatment in rheumatoid arthritis, is related to autophagy. Front Immunol. 2022;12:795912. doi:10.3389/fimmu.2021.795912
  • Sanayama Y, Ikeda K, Saito Y, et al. Prediction of therapeutic responses to tocilizumab in patients with rheumatoid arthritis: biomarkers identified by analysis of gene expression in peripheral blood mononuclear cells using genome‐wide DNA microarray. Arthritis Rheumatol. 2014;66(6):1421–1431. doi:10.1002/art.38400
  • Teitsma XM, Jacobs JW, Mokry M, et al. Identification of differential co-expressed gene networks in early rheumatoid arthritis achieving sustained drug-free remission after treatment with a tocilizumab-based or methotrexate-based strategy. Arthritis Res Ther. 2017;19(1):1–10. doi:10.1186/s13075-017-1378-x
  • Yokoyama-Kokuryo W, Yamazaki H, Takeuchi T, et al. Identification of molecules associated with response to Abatacept in patients with rheumatoid arthritis. Arthritis Res Ther. 2020;22(1):1–8. doi:10.1186/s13075-020-2137-y
  • Thurlings RM, Boumans M, Tekstra J, et al. Relationship between the type I interferon signature and the response to rituximab in rheumatoid arthritis patients. Arthritis Rheum. 2010;62(12):3607–3614. doi:10.1002/art.27702
  • Raterman HG, Vosslamber S, de Ridder S, et al. The interferon type I signature towards prediction of non-response to rituximab in rheumatoid arthritis patients. Arthritis Res Ther. 2012;14(2):R95. doi:10.1186/ar3819
  • Sellam J, Marion‐Thore S, Dumont F, et al. Use of whole‐blood transcriptomic profiling to highlight several pathophysiologic pathways associated with response to rituximab in patients with rheumatoid arthritis: data from a randomized, controlled, open‐label trial. Arthritis Rheumatol. 2014;66(8):2015–2025. doi:10.1002/art.38671
  • Nakamura S, Suzuki K, Iijima H, et al. Identification of baseline gene expression signatures predicting therapeutic responses to three biologic agents in rheumatoid arthritis: a retrospective observational study. Arthritis Res Ther. 2016;18(1):1–12. doi:10.1186/s13075-016-1052-8
  • Triaille C, Durez P, Sokolova T, et al. Common transcriptomic effects of Abatacept and other DMARDs on rheumatoid arthritis synovial tissue. Front Immunol. 2021;12:724895. doi:10.3389/fimmu.2021.724895
  • Rivellese F, Nerviani A, Giorli G, et al. Stratification of biological therapies by pathobiology in biologic-naive patients with rheumatoid arthritis (STRAP and STRAP-EU): two parallel, open-label, biopsy-driven, randomised trials. Lancet Rheumatol. 2023;5(11):e648–e659. doi:10.1016/S2665-9913(23)00241-2
  • Humby F, Durez P, Buch MH, et al. Rituximab versus tocilizumab in anti-TNF inadequate responder patients with rheumatoid arthritis (R4RA): 16-week outcomes of a stratified, biopsy-driven, multicentre, open-label, Phase 4 randomised controlled trial. Lancet. 2021;397(10271):305–317. doi:10.1016/S0140-6736(20)32341-2
  • Sobsey CA, Ibrahim S, Richard VR, et al. Targeted and untargeted proteomics approaches in biomarker development. Proteomics. 2020;20(9):1900029. doi:10.1002/pmic.201900029
  • Lee AN, Beck CE, Hall M. Rheumatoid factor and anti-CCP autoantibodies in rheumatoid arthritis: a review. Clin Lab Sci. 2008;21(1):15.
  • Julià A, López-Lasanta M, Blanco F, et al. Interactions between rheumatoid arthritis antibodies are associated with the response to anti-tumor necrosis factor therapy. BMC Musculoskel Disorder. 2021;22(1):1–7. doi:10.1186/s12891-021-04248-y
  • Courvoisier DS, Chatzidionysiou K, Mongin D, et al. The impact of seropositivity on the effectiveness of biologic anti-rheumatic agents: results from a collaboration of 16 registries. Rheumatology. 2020;60(2):820–828. doi:10.1093/rheumatology/keaa393
  • Bird P, Hall S, Nash P, et al. Treatment outcomes in patients with seropositive versus seronegative rheumatoid arthritis in Phase III randomised clinical trials of tofacitinib. RMD Open. 2019;5(1):e000742. doi:10.1136/rmdopen-2018-000742
  • Genovese MC, Fleischmann R, Kivitz A, et al. Efficacy and safety of sarilumab in combination with csDMARDs or as monotherapy in subpopulations of patients with moderately to severely active rheumatoid arthritis in three phase III randomized, controlled studies. Arthritis Res Ther. 2020;22(1):139. doi:10.1186/s13075-020-02194-z
  • Isaacs JD, Cohen SB, Emery P, et al. Effect of baseline rheumatoid factor and anticitrullinated peptide antibody serotype on rituximab clinical response: a meta-analysis. Ann Rheum Dis. 2013;72(3):329–336. doi:10.1136/annrheumdis-2011-201117
  • Halvorsen EH, Haavardsholm EA, Pollmann S, et al. Serum IgG antibodies to peptidylarginine deiminase 4 predict radiographic progression in patients with rheumatoid arthritis treated with tumour necrosis factor-alpha blocking agents. Ann Rheum Dis. 2009;68(2):249–252. doi:10.1136/ard.2008.094490
  • Ferraccioli G, Tolusso B, Bobbio-Pallavicini F, et al. Biomarkers of good EULAR response to the B cell depletion therapy in all seropositive rheumatoid arthritis patients: clues for the pathogenesis. PLoS One. 2012;7(7):e40362. doi:10.1371/journal.pone.0040362
  • de Moel EC, Derksen VFAM, Stoeken G, et al. Baseline autoantibody profile in rheumatoid arthritis is associated with early treatment response but not long-term outcomes. Arthritis Res Ther. 2018;20(1):33. doi:10.1186/s13075-018-1520-4
  • Darrah E, Yu F, Cappelli LC, Rosen A, O’Dell JR, Mikuls TR. Association of baseline peptidylarginine deiminase 4 autoantibodies with favorable response to treatment escalation in rheumatoid arthritis. Arthritis Rheumatol. 2019;71(5):696–702. doi:10.1002/art.40791
  • Lourido L, Ruiz-Romero C, Picchi F, et al. Association of Serum Anti-Centromere Protein F Antibodies with Clinical Response to Infliximab in Patients with Rheumatoid Arthritis: A Prospective Study. Elsevier; 2020:1101–1108.
  • Kumar R, Piantoni S, Boldini M, et al. Anti-carbamylated protein antibodies as a clinical response predictor in rheumatoid arthritis patients treated with Abatacept. Clin Exp Rheumatol. 2021;39(1):91–97. doi:10.55563/clinexprheumatol/g8xqxr
  • Fabre S, Dupuy AM, Dossat N, et al. Protein biochip array technology for cytokine profiling predicts etanercept responsiveness in rheumatoid arthritis. Clin Exp Immunol. 2008;153(2):188–195. doi:10.1111/j.1365-2249.2008.03691.x
  • Trocmé C, Marotte H, Baillet A, et al. Apolipoprotein A-I and platelet factor 4 are biomarkers for infliximab response in rheumatoid arthritis. Ann Rheum Dis. 2009;68(8):1328–1333. doi:10.1136/ard.2008.093153
  • Hueber W, Tomooka BH, Batliwalla F, et al. Blood autoantibody and cytokine profiles predict response to anti-tumor necrosis factor therapy in rheumatoid arthritis. Arthritis Res Ther. 2009;11(3):R76. doi:10.1186/ar2706
  • Fabre S, Guisset C, Tatem L, et al. Protein biochip array technology to monitor rituximab in rheumatoid arthritis. Clin Exp Immunol. 2009;155(3):395–402. doi:10.1111/j.1365-2249.2008.03804.x
  • Hirata S, Marotta A, Gui Y, Hanami K, Tanaka Y. Serum 14-3-3η level is associated with severity and clinical outcomes of rheumatoid arthritis, and its pretreatment level is predictive of DAS28 remission with tocilizumab. Arthritis Res Ther. 2015;17(1):280. doi:10.1186/s13075-015-0799-7
  • Uno K, Yoshizaki K, Iwahashi M, et al. Pretreatment Prediction of individual rheumatoid arthritis patients’ response to anti-cytokine therapy using serum cytokine/chemokine/soluble receptor biomarkers. PLoS One. 2015;10(7):e0132055. doi:10.1371/journal.pone.0132055
  • Obry A, Hardouin J, Lequerré T, et al. Identification of 7 Proteins in sera of RA patients with potential to predict ETA/MTX treatment response. Theranostics. 2015;5(11):1214–1224. doi:10.7150/thno.12403
  • Han BK, Kuzin I, Gaughan JP, Olsen NJ, Bottaro A. Baseline CXCL10 and CXCL13 levels are predictive biomarkers for tumor necrosis factor inhibitor therapy in patients with moderate to severe rheumatoid arthritis: a pilot, prospective study. Arthritis Res Ther. 2016;18:93. doi:10.1186/s13075-016-0995-0
  • Folkersen L, Brynedal B, Diaz-Gallo LM, et al. Integration of known DNA, RNA and protein biomarkers provides prediction of anti-TNF response in rheumatoid arthritis: results from the COMBINE study. Mol Med. 2016;22(1):322–328. doi:10.2119/molmed.2016.00078
  • Rinaudo-Gaujous M, Blasco-Baque V, Miossec P, et al. Infliximab induced a dissociated response of severe periodontal biomarkers in rheumatoid arthritis patients. J Clin Med. 2019;8(5):751. doi:10.3390/jcm8050751
  • Chen J, Tang MS, Xu LC, et al. Proteomic analysis of biomarkers predicting the response to triple therapy in patients with rheumatoid arthritis. Biomed Pharmacother. 2019;116:109026. doi:10.1016/j.biopha.2019.109026
  • Ling SF, Nair N, Verstappen SM, et al. Proteomic analysis to define predictors of treatment response to Adalimumab or methotrexate in rheumatoid arthritis patients. Pharmacogeno J. 2020;20(3):516–523. doi:10.1038/s41397-019-0139-4
  • Zhao J, Ye X, Zhang Z. The predictive value of serum soluble ICAM-1 and CXCL13 in the therapeutic response to TNF inhibitor in rheumatoid arthritis patients who are refractory to csDMARDs. Clin Rheumatol. 2020;39(9):2573–2581. doi:10.1007/s10067-020-05043-1
  • Ling SF, Yap CF, Nair N, et al. A proteomics study of rheumatoid arthritis patients on etanercept identifies putative biomarkers associated with clinical outcome measures. Rheumatology. 2023;(Supplement_2). doi:10.1093/rheumatology/kead321
  • Roessner U, Bowne J. What is metabolomics all about? Biotechniques. 2009;46(5):363–365. doi:10.2144/000113133
  • Gosselt HR, Muller IB, Jansen G, et al. Identification of metabolic biomarkers in relation to methotrexate response in early rheumatoid arthritis. J Pers Med. 2020;10(4):271. doi:10.3390/jpm10040271
  • Medcalf MR, Bantis LE, Shi P, et al. Plasma metabolomic profiling as a tool to identify predictive biomarkers of methotrexate efficacy in rheumatoid arthritis. Semin Arthritis Rheumatism. 2022;56:152056. doi:10.1016/j.semarthrit.2022.152056
  • Kapoor SR, Filer A, Fitzpatrick MA, et al. Metabolic profiling predicts response to anti-tumor necrosis factor α therapy in patients with rheumatoid arthritis. Arthritis Rheum. 2013;65(6):1448–1456. doi:10.1002/art.37921
  • Takahashi S, Saegusa J, Onishi A, Morinobu A. Biomarkers identified by serum metabolomic analysis to predict biologic treatment response in rheumatoid arthritis patients. Rheumatology. 2019;58(12):2153–2161. doi:10.1093/rheumatology/kez199
  • Priori R, Casadei L, Valerio M, Scrivo R, Valesini G, Manetti C. ¹H-NMR-Based metabolomic study for identifying serum profiles associated with the response to etanercept in patients with rheumatoid arthritis. PLoS One. 2015;10(11):e0138537. doi:10.1371/journal.pone.0138537
  • Cuppen BV, Fu J, van Wietmarschen HA, et al. Exploring the inflammatory metabolomic profile to predict response to TNF-α inhibitors in rheumatoid arthritis. PLoS One. 2016;11(9):e0163087. doi:10.1371/journal.pone.0163087
  • Murillo-Saich JD, Diaz-Torne C, Ortiz MA, et al. Metabolomics profiling predicts outcome of tocilizumab in rheumatoid arthritis: an exploratory study. Metabolomics. 2021;17(9):74. doi:10.1007/s11306-021-01822-2
  • Teitsma XM, Yang W, Jacobs JW, et al. Baseline metabolic profiles of early rheumatoid arthritis patients achieving sustained drug-free remission after initiating treat-to-target tocilizumab, methotrexate, or the combination: insights from systems biology. Arthritis Res Ther. 2018;20(1):1–11. doi:10.1186/s13075-018-1729-2
  • Dudka I, Chachaj A, Sebastian A, et al. Metabolomic profiling reveals plasma GlycA and GlycB as a potential biomarkers for treatment efficiency in rheumatoid arthritis. J Pharma Biomed Analysis. 2021;197:113971. doi:10.1016/j.jpba.2021.113971
  • Sweeney SR, Kavanaugh A, Lodi A, et al. Metabolomic profiling predicts outcome of rituximab therapy in rheumatoid arthritis. RMD Open. 2016;2(2):e000289. doi:10.1136/rmdopen-2016-000289
  • Kang J-S, Lee M-H. Overview of therapeutic drug monitoring. Korean J Int Med. 2009;24(1):1. doi:10.3904/kjim.2009.24.1.1
  • Rosas J, Llinares-Tello F, de la Torre I, et al. Clinical relevance of monitoring serum levels of Adalimumab in patients with rheumatoid arthritis in daily practice. Clin Exp Rheumatol. 2014;32(6):942–948.
  • van den Bemt BJ, den Broeder AA, Wolbink GJ, et al. The combined use of disease activity and infliximab serum trough concentrations for early prediction of (non-)response to infliximab in rheumatoid arthritis. Br J Clin Pharmacol. 2013;76(6):939–945. doi:10.1111/bcp.12142
  • Jurado T, Plasencia-Rodríguez C, Martínez-Feito A, et al. Predictive value of serum infliximab levels at induction phase in rheumatoid arthritis patients. Open Rheumatol J. 2017;11:75. doi:10.2174/1874312901711010075
  • Takeuchi T, Miyasaka N, Inoue K, Abe T, Koike T. Impact of trough serum level on radiographic and clinical response to infliximab plus methotrexate in patients with rheumatoid arthritis: results from the RISING study. Mod Rheumatol. 2009;19(5):478–487. doi:10.3109/s10165-009-0195-8
  • Jamnitski A, Krieckaert C, Nurmohamed M, et al. Patients non-responding to etanercept obtain lower etanercept concentrations compared with responding patients. Ann Rheumatic Dis. 2012;71(1):88–91. doi:10.1136/annrheumdis-2011-200184
  • Arad U, Elkayam O. Association of serum tocilizumab trough concentrations with clinical disease activity index scores in adult patients with rheumatoid arthritis. J Rheumatol. 2019;46(12):1577–1581. doi:10.3899/jrheum.181431
  • Kneepkens E, van den Oever I, Plasencia C, et al. Serum tocilizumab trough concentration can be used to monitor systemic IL-6 receptor blockade in patients with rheumatoid arthritis: a prospective observational cohort study. Scand J Rheumatol. 2017;46(2):87–94. doi:10.1080/03009742.2016.1183039
  • Ruscitti P, Sinigaglia L, Cazzato M, et al. Dose adjustments and discontinuation in TNF inhibitors treated patients: when and how. A systematic review of literature. Rheumatology. 2018;57(Supplement_7):vii23–vii31. doi:10.1093/rheumatology/key132
  • Balsa A, Sanmarti R, Rosas J, et al. Drug immunogenicity in patients with inflammatory arthritis and secondary failure to tumour necrosis factor inhibitor therapies: the REASON study. Rheumatology. 2018;57(4):688–693. doi:10.1093/rheumatology/kex474
  • Strand V, Goncalves J, Isaacs JD. Immunogenicity of biologic agents in rheumatology. Nat Rev Rheumatol. 2021;17(2):81–97. doi:10.1038/s41584-020-00540-8
  • Bartelds GM, Krieckaert CL, Nurmohamed MT, et al. Development of antidrug antibodies against Adalimumab and association with disease activity and treatment failure during long-term follow-up. JAMA. 2011;305(14):1460–1468. doi:10.1001/jama.2011.406
  • Ducourau E, Mulleman D, Paintaud G, et al. Antibodies toward infliximab are associated with low infliximab concentration at treatment initiation and poor infliximab maintenance in rheumatic diseases. Arthritis Res Ther. 2011;13(3):1–7. doi:10.1186/ar3386
  • Strand V, Balsa A, Al-Saleh J, et al. Immunogenicity of biologics in chronic inflammatory diseases: a systematic review. BioDrugs. 2017;31(4):299–316. doi:10.1007/s40259-017-0231-8
  • Wells AF, Parrino J, Mangan EK, et al. Immunogenicity of sarilumab monotherapy in patients with rheumatoid arthritis who were inadequate responders or intolerant to disease-modifying antirheumatic drugs. Rheumatol Ther. 2019;6(3):339–352. doi:10.1007/s40744-019-0157-3
  • Xu C, Su Y, Paccaly A, Kanamaluru V. Population pharmacokinetics of sarilumab in patients with rheumatoid arthritis. Clin Pharmacokinet. 2019;58(11):1455–1467. doi:10.1007/s40262-019-00765-1
  • Wincup C, Dunn N, Ruetsch-Chelli C, et al. Anti-rituximab antibodies demonstrate neutralizing capacity, associate with lower circulating drug levels and earlier relapse in lupus. Rheumatology. 2022;62(7):2601–2610. doi:10.1093/rheumatology/keac608
  • Moots RJ, Xavier RM, Mok CC, et al. The impact of anti-drug antibodies on drug concentrations and clinical outcomes in rheumatoid arthritis patients treated with Adalimumab, etanercept, or infliximab: results from a multinational, real-world clinical practice, non-interventional study. PLoS One. 2017;12:4.
  • Maid PJ, Xavier R, Real RM, et al. Incidence of antidrug antibodies in rheumatoid arthritis patients from Argentina treated with Adalimumab, etanercept, or infliximab in a real-world setting. JCR. 2018;24(4):177–182.
  • Quistrebert J, Hässler S, Bachelet D, et al. Incidence and risk factors for Adalimumab and infliximab anti-drug antibodies in rheumatoid arthritis: a European retrospective multicohort analysis. Semin Arthritis Rheum. 2019;48(6):967–975. doi:10.1016/j.semarthrit.2018.10.006
  • Burmester GR, Choy E, Kivitz A, et al. Low immunogenicity of tocilizumab in patients with rheumatoid arthritis. Ann Rheum Dis. 2017;76(6):1078–1085. doi:10.1136/annrheumdis-2016-210297
  • Sigaux J, Hamze M, Daien C, et al. Immunogenicity of tocilizumab in patients with rheumatoid arthritis. Joint Bone Spine. 2017;84(1):39–45. doi:10.1016/j.jbspin.2016.04.013
  • Genovese MC, Fleischmann R, Kivitz AJ, et al. Sarilumab plus methotrexate in patients with active rheumatoid arthritis and inadequate response to methotrexate: results of a phase III study. Arthritis Rheumatol. 2015;67(6):1424–1437. doi:10.1002/art.39093
  • Thurlings RM, Teng O, Vos K, et al. Clinical response, pharmacokinetics, development of human anti-chimaeric antibodies, and synovial tissue response to rituximab treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 2010;69(2):409–412. doi:10.1136/ard.2009.109041
  • Bitoun S, Hässler S, Ternant D, et al. Response to biologic drugs in patients with rheumatoid arthritis and antidrug antibodies. JAMA Network Open. 2023;6(7):e2323098–e2323098. doi:10.1001/jamanetworkopen.2023.23098
  • Jamnitski A, Bartelds GM, Nurmohamed MT, et al. The presence or absence of antibodies to infliximab or Adalimumab determines the outcome of switching to etanercept. Ann Rheum Dis. 2011;70(2):284–288. doi:10.1136/ard.2010.135111
  • Ucar E, Gorostiza Í, Gόmez C, et al. SAT0150 Prospective, Intervention, Multicenter Study of Utility of Biologic Drug Monitoring with Respect to the Efficacy and Cost of Adalimumab Tapering in Patients with Rheumatic Diseases: Preliminary Results of INGEBIO Study. BMJ Publishing Group Ltd; 2017.
  • Syversen SW, Goll GL, Jørgensen KK, et al. Therapeutic drug monitoring of infliximab compared to standard clinical treatment with infliximab: study protocol for a randomised, controlled, open, parallel-group, Phase IV study (the NOR-DRUM study). Trials. 2020;21(1):13. doi:10.1186/s13063-019-3734-4
  • (NICE) NIfHaCE. Therapeutic monitoring of TNF-alpha inhibitors in rheumatoid arthritis. Available from: https://www.nice.org.uk/guidance/dg36. Accessed December 19, 2023.
  • Krieckaert CL, van Tubergen A, Gehin JE, et al. EULAR points to consider for therapeutic drug monitoring of biopharmaceuticals in inflammatory rheumatic and musculoskeletal diseases. Ann Rheumatic Dis. 2023;82(1):65–73. doi:10.1136/annrheumdis-2022-222155
  • Johnson KB, Wei WQ, Weeraratne D, et al. Precision medicine, AI, and the future of personalized health care. Clin Transl Sci. 2021;14(1):86–93. doi:10.1111/cts.12884
  • Deo RC. Machine learning in medicine. Circulation. 2015;132(20):1920–1930. doi:10.1161/CIRCULATIONAHA.115.001593
  • Mirza B, Wang W, Wang J, Choi H, Chung NC, Ping P. Machine learning and integrative analysis of biomedical big data. Genes. 2019;10(2):87. doi:10.3390/genes10020087
  • Westerlind H, Maciejewski M, Frisell T, Jelinsky SA, Ziemek D, Askling J. What Is the persistence to methotrexate in rheumatoid arthritis, and does machine learning outperform hypothesis-based approaches to its prediction? ACR Open Rheumatol. 2021;3(7):457–463. doi:10.1002/acr2.11266
  • Miyoshi F, Honne K, Minota S, Okada M, Ogawa N, Mimura T. A novel method predicting clinical response using only background clinical data in RA patients before treatment with infliximab. Mod Rheumatol. 2016;26(6):813–816. doi:10.3109/14397595.2016.1168536
  • Koo BS, Eun S, Shin K, et al. Machine learning model for identifying important clinical features for predicting remission in patients with rheumatoid arthritis treated with biologics. Arthritis Res Ther. 2021;23(1):178. doi:10.1186/s13075-021-02567-y
  • Gosselt HR, Verhoeven MMA, Bulatović-ćalasan M, et al. Complex machine-learning algorithms and multivariable logistic regression on par in the prediction of insufficient clinical response to methotrexate in rheumatoid arthritis. J Pers Med. 2021;11(1):44. doi:10.3390/jpm11010044
  • Lim AJ, Lim LJ, Ooi BN, et al. Functional coding haplotypes and machine-learning feature elimination identifies predictors of methotrexate response in rheumatoid arthritis patients. EBioMedicine. 2022;3:75.
  • Plant D, Maciejewski M, Smith S, et al. Profiling of gene expression biomarkers as a classifier of methotrexate nonresponse in patients with rheumatoid arthritis. Arthritis Rheumatol. 2019;71(5):678–684. doi:10.1002/art.40810
  • Guan Y, Zhang H, Quang D, et al. Machine learning to predict anti-tumor necrosis factor drug responses of rheumatoid arthritis patients by integrating clinical and genetic markers. Arthritis Rheumatol. 2019;71(12):1987–1996. doi:10.1002/art.41056
  • Luque-Tévar M, Perez-Sanchez C, Patiño-Trives AM, et al. Integrative clinical, molecular, and computational analysis identify novel biomarkers and differential profiles of anti-TNF response in rheumatoid arthritis. Front Immunol. 2021;12:631662. doi:10.3389/fimmu.2021.631662
  • Yoosuf N, Maciejewski M, Ziemek D, et al. Early prediction of clinical response to anti-TNF treatment using multi-omics and machine learning in rheumatoid arthritis. Rheumatology. 2022;61(4):1680–1689. doi:10.1093/rheumatology/keab521
  • Tao W, Concepcion AN, Vianen M, et al. Multiomics and machine learning accurately predict clinical response to Adalimumab and etanercept therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2021;73(2):212–222. doi:10.1002/art.41516
  • Tasaki S, Suzuki K, Kassai Y, et al. Multi-omics monitoring of drug response in rheumatoid arthritis in pursuit of molecular remission. Nat Commun. 2018;9(1):2755. doi:10.1038/s41467-018-05044-4
  • Jung SM, Park K-S, Kim K-J. Deep phenotyping of synovial molecular signatures by integrative systems analysis in rheumatoid arthritis. Rheumatology. 2021;60(7):3420–3431. doi:10.1093/rheumatology/keaa751
  • MacEachern SJ, Forkert ND. Machine learning for precision medicine. Genome. 2021;64(4):416–425. doi:10.1139/gen-2020-0131