250
Views
1
CrossRef citations to date
0
Altmetric
REVIEW

Potential Mechanism of Fatigue Induction and Its Management by JAK Inhibitors in Inflammatory Rheumatic Diseases

ORCID Icon, ORCID Icon, , ORCID Icon & ORCID Icon
Pages 3949-3965 | Received 28 Mar 2023, Accepted 27 Jul 2023, Published online: 08 Sep 2023

References

  • Surowiec I, Gjesdal CG, Jonsson G, et al. Metabolomics study of fatigue in patients with rheumatoid arthritis naive to biological treatment. Rheumatol Int. 2016;36(5):703–711. doi:10.1007/s00296-016-3426-2
  • Bower JE. Cancer-related fatigue--mechanisms, risk factors, and treatments. Nature reviews. Clin Oncol. 2014;11(10):597–609.
  • Norheim KB, Jonsson G, Omdal R. Biological mechanisms of chronic fatigue. Rheumatology. 2011;50(6):1009–1018.
  • Korte SM, Straub RH. Fatigue in inflammatory rheumatic disorders: pathophysiological mechanisms. Rheumatology. 2019;58(Suppl 5):v35–v50.
  • Versteeg GA, Ten Klooster PM, van de Laar M. Fatigue is associated with disease activity in some, but not all, patients living with rheumatoid arthritis: disentangling “between-person” and “within-person” associations. BMC Rheumatol. 2022;6(1):3.
  • Druce KL, Basu N. Predictors of fatigue in rheumatoid arthritis. Rheumatology. 2019;58(Suppl 5):v29–v34.
  • Westhoff G, Dorner T, Zink A. Fatigue and depression predict physician visits and work disability in women with primary Sjogren’s syndrome: results from a cohort study. Rheumatology. 2012;51(2):262–269.
  • Pollard LC, Choy EH, Gonzalez J, Khoshaba B, Scott DL. Fatigue in rheumatoid arthritis reflects pain, not disease activity. Rheumatology. 2006;45(7):885–889.
  • Husted JA, Gladman DD, Farewell VT, Cook RJ. Health-related quality of life of patients with psoriatic arthritis: a comparison with patients with rheumatoid arthritis. Arthritis Rheum. 2001;45(2):151–158.
  • Dagfinrud H, Vollestad NK, Loge JH, Kvien TK, Mengshoel AM. Fatigue in patients with ankylosing spondylitis: a comparison with the general population and associations with clinical and self-reported measures. Arthritis Rheum. 2005;53(1):5–11. doi:10.1002/art.20910
  • Massalska M, Maslinski W, Ciechomska M. Small molecule inhibitors in the treatment of rheumatoid arthritis and beyond: latest updates and potential strategy for fighting COVID-19. Cells. 2020;9(8):1876. doi:10.3390/cells9081876
  • Pope JE. Management of Fatigue in Rheumatoid Arthritis. RMD Open. 2020;6(1):e001084. doi:10.1136/rmdopen-2019-001084
  • Howard Tripp N, Tarn J, Natasari A, et al. Fatigue in primary Sjogren’s syndrome is associated with lower levels of proinflammatory cytokines. RMD Open. 2016;2(2):e000282. doi:10.1136/rmdopen-2016-000282
  • Muskardin TLW, Niewold TB. Type I interferon in rheumatic diseases. Nat Rev Rheumatol. 2018;14(4):214–228. doi:10.1038/nrrheum.2018.31
  • Druce KL, Bhattacharya Y, Jones GT, Macfarlane GJ, Basu N. Most patients who reach disease remission following anti-TNF therapy continue to report fatigue: results from the British society for rheumatology biologics register for rheumatoid arthritis. Rheumatology. 2016;55(10):1786–1790. doi:10.1093/rheumatology/kew241
  • Almeida C, Choy EH, Hewlett S, et al. Biologic interventions for fatigue in rheumatoid arthritis. Cochrane Database Syst. Rev. 2016;2016(6):CD008334. doi:10.1002/14651858.CD008334.pub2
  • Bonek K, Roszkowski L, Massalska M, Maslinski W, Ciechomska M. Biologic drugs for rheumatoid arthritis in the context of Biosimilars, Genetics, Epigenetics and COVID-19 Treatment. Cells. 2021;10(2):323. doi:10.3390/cells10020323
  • Norheim KB, Imgenberg-Kreuz J, Alexsson A, et al. Genetic variants at the RTP4/MASP1 locus are associated with fatigue in Scandinavian patients with primary Sjögren’s syndrome. RMD Open. 2021;7(3):e001832. doi:10.1136/rmdopen-2021-001832
  • Deepak HB, Prince SE, Deshpande P. Effect of baricitinib in regulating programmed death 1 and ligand programmed cell death ligand 1 through JAK/STAT pathway in psoriasis. Indian J Pharmacol. 2022;54(3):183–193. doi:10.4103/ijp.ijp_1089_20
  • Morris G, Berk M, Galecki P, Walder K, Maes M. The neuro-immune pathophysiology of central and peripheral fatigue in systemic immune-inflammatory and neuro-immune diseases. Mol Neurobiol. 2016;53(2):1195–1219.
  • Teleanu DM, Niculescu AG, Lungu II, et al. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int J Mol Sci. 2022;23:11.
  • Avalos I, Chung CP, Oeser A, et al. Oxidative stress in systemic lupus erythematosus: relationship to disease activity and symptoms. Lupus. 2007;16(3):195–200.
  • Sharpe M, Wilks D. Fatigue. BMJ. 2002;325(7362):480–483.
  • Davies K, Dures E, Ng WF. Fatigue in inflammatory rheumatic diseases: current knowledge and areas for future research. Nat Rev Rheumatol. 2021;17(11):651–664.
  • McCusker RH, Kelley KW. Immune-neural connections: how the immune system’s response to infectious agents influences behavior. J Exp Biol. 2013;216(Pt 1):84–98.
  • Schaible HG. Nociceptive neurons detect cytokines in arthritis. Arthritis Res Ther. 2014;16(5):470.
  • Eddleston M, Mucke L. Molecular profile of reactive astrocytes--implications for their role in neurologic disease. Neuroscience. 1993;54(1):15–36.
  • Schwarcz R, Pellicciari R. Manipulation of brain kynurenines: glial targets, neuronal effects, and clinical opportunities. J Pharmacol Exp Ther. 2002;303(1):1–10.
  • Maes M, Leonard BE, Myint AM, Kubera M, Verkerk R. The new ‘5-HT’ hypothesis of depression: cell-mediated immune activation induces indoleamine 2,3-dioxygenase, which leads to lower plasma tryptophan and an increased synthesis of detrimental tryptophan catabolites (TRYCATs), both of which contribute to the onset of depression. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35(3):702–721.
  • Sas K, Robotka H, Toldi J, Vecsei L. Mitochondria, metabolic disturbances, oxidative stress and the kynurenine system, with focus on neurodegenerative disorders. J Neurol Sci. 2007;257(1–2):221–239.
  • Akesson K, Pettersson S, Stahl S, et al. Kynurenine pathway is altered in patients with SLE and associated with severe fatigue. Lupus Sci Med. 2018;5(1):e000254.
  • Schrocksnadel K, Wirleitner B, Winkler C, Fuchs D. Monitoring tryptophan metabolism in chronic immune activation. Clin Chim Acta. 2006;364(1–2):82–90.
  • Tykocinski LO, Lauffer AM, Bohnen A, et al. Synovial Fibroblasts selectively suppress Th1 Cell Responses through IDO1-mediated tryptophan catabolism. J Immunol. 2017;198(8):3109–3117.
  • Massalska M, Kuca-Warnawin E, Janicka I, et al. Survival of lymphocytes is not restricted by IDO-expressing fibroblast from rheumatoid arthritis patients. Immunopharmacol Immunotoxicol. 2019;41(2):214–223.
  • Yao N, Tretter T, Kvacskay P, et al. Targeting of janus kinases limits pro-inflammatory but also immunosuppressive circuits in the crosstalk between synovial fibroblasts and lymphocytes. Biomedicines. 2021;9:10.
  • Lamba M, Wang R, Fletcher T, Alvey C, Kushner J, Stock TC. Extended-release once-daily formulation of tofacitinib: evaluation of pharmacokinetics compared with immediate-release tofacitinib and impact of food. J Clin Pharmacol. 2016;56(11):1362–1371.
  • Shi JG, Chen X, Lee F, et al. The pharmacokinetics, pharmacodynamics, and safety of baricitinib, an oral JAK 1/2 inhibitor, in healthy volunteers. J Clin Pharmacol. 2014;54(12):1354–1361.
  • Mohamed MF, Camp HS, Jiang P, Padley RJ, Asatryan A, Othman AA. Pharmacokinetics, safety and tolerability of ABT-494, a novel selective JAK 1 inhibitor, in healthy volunteers and subjects with rheumatoid arthritis. Clin Pharmacokinet. 2016;55(12):1547–1558.
  • Larssen E, Brede C, Hjelle A, et al. Fatigue in primary Sjogren’s syndrome: a proteomic pilot study of cerebrospinal fluid. SAGE Open Medicine. 2019;7:2050312119850390.
  • Holdren M, Schieir O, Bartlett SJ, et al. Improvements in fatigue lag behind disease remission in early rheumatoid arthritis: results from the Canadian early arthritis cohort. Arthrit Rheumatol. 2021;73(1):53–60.
  • Dowell NG, Bouyagoub S, Tibble J, Voon V, Cercignani M, Harrison NA. Interferon-alpha-induced changes in NODDI predispose to the development of fatigue. Neuroscience. 2019;403:111–117.
  • Davies K, Mirza K, Tarn J, et al. Fatigue in primary Sjogren’s syndrome (pSS) is associated with lower levels of proinflammatory cytokines: a validation study. Rheumatol Int. 2019;39(11):1867–1873.
  • Azizoddin DR, Gandhi N, Weinberg S, Sengupta M, Nicassio PM, Jolly M. Fatigue in systemic lupus: the role of disease activity and its correlates. Lupus. 2019;28(2):163–173.
  • Wu Q, Inman RD, Davis KD. Tumor necrosis factor inhibitor therapy in ankylosing spondylitis: differential effects on pain and fatigue and brain correlates. Pain. 2015;156(2):297–304.
  • Reygaerts T, Mitrovic S, Fautrel B, Gossec L. Effect of biologics on fatigue in psoriatic arthritis: a systematic literature review with meta-analysis. Joint Bone Spine. 2018;85(4):405–410.
  • Atzeni F, Nucera V, Masala IF, Sarzi-Puttini P, Bonitta G. Il-6 Involvement in pain, fatigue and mood disorders in rheumatoid arthritis and the effects of Il-6 inhibitor sarilumab. Pharmacol Res. 2019;149:104402.
  • Wu P, Wu X, Zhou G, et al. P2X7 receptor-induced bone cancer pain by regulating microglial activity via NLRP3/IL-1beta Signaling. Pain Physician. 2022;25(8):E1199–E1210.
  • Alciati A, Di Carlo M, Siragusano C, Palumbo A, Masala IF, Atzeni F. Effect of biological DMARDs and JAK inhibitors in pain of chronic inflammatory arthritis. Expert Opin Biol Ther. 2022;22(10):1311–1322.
  • Moulton VR. Sex Hormones in Acquired Immunity and Autoimmune Disease. Front Immunol. 2018;9:2279.
  • Sokka T, Toloza S, Cutolo M, et al. Women, men, and rheumatoid arthritis: analyses of disease activity, disease characteristics, and treatments in the QUEST-RA study. Arthritis Res Ther. 2009;11(1):R7.
  • Fleischmann R, Schiff M, van der Heijde D, et al. Baricitinib, methotrexate, or combination in patients with rheumatoid arthritis and no or limited prior disease-modifying antirheumatic drug treatment. Arthrit Rheumatol. 2017;69(3):506–517.
  • Felson DT, Anderson JJ, Boers M, et al. The American College of Rheumatology preliminary core set of disease activity measures for rheumatoid arthritis clinical trials. The committee on outcome measures in rheumatoid arthritis clinical trials. Arthritis Rheum. 1993;36(6):729–740.
  • Singh JA, Saag KG, Bridges SL Jr, et al. 2015 American college of rheumatology guideline for the treatment of rheumatoid arthritis. Arthrit Care Res. 2016;68(1):1–25.
  • Smolen JS, Landewe R, Bijlsma J, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017;76(6):960–977.
  • Food and Drug Administration. Guidance for industry rheumatoid arthritis: developing drug products for treatment https://www.fda.gov/regulatory-information/search-fda-guidance-documents/rheumatoid-arthritis-developing-drug-products-treatment. Accessed August 11, 2023.
  • Whalley D, McKenna SP, de Jong Z, van der Heijde D. Quality of life in rheumatoid arthritis. Br J Rheumatol. 1997;36(8):884–888.
  • Cole JC, Motivala SJ, Khanna D, Lee JY, Paulus HE, Irwin MR. Validation of single-factor structure and scoring protocol for the Health Assessment Questionnaire-Disability Index. Arthritis Rheum. 2005;53(4):536–542.
  • Saag KG, Teng GG, Patkar NM, et al. American College of Rheumatology 2008 recommendations for the use of nonbiologic and biologic disease-modifying antirheumatic drugs in rheumatoid arthritis. Arthritis Rheum. 2008;59(6):762–784.
  • Luqmani R, Hennell S, Estrach C, et al. British society for rheumatology and British health professionals in rheumatology guideline for the management of rheumatoid arthritis (after the first 2 years). Rheumatology. 2009;48(4):436–439.
  • Jansen JP, Buckley F, Dejonckheere F, Ogale S. Comparative efficacy of biologics as monotherapy and in combination with methotrexate on patient reported outcomes (PROs) in rheumatoid arthritis patients with an inadequate response to conventional DMARDs--a systematic review and network meta-analysis. Health Qual Life Outcomes. 2014;12:102.
  • Ramey DR, Raynauld JP, Fries JF. The health assessment questionnaire 1992: status and review. Arthritis Care Res. 1992;5(3):119–129.
  • Strand V, Singh JA. Improved health-related quality of life with effective disease-modifying antirheumatic drugs: evidence from randomized controlled trials. Am J Manag Care. 2008;14(4):234–254.
  • Mathias SD, Colwell HH, Miller DP, Moreland LW, Buatti M, Wanke L. Health-related quality of life and functional status of patients with rheumatoid arthritis randomly assigned to receive etanercept or placebo. Clin Ther. 2000;22(1):128–139.
  • van de Putte LB, Atkins C, Malaise M, et al. Efficacy and safety of Adalimumab as monotherapy in patients with rheumatoid arthritis for whom previous disease modifying antirheumatic drug treatment has failed. Ann Rheum Dis. 2004;63(5):508–516.
  • Holten K, Paulshus Sundlisater N, Lillegraven S, et al. Fatigue in patients with early rheumatoid arthritis undergoing treat-to-target therapy: predictors and response to treatment. Ann Rheum Dis. 2022;81(3):344–350.
  • Calin A, Edmunds L, Kennedy LG. Fatigue in ankylosing spondylitis--why is it ignored? J Rheumatol. 1993;20(6):991–995.
  • Hammoudeh M, Zack DJ, Li W, Stewart VM, Koenig AS. Associations between inflammation, nocturnal back pain and fatigue in ankylosing spondylitis and improvements with etanercept therapy. J Int Med Res. 2013;41(4):1150–1159.
  • Kotsis K, Voulgari PV, Drosos AA, Carvalho AF, Hyphantis T. Health-related quality of life in patients with ankylosing spondylitis: a comprehensive review. Expert Rev Pharmacoecon Outcomes Res. 2014;14(6):857–872.
  • Garrett S, Jenkinson T, Kennedy LG, Whitelock H, Gaisford P, Calin A. A new approach to defining disease status in ankylosing spondylitis: the bath ankylosing spondylitis disease activity index. J Rheumatol. 1994;21(12):2286–2291.
  • Calin A, Garrett S, Whitelock H, et al. A new approach to defining functional ability in ankylosing spondylitis: the development of the bath ankylosing spondylitis functional index. J Rheumatol. 1994;21(12):2281–2285.
  • Boonen A, van der Heijde D, Landewe R, et al. How do the EQ-5D, SF-6D and the well-being rating scale compare in patients with ankylosing spondylitis? Ann Rheum Dis. 2007;66(6):771–777.
  • Krupp LB, LaRocca NG, Muir-Nash J, Steinberg AD. The fatigue severity scale. Application to patients with multiple sclerosis and systemic lupus erythematosus. Arch Neurol. 1989;46(10):1121–1123.
  • Doward LC, Spoorenberg A, Cook SA, et al. Development of the ASQoL: a quality of life instrument specific to ankylosing spondylitis. Ann Rheum Dis. 2003;62(1):20–26.
  • Salaffi F, Carotti M, Gasparini S, Intorcia M, Grassi W. The health-related quality of life in rheumatoid arthritis, ankylosing spondylitis, and psoriatic arthritis: a comparison with a selected sample of healthy people. Health Qual Life Outcomes. 2009;7:25.
  • Singh H, Arya S, Talapatra P, et al. Assessment of fatigue in rheumatoid arthritis (by functional assessment of chronic illness therapy-fatigue score) and its relation to disease activity and anemia. J Clin Rheumatol. 2014;20(2):87–90.
  • Carneiro C, Chaves M, Verardino G, et al. Evaluation of fatigue and its correlation with quality of life index, anxiety symptoms, depression and activity of disease in patients with psoriatic arthritis. Clin Cosmet Investig Dermatol. 2017;10:155–163.
  • Gudu T, Gossec L. Quality of life in psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(5):405–417.
  • Gudu T, Etcheto A, de Wit M, et al. Fatigue in psoriatic arthritis - a cross-sectional study of 246 patients from 13 countries. Joint Bone Spine. 2016;83(4):439–443.
  • Orbai AM, de Wit M, Mease P, et al. International patient and physician consensus on a psoriatic arthritis core outcome set for clinical trials. Ann Rheum Dis. 2017;76(4):673–680.
  • Gossec L, de Wit M, Kiltz U, et al. A patient-derived and patient-reported outcome measure for assessing psoriatic arthritis: elaboration and preliminary validation of the Psoriatic Arthritis Impact of Disease (PsAID) questionnaire, a 13-country EULAR initiative. Ann Rheum Dis. 2014;73(6):1012–1019.
  • European Union. Olumiant. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/olumiant. Accessed August 11, 2023.
  • European Union. Xeljanz. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/xeljanz. Accessed August 11, 2023.
  • European Union. Rinvoq. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/rinvoq. Accessed August 11, 2023.
  • Smolen JS, Breedveld FC, Burmester GR, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75(1):3–15.
  • Lee EB, Fleischmann R, Hall S, et al. Tofacitinib versus methotrexate in rheumatoid arthritis. N Engl J Med. 2014;370(25):2377–2386.
  • Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010;69(1):88–96.
  • Emery P, Breedveld FC, Hall S, et al. Comparison of methotrexate monotherapy with a combination of methotrexate and etanercept in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised, double-blind, parallel treatment trial. Lancet. 2008;372(9636):375–382.
  • Nash P, Nicholls D. Perceptions of methotrexate use in rheumatoid arthritis by rheumatologists and their patients: an Australian survey study. Int J Rheum Dis. 2013;16(6):652–661.
  • Palmroth M, Kuuliala K, Peltomaa R, et al. Tofacitinib suppresses several JAK-STAT pathways in rheumatoid arthritis in vivo and baseline signaling profile associates with treatment response. Front Immunol. 2021;12:738481.
  • Choy EHS, Calabrese LH. Neuroendocrine and neurophysiological effects of interleukin 6 in rheumatoid arthritis. Rheumatology. 2018;57(11):1885–1895.
  • Li Y, Yuan L, Yang J, et al. Changes in serum cytokines may predict therapeutic efficacy of tofacitinib in rheumatoid arthritis. Mediators Inflamm. 2019;2019:5617431.
  • Wallenstein GV, Kanik KS, Wilkinson B, et al. Effects of the oral Janus kinase inhibitor tofacitinib on patient-reported outcomes in patients with active rheumatoid arthritis: results of two Phase 2 randomised controlled trials. Clin Exp Rheumatol. 2016;34(3):430–442.
  • Strand V, Kremer J, Wallenstein G, et al. Effects of tofacitinib monotherapy on patient-reported outcomes in a randomized Phase 3 study of patients with active rheumatoid arthritis and inadequate responses to DMARDs. Arthritis Res Ther. 2015;17:307.
  • Strand V, van Vollenhoven RF, Lee EB, et al. Tofacitinib or Adalimumab versus placebo: patient-reported outcomes from a phase 3 study of active rheumatoid arthritis. Rheumatology. 2016;55(6):1031–1041.
  • Strand V, Lee EB, Fleischmann R, et al. Tofacitinib versus methotrexate in rheumatoid arthritis: patient-reported outcomes from the randomised phase III ORAL Start trial. RMD Open. 2016;2(2):e000308.
  • Strand V, Kremer JM, Gruben D, Krishnaswami S, Zwillich SH, Wallenstein GV. Tofacitinib in combination with conventional disease-modifying antirheumatic drugs in patients with active rheumatoid arthritis: patient-reported outcomes from a phase iii randomized controlled trial. Arthrit Care Res. 2017;69(4):592–598.
  • Strand V, Burmester GR, Zerbini CA, et al. Tofacitinib with methotrexate in third-line treatment of patients with active rheumatoid arthritis: patient-reported outcomes from a phase III trial. Arthrit Care Res. 2015;67(4):475–483.
  • Strand V, Mysler E, Moots RJ, et al. Patient-reported outcomes for tofacitinib with and without methotrexate, or Adalimumab with methotrexate, in rheumatoid arthritis: a phase IIIB/IV trial. RMD Open. 2019;5(2):e001040.
  • Mease P, Hall S, FitzGerald O, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377(16):1537–1550.
  • Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377(16):1525–1536. doi:10.1056/NEJMoa1615977
  • Coates LC, Bushmakin AG, FitzGerald O, et al. Relationships between psoriatic arthritis composite measures of disease activity with patient-reported outcomes in phase 3 studies of tofacitinib. Arthritis Res Ther. 2021;23(1):94. doi:10.1186/s13075-021-02474-2
  • Overman CL, Kool MB, Da Silva JA, Geenen R. The prevalence of severe fatigue in rheumatic diseases: an international study. Clin Rheumatol. 2016;35(2):409–415. doi:10.1007/s10067-015-3035-6
  • Strand V, de Vlam K, Covarrubias-Cobos JA, et al. Tofacitinib or Adalimumab versus placebo: patient-reported outcomes from OPAL Broaden-a phase III study of active psoriatic arthritis in patients with an inadequate response to conventional synthetic disease-modifying antirheumatic drugs. RMD Open. 2019;5(1):e000806. doi:10.1136/rmdopen-2018-000806
  • Strand V, de Vlam K, Covarrubias-Cobos JA, et al. Effect of tofacitinib on patient-reported outcomes in patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors in the Phase III, randomised controlled trial: OPAL Beyond. RMD Open. 2019;5(1):e000808. doi:10.1136/rmdopen-2018-000808
  • van der Heijde D, Deodhar A, Wei JC, et al. Tofacitinib in patients with ankylosing spondylitis: a Phase II, 16-week, randomised, placebo-controlled, dose-ranging study. Ann Rheum Dis. 2017;76(8):1340–1347. doi:10.1136/annrheumdis-2016-210322
  • Navarro-Compan V, Wei JC, Van den Bosch F, et al. Effect of tofacitinib on pain, fatigue, health-related quality of life and work productivity in patients with active ankylosing spondylitis: results from a phase III, randomised, double-blind, placebo-controlled trial. RMD Open. 2022;8(2):e002253. doi:10.1136/rmdopen-2022-002253
  • Ruperto N, Brunner HI, Synoverska O, et al. Tofacitinib in juvenile idiopathic arthritis: a double-blind, placebo-controlled, withdrawal phase 3 randomised trial. Lancet. 2021;398(10315):1984–1996. doi:10.1016/S0140-6736(21)01255-1
  • McInnes IB, Byers NL, Higgs RE, et al. Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations. Arthritis Res Ther. 2019;21(1):183. doi:10.1186/s13075-019-1964-1
  • Weston S, Macdonald JL, Williams LM, et al. The JAK inhibitor baricitinib inhibits oncostatin M induction of proinflammatory mediators in ex-vivo synovial derived cells. Clin Exp Rheumatol. 2021;40:1620–1628. doi:10.55563/clinexprheumatol/cfsajk
  • Choy EHS, Miceli-Richard C, Gonzalez-Gay MA, et al. The effect of JAK1/JAK2 inhibition in rheumatoid arthritis: efficacy and safety of baricitinib. Clin Exp Rheumatol. 2019;37(4):694–704.
  • Schiff M, Takeuchi T, Fleischmann R, et al. Patient-reported outcomes of baricitinib in patients with rheumatoid arthritis and no or limited prior disease-modifying antirheumatic drug treatment. Arthritis Res Ther. 2017;19(1):208. doi:10.1186/s13075-017-1410-1
  • Taylor PC, Alten R, Alvaro Gracia JM, et al. Achieving pain control in early rheumatoid arthritis with baricitinib monotherapy or in combination with methotrexate versus methotrexate monotherapy. RMD Open. 2022;8(1):e001994. doi:10.1136/rmdopen-2021-001994
  • Michaud K, Pope JE, Emery P, et al. Relative Impact of pain and fatigue on work productivity in patients with rheumatoid arthritis from the RA-BEAM baricitinib trial. Rheumatol Therap. 2019;6(3):409–419. doi:10.1007/s40744-019-0164-4
  • Keystone EC, Taylor PC, Tanaka Y, et al. Patient-reported outcomes from a phase 3 study of baricitinib versus placebo or Adalimumab in rheumatoid arthritis: secondary analyses from the RA-BEAM study. Ann Rheum Dis. 2017;76(11):1853–1861. doi:10.1136/annrheumdis-2017-211259
  • Genovese MC, Kremer J, Zamani O, et al. Baricitinib in patients with refractory rheumatoid arthritis. N Engl J Med. 2016;374(13):1243–1252. doi:10.1056/NEJMoa1507247
  • Smolen JS, Kremer JM, Gaich CL, et al. Patient-reported outcomes from a randomised phase III study of baricitinib in patients with rheumatoid arthritis and an inadequate response to biological agents (RA-BEACON). Ann Rheum Dis. 2017;76(4):694–700. doi:10.1136/annrheumdis-2016-209821
  • Fautrel B, Wu J, Wang D, Haladyj E, van de Laar M, Takeuchi T. Relative impact of pain and disease activity on improvements in fatigue: results from 2 baricitinib phase 3 clinical trials. J Clin Rheumatol. 2022;2022:1.
  • Parmentier JM, Voss J, Graff C, et al. In vitro and in vivo characterization of the JAK1 selectivity of upadacitinib (ABT-494). BMC Rheumatol. 2018;2:23. doi:10.1186/s41927-018-0031-x
  • Strand V, Pope J, Tundia N, et al. Upadacitinib improves patient-reported outcomes in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying antirheumatic drugs: results from SELECT-NEXT. Arthritis Res Ther. 2019;21(1):272. doi:10.1186/s13075-019-2037-1
  • Strand V, Tundia N, Bergman M, et al. Upadacitinib improves patient-reported outcomes vs placebo or Adalimumab in patients with rheumatoid arthritis: results from SELECT-COMPARE. Rheumatology. 2021;60(12):5583–5594. doi:10.1093/rheumatology/keab158
  • McInnes IB, Ostor AJK, Mease PJ, et al. Effect of upadacitinib on reducing pain in patients with active psoriatic arthritis or ankylosing spondylitis: post hoc analysis of three randomised clinical trials. RMD Open. 2022;8(1):e002049. doi:10.1136/rmdopen-2021-002049
  • Deodhar A, van der Heijde D, Sieper J, et al. Safety and efficacy of upadacitinib in patients with active ankylosing spondylitis and an inadequate response to nonsteroidal antiinflammatory drug therapy: one-year results of a double-blind, placebo-controlled study and open-label extension. Arthrit Rheumatol. 2022;74(1):70–80. doi:10.1002/art.41911
  • van der Heijde D, Deodhar A, Maksymowych WP, et al. Upadacitinib in active ankylosing spondylitis: results of the 2-year, double-blind, placebo-controlled SELECT-AXIS 1 study and open-label extension. RMD Open. 2022;8(2):e002280. doi:10.1136/rmdopen-2022-002280
  • European Union. Jyseleca. Available from: https://www.ema.europa.eu/en/medicines/human/EPAR/jyseleca. Accessed August 11, 2023.
  • Namour F, Anderson K, Nelson C, Tasset C. Filgotinib: a clinical pharmacology review. Clin Pharmacokinet. 2022;61:819–832. doi:10.1007/s40262-022-01129-y
  • Traves PG, Murray B, Campigotto F, Galien R, Meng A, Di Paolo JA. JAK selectivity and the implications for clinical inhibition of pharmacodynamic cytokine signalling by filgotinib, upadacitinib, tofacitinib and baricitinib. Ann Rheum Dis. 2021;80(7):865–875. doi:10.1136/annrheumdis-2020-219012
  • Dowty ME, Lin TH, Jesson MI, et al. Janus kinase inhibitors for the treatment of rheumatoid arthritis demonstrate similar profiles of in vitro cytokine receptor inhibition. Pharmacol Res Perspect. 2019;7(6):e00537. doi:10.1002/prp2.537
  • Bingham CO 3rd, Walker D, Nash P, et al. The impact of filgotinib on patient-reported outcomes and health-related quality of life for patients with active rheumatoid arthritis: a post hoc analysis of Phase 3 studies. Arthritis Res Ther. 2022;24(1):11. doi:10.1186/s13075-021-02677-7
  • Huarte E, Peel MT, Verbist K, et al. Ruxolitinib, a JAK1/2 Inhibitor, ameliorates cytokine storm in experimental models of hyperinflammation syndrome. Front Pharmacol. 2021;12:650295. doi:10.3389/fphar.2021.650295
  • Bader-Meunier B, Hadchouel A, Berteloot L, et al. Effectiveness and safety of ruxolitinib for the treatment of refractory systemic idiopathic juvenile arthritis like associated with interstitial lung disease: a case report. Ann Rheum Dis. 2022;81(2):e20. doi:10.1136/annrheumdis-2020-216983
  • Jain P, Keating M, Renner S, et al. Ruxolitinib for symptom control in patients with chronic lymphocytic leukaemia: a single-group, phase 2 trial. Lancet Haematol. 2017;4(2):e67–e74. doi:10.1016/S2352-3026(16)30194-6
  • Hardwick RN, Brassil P, Badagnani I, et al. Gut-selective design of orally administered izencitinib (TD-1473) limits systemic exposure and effects of janus kinase inhibition in nonclinical species. Toxicol Sci. 2022;186(2):323–337. doi:10.1093/toxsci/kfac002
  • Toth L, Juhasz MF, Szabo L, et al. Janus kinase inhibitors improve disease activity and patient-reported outcomes in rheumatoid arthritis: a systematic review and meta-analysis of 24,135 patients. Int J Mol Sci. 2022;23(3):1246. doi:10.3390/ijms23031246
  • Odriozola ICC, Barnetche T, Richez C, Bannwarth B, Schaeverbeke T Is there a specific effect of jak-inhibitors on pain and fatigue in rheumatoid arthritis? Available from: https://acrabstracts.org/abstract/is-there-a-specific-effect-of-jak-inhibitors-on-pain-and-fatigue-in-rheumatoid-arthritis/. Accessed August 11, 2023.
  • Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med. 2017;376(7):652–662. doi:10.1056/NEJMoa1608345
  • Fleischmann R, Pangan AL, Song IH, et al. Upadacitinib versus placebo or adalimumab in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase III, double-blind, randomized controlled trial. Arthrit Rheumatol. 2019;71(11):1788–1800. doi:10.1002/art.41032
  • Fleischmann R, Mysler E, Hall S, et al. Efficacy and safety of tofacitinib monotherapy, tofacitinib with methotrexate, and Adalimumab with methotrexate in patients with rheumatoid arthritis (ORAL Strategy): a phase 3b/4, double-blind, head-to-head, randomised controlled trial. Lancet. 2017;390(10093):457–468. doi:10.1016/S0140-6736(17)31618-5
  • Combe B, Kivitz A, Tanaka Y, et al. Filgotinib versus placebo or Adalimumab in patients with rheumatoid arthritis and inadequate response to methotrexate: a phase III randomised clinical trial. Ann Rheum Dis. 2021;80(7):848–858. doi:10.1136/annrheumdis-2020-219214
  • EMA confirms measures to minimise risk of serious side effects with Janus kinase inhibitors for chronic inflammatory disorders. Available from: https://www.ema.europa.eu/en/news/ema-confirms-measures-minimise-risk-serious-side-effects-janus-kinase-inhibitors-chronic. Accessed August 11, 2023.