The Interleukin-1 Receptor-Associated Kinase 4 Inhibitor PF-06650833 Blocks Inflammation in Preclinical Models of Rheumatic Disease and in Humans Enrolled in a Randomized Clinical Trial

Aaron Winkler, Weiyong Sun, Saurav De, Aiping Jiao, M Nusrat Sharif, Peter T Symanowicz, Shruti Athale, Julia H Shin, Ju Wang, Bruce A Jacobson, Simeon J Ramsey, Ken Dower, Tatyana Andreyeva, Heng Liu, Martin Hegen, Bruce L Homer, Joanne Brodfuehrer, Mera Tilley, Steven A Gilbert, Spencer I Danto, Jean J Beebe, Betsy J Barnes, Virginia Pascual, Lih-Ling Lin, Iain Kilty, Margaret Fleming, Vikram R Rao, Aaron Winkler, Weiyong Sun, Saurav De, Aiping Jiao, M Nusrat Sharif, Peter T Symanowicz, Shruti Athale, Julia H Shin, Ju Wang, Bruce A Jacobson, Simeon J Ramsey, Ken Dower, Tatyana Andreyeva, Heng Liu, Martin Hegen, Bruce L Homer, Joanne Brodfuehrer, Mera Tilley, Steven A Gilbert, Spencer I Danto, Jean J Beebe, Betsy J Barnes, Virginia Pascual, Lih-Ling Lin, Iain Kilty, Margaret Fleming, Vikram R Rao

Abstract

Objective: To investigate the role of PF-06650833, a highly potent and selective small-molecule inhibitor of interleukin-1-associated kinase 4 (IRAK4), in autoimmune pathophysiology in vitro, in vivo, and in the clinical setting.

Methods: Rheumatoid arthritis (RA) inflammatory pathophysiology was modeled in vitro through 1) stimulation of primary human macrophages with anti-citrullinated protein antibody immune complexes (ICs), 2) RA fibroblast-like synoviocyte (FLS) cultures stimulated with Toll-like receptor (TLR) ligands, as well as 3) additional human primary cell cocultures exposed to inflammatory stimuli. Systemic lupus erythematosus (SLE) pathophysiology was simulated in human neutrophils, dendritic cells, B cells, and peripheral blood mononuclear cells stimulated with TLR ligands and SLE patient ICs. PF-06650833 was evaluated in vivo in the rat collagen-induced arthritis (CIA) model and the mouse pristane-induced and MRL/lpr models of lupus. Finally, RNA sequencing data generated with whole blood samples from a phase I multiple-ascending-dose clinical trial of PF-06650833 were used to test in vivo human pharmacology.

Results: In vitro, PF-06650833 inhibited human primary cell inflammatory responses to physiologically relevant stimuli generated with RA and SLE patient plasma. In vivo, PF-06650833 reduced circulating autoantibody levels in the pristane-induced and MRL/lpr murine models of lupus and protected against CIA in rats. In a phase I clinical trial (NCT02485769), PF-06650833 demonstrated in vivo pharmacologic action pertinent to SLE by reducing whole blood interferon gene signature expression in healthy volunteers.

Conclusion: These data demonstrate that inhibition of IRAK4 kinase activity can reduce levels of inflammation markers in humans and provide confidence in the rationale for clinical development of IRAK4 inhibitors for rheumatologic indications.

© 2021 The Authors. Arthritis & Rheumatology published by Wiley Periodicals LLC on behalf of American College of Rheumatology.

Figures

Figure 1
Figure 1
Pharmacologic properties of PF‐06650833, an interleukin‐1–associated kinase 4 (IRAK4) inhibitor. A, Structure of PF‐06650833. B, Selectivity of PF‐06650833 in an ActivX ATP occupancy assay using THP‐1 lysates. The top 26 of >200+ kinases are shown. Fifty percent inhibition concentrations (IC50) were determined using a 5‐point dose‐response curve (full data set shown in Supplementary Table 1, on the Arthritis & Rheumatology website at http://onlinelibrary.wiley.com/doi/10.1002/art.41953/abstract). C, Potency of PF‐06650833 in enzyme and cell‐based assays. IC50 values for enzyme and peripheral blood mononuclear cell (PBMC) assays are a single value, whereas whole blood (wb) assays for human, mouse, and rat are denoted as total concentration of compound over the non–protein‐bound (free [f]) concentration of compound, calculated as (wb IC50 × [fu/(B/P)] = IC50 free, where fraction unbound [fu] = 0.22 and blood/plasma ratio [B/P] = 0.91). D, Demonstration of the relationship between free drug concentration, IRAK4 ATP binding site occupancy, and inhibition of downstream biology by PF‐06650833 in a whole blood sample from a patient with systemic lupus erythematosus. Values are the mean ± SEM. E, Use of 50% maximum response concentration data from each preclinical assay to determine that 100 nM was the target free compound concentration required to maintain >90% IRAK4 inhibition across species and assays. CK1g2 = casein kinase 1 gamma 2; PIP4K2C = phosphatidylinositol‐5‐phosphate 4‐kinase 2C; NDR1 = nuclear Dbf2‐related kinase 1; PRP4 = pre–mRNA processing factor 4; MPSK1 = myristoylated and palmitoylated serine/threonine kinase 1; LKB1 = liver kinase B1; LATS2 = large tumor suppressor kinase 2; LRRK2 = leucine‐rich repeat kinase 2; TLK2 = tousled‐like kinase 2; MASTL = microtubule‐associated serine/threonine kinase–like; MLKL = mixed‐lineage kinase domain–like; CHK2 = checkpoint kinase 2; AurA = aurora A kinase; AXL = AXL receptor tyrosine kinase; CAMK2g = Ca2+/calmodulin‐dependent protein kinase 2g; GAK = cyclin G–associated kinase; TNF = tumor necrosis factor; IL‐6 = interleukin‐6; HWB = human whole blood; LPS = lipopolysaccharide; IRF‐5 = interferon regulatory factor 5.
Figure 2
Figure 2
PF‐06650833 inhibits rheumatoid arthritis (RA) pathophysiology. A, Human macrophages were exposed to anti–citrullinated protein antibody (ACPA) immune complexes (ICs) formed with cyclic citrullinated peptide–positive RA sera in the presence or absence of 100 nM interleukin‐1–associated kinase 4 inhibitor (IRAK4i) (PF‐06650833) or 100 nM Bruton’s tyrosine kinase inhibitor (BTKi) (PF‐303). Supernatants were analyzed by enzyme‐linked immunosorbent assay for induction of tumor necrosis factor (TNF). Values are the percent in relation to DMSO treatment (set at 100). Symbols represent individual subjects (n = 11 per group); bars show the mean. B, Human RA fibroblast‐like synoviocytes were stimulated with 10 μg/ml Pam3Cys (Toll‐like receptor 1/2 [TLR‐1/2]), 10 ng/ml lipopolysaccharide (LPS) (TLR‐4), 100 ng/ml flagellin (TLR‐5), or 0.1 ng/ml interleukin‐1β (IL‐1β) in the presence or absence of 100 nM PF‐06650833. Cytokine and matrix metalloproteinase (MMP) content in supernatants was assessed by Meso Scale Discovery assay. Values are the percent of those observed with vehicle control. C, Compounds at the noted concentrations were profiled with DiscoverRx on the BioMAP platform. Data represent the log ratio of values in compound‐treated samples to controls, with negative values indicating inhibition and points outside of the gray shading demonstrating a statistically significant difference based on assay variability. Each point on the x‐axis represents the result of a different end point in each assay system: venular endothelial cells (4H), peripheral blood mononuclear cells (PBMCs) plus venular endothelial cells stimulated with LPS (LPS), PBMCs plus venular endothelial cells stimulated with superantigen (SAg), B cells plus PBMCs (BT), lung fibroblasts (MyoF), and macrophages plus venular endothelial cells (/Mphg). Unstim = unstimulated; MCP‐1 = monocyte chemotactic factor 1; VCAM‐1 = vascular cell adhesin molecule 1; uPAR = urokinase‐type plasminogen activator receptor; SRB = sulforhodamine B staining; VEGFR‐2 = vascular endothelial growth factor receptor 2; M‐CSF = macrophage colony‐stimulating factor; sPGE2 = soluble prostaglandin E2; MIG = monokine induced by interferon‐γ; bFGF = basic fibroblast growth factor; PAI‐1 = plasminogen activator inhibitor 1; TIMP‐1 = tissue inhibitor of metalloproteinases 1.
Figure 3
Figure 3
PF‐06650833 is efficacious in rat collagen‐induced arthritis (CIA). Rats with CIA were treated for 7 days with PF‐06650833 3 mg/kg twice daily (bid), tofacitinib 10 mg/kg daily (qd), or vehicle, and paw volume was measured daily. Data are from a representative experiment (of 3 experiments performed). Values are the mean ± SEM.
Figure 4
Figure 4
PF‐06650833 inhibits pathophysiology in human systemic lupus erythematosus (SLE). A, DNA release was measured in neutrophil supernatant (sup) following exposure to R837 in the presence or absence of 100 nM PF‐06650833. Values are the mean ± SEM (n = 3 donors). B, Peripheral blood mononuclear cells (PBMCs) were exposed to either R848 or SLE sera in the presence or absence of 100 nM PF‐06650833, incubated for 2 hours at 37°C, and then interferon regulatory factor 5 (IRF‐5) nuclear translocation was analyzed by Amnis imaging cytometry. C, B cells were exposed to interferon‐α (IFNα) plus R848 in the presence or absence of 100 nM PF‐06650833. Supernatants were harvested at 24 hours to assess interleukin‐6 (IL‐6) release and at 72 hours to assess IL‐10 release. Plasmablasts were quantified by flow cytometry after 6 days. D, Neutrophils were isolated from SLE patients and NETosis induced with R837. Supernatants were used to stimulate plasmacytoid dendritic cells for 24 hours in the presence or absence of 200 nM PF‐06650833 prior to quantification of CD83 induction by flow cytometry (n = 5) or IFNα release by enzyme‐linked immunosorbent assay (ELISA) (mean ± SEM; n = 3). Iso = isotype. E, PBMCs were exposed to SLE immune complexes (ICs) in the presence or absence of 100 nM PF‐06650833. After 24 hours, supernatant was analyzed by ELISA for IFNα, and IFN‐induced gene expression in cells was measured by quantitative reverse transcription–polymerase chain reaction. RQ = relative quantification.
Figure 5
Figure 5
Effect of PF‐06650833 on pristane‐induced systemic lupus erythematosus. PF‐06650833 was administered to BALB/c mice by chow dosing during weeks 8–20 following pristane administration. Anti–double‐stranded DNA (anti‐dsDNA) was quantified by enzyme‐linked immunosorbent assay at weeks 4, 8, 12, and 20, and the mean ± SEM value at each of these time points is shown. Additional autoantibody titers (anti‐SSA, anti‐RNP) were determined, and immunohistochemical assessments (periodic acid–Schiff [PAS] staining) were performed, at week 20. Data are from a representative experiment (of 2 experiments performed). Symbols represent individual subjects; bars show the mean. PBS = phosphate buffered saline; mpk = mg/kg.
Figure 6
Figure 6
PF‐06650833 inhibits type I interferon signature in vivo in humans. Moderate‐release (MR) PF‐06650833 (300 mg/day [qd]) (n = 7) or placebo (n = 11) was administered for 14 days in a phase I multiple‐ascending‐dose trial in healthy human volunteer subjects. Whole blood was collected in a PAXgene tube on day 0 prior to administration of the first dose and on day 14 prior to administration of the last dose, RNA extracted, and a composite gene signature calculated. The percent change in the composite gene signature for each individual participant between the 2 time points is shown. Each box represents the 25th to 75th percentiles. Lines inside the boxes represent the median. Lines outside the boxes represent the full range of values. Symbols represent individual patients.

References

    1. Malmström V, Catrina AI, Klareskog L. The immunopathogenesis of seropositive rheumatoid arthritis: from triggering to targeting [review]. Nat Rev Immunol 2016;17:60–75.
    1. Smolen JS, Aletaha D, Barton A, Burmester GR, Emery P, Firestein GS, et al. Rheumatoid arthritis [review]. Nat Rev Dis Primers 2018;4:18001.
    1. Aletaha D, Smolen JS. Diagnosis and management of rheumatoid arthritis: a review. JAMA 2018;320:1360–72.
    1. Smolen JS, Aletaha D. Rheumatoid arthritis therapy reappraisal: strategies, opportunities and challenges [review]. Nat Rev Rheumatol 2015;11:276–89.
    1. Dörner T, Furie R. Novel paradigms in systemic lupus erythematosus [review]. Lancet 2019;393:2344–58.
    1. Tsokos GC, Lo MS, Reis PC, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus [review]. Nat Rev Rheumatol 2016;12:716–30.
    1. Cushing L, Winkler A, Jelinsky SA, Lee K, Korver W, Hawtin R, et al. IRAK4 kinase activity controls Toll‐like receptor–induced inflammation through the transcription factor IRF5 in primary human monocytes. J Biol Chem 2017;292:18689–98.
    1. De S, Karim F, Kiessu E, Cushing L, Lin LL, Ghandil P, et al. Mechanism of dysfunction of human variants of the IRAK4 kinase and a role for its kinase activity in interleukin‐1 receptor signaling. J Biol Chem 2018;293:15208–20.
    1. Balka KR, De Nardo D. Understanding early TLR signaling through the Myddosome [review]. J Leukoc Biol 2019;105:339–51.
    1. Suzuki N, Suzuki S, Duncan GS, Millar DG, Wada T, Mirtsos C, et al. Severe impairment of interleukin‐1 and Toll‐like receptor signalling in mice lacking IRAK‐4. Nature 2002;416:750–6.
    1. Kim TW, Staschke K, Bulek K, Yao J, Peters K, Oh KH, et al. A critical role for IRAK4 kinase activity in Toll‐like receptor‐mediated innate immunity. J Exp Med 2007;204:1025–36.
    1. Koziczak‐Holbro M, Littlewood‐Evans A, Pöllinger B, Kovarik J, Dawson J, Zenke G, et al. The critical role of kinase activity of interleukin‐1 receptor–associated kinase 4 in animal models of joint inflammation. Arthritis Rheum 2009;60:1661–71.
    1. Nanda SK, Lopez‐Pelaez M, Arthur JS, Marchesi F, Cohen P. Suppression of IRAK1 or IRAK4 catalytic activity, but not type 1 IFN signaling, prevents lupus nephritis in mice expressing a ubiquitin binding‐defective mutant of ABIN1. J Immunol 2016;197:4266–73.
    1. Kawagoe T, Sato S, Jung A, Yamamoto M, Matsui K, Kato H, et al. Essential role of IRAK‐4 protein and its kinase activity in Toll‐like receptor‐mediated immune responses but not in TCR signaling. J Exp Medicine 2007;204:1013–24.
    1. Picard C, Puel A, Bonnet M, Ku CL, Bustamante J, Yang K, et al. Pyogenic bacterial infections in humans with IRAK‐4 deficiency. Science 2003;299:2076–9.
    1. Kelly PN, Romero DL, Yang Y, Shaffer AL III, Chaudhary D, Robinson S, et al. Selective interleukin‐1 receptor‐associated kinase 4 inhibitors for the treatment of autoimmune disorders and lymphoid malignancy. J Exp Med 2015;212:2189–201.
    1. Chaudhary D, Robinson S, Romero DL. Recent advances in the discovery of small molecule inhibitors of interleukin‐1 receptor‐associated kinase 4 (IRAK4) as a therapeutic target for inflammation and oncology disorders [review]. J Med Chem 2015;58:96–110.
    1. Lee KL, Ambler CM, Anderson DR, Boscoe BP, Bree AG, Brodfuehrer JI, et al. Discovery of clinical candidate 1‐{[(2S,3S,4S)‐3‐Ethyl‐4‐fluoro‐5‐oxopyrrolidin‐2‐yl]methoxy}‐7‐methoxyisoquinoline‐6‐carboxamide (PF‐06650833), a potent, selective inhibitor of interleukin‐1 receptor associated kinase 4 (IRAK4), by fragment‐based drug design. J Med Chem 2017;60:5521–42.
    1. Dudhgaonkar S, Ranade S, Nagar J, Subramani S, Prasad DS, Karunanithi P, et al. Selective IRAK4 inhibition attenuates disease in murine lupus models and demonstrates steroid sparing activity. J Immunol 2017;198:1308–19.
    1. Corzo CA, Varfolomeev E, Setiadi AF, Francis R, Klabunde S, Senger K, et al. The kinase IRAK4 promotes endosomal TLR and immune complex signaling in B cells and plasmacytoid dendritic cells. Sci Signal 2020;13:eeaz1053.
    1. Qin J, Jiang Z, Qian Y, Casanova JL, Li X. IRAK4 kinase activity is redundant for interleukin‐1 (IL‐1) receptor‐associated kinase phosphorylation and IL‐1 responsiveness. J Biol Chem 2004;279:26748–53.
    1. Song KW, Talamas FX, Suttmann RT, Olson PS, Barnett JW, Lee SW, et al. The kinase activities of interleukin‐1 receptor associated kinase (IRAK)‐1 and 4 are redundant in the control of inflammatory cytokine expression in human cells. Mol Immunol 2009;46:1458–66.
    1. Cushing L, Stochaj W, Siegel M, Czerwinski R, Dower K, Wright Q, et al. Interleukin 1/Toll‐like receptor‐induced autophosphorylation activates interleukin 1 receptor‐associated kinase 4 and controls cytokine induction in a cell type‐specific manner. J Biol Chem 2014;289:10865–75.
    1. Chiang EY, Yu X, Grogan JL. Immune complex‐mediated cell activation from systemic lupus erythematosus and rheumatoid arthritis patients elaborate different requirements for IRAK1/4 kinase activity across human cell types. J Immunol 2011;186:1279–88.
    1. Sun J, Li N, Oh KS, Dutta B, Vayttaden SJ, Lin B, et al. Comprehensive RNAi‐based screening of human and mouse TLR pathways identifies species‐specific preferences in signaling protein use. Sci Signal 2016;9:ra3.
    1. Danto SI, Shojaee N, Singh RS, Li C, Gilbert SA, Manukyan Z, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of PF‐06650833, a selective interleukin‐1 receptor‐associated kinase 4 (IRAK4) inhibitor, in single and multiple ascending dose randomized phase 1 studies in healthy subjects. Arthritis Res Ther 2019;21:269.
    1. O'Mahony A, John MR, Cho H, Hashizume M, Choy EH. Discriminating phenotypic signatures identified for tocilizumab, adalimumab, and tofacitinib monotherapy and their combinations with methotrexate. J Transl Med 2018;16:156.
    1. Shah F, Stepan AF, O'Mahony A, Velichko S, Folias AE, Houle C, et al. Mechanisms of skin toxicity associated with metabotropic glutamate receptor 5 negative allosteric modulators. Cell Chem Biol 2017;24:858–69.
    1. Rankin AL, Seth N, Keegan S, Andreyeva T, Cook TA, Edmonds J, et al. Selective inhibition of BTK prevents murine lupus and antibody‐mediated glomerulonephritis. J Immunol 2013;191:4540–50.
    1. Yao Y, Higgs BW, Richman L, White B, Jallal B. Use of type I interferon‐inducible mRNAs as pharmacodynamic markers and potential diagnostic markers in trials with sifalimumab, an anti‐IFNα antibody, in systemic lupus erythematosus [review]. Arthritis Res Ther 2010;12 Suppl 1:S6.
    1. Patricelli MP, Nomanbhoy TK, Wu J, Brown H, Zhou D, Zhang J, et al. In situ kinase profiling reveals functionally relevant properties of native kinases. Chem Biol 2011;18:699–710.
    1. Morgan P, Van Der Graaf PH, Arrowsmith J, Feltner DE, Drummond KS, Wegner CD, et al. Can the flow of medicines be improved? Fundamental pharmacokinetic and pharmacological principles toward improving Phase II survival. Drug Discov Today 2012;17:419–24.
    1. Castelar‐Pinheiro GR, Xavier RM. The spectrum and clinical significance of autoantibodies in rheumatoid arthritis. Front Immunol 2015;6:320.
    1. Catrina AI, Svensson CI, Malmström V, Schett G, Klareskog L. Mechanisms leading from systemic autoimmunity to joint‐specific disease in rheumatoid arthritis [review]. Nat Rev Rheumatol 2016;13:79–86.
    1. Sokolove J, Zhao X, Chandra PE, Robinson WH. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll‐like receptor 4 and Fcγ receptor. Arthritis Rheum 2011;63:53–62.
    1. Horwood NJ, Page TH, McDaid JP, Palmer CD, Campbell J, Mahon T, et al. Bruton's tyrosine kinase is required for TLR2 and TLR4‐induced TNF, but not IL‐6, production. J Immunol 2006;176:3635–41.
    1. Falconer J, Murphy AN, Young SP, Clark AR, Tiziani S, Guma M, et al. Synovial cell metabolism and chronic inflammation in rheumatoid arthritis [review]. Arthritis Rheumatol 2018;70:984–99.
    1. Doody KM, Bottini N, Firestein GS. Epigenetic alterations in rheumatoid arthritis fibroblast‐like synoviocytes [review]. Epigenomics 2017;9:479–92.
    1. Pisetsky DS. Anti‐DNA antibodies: quintessential biomarkers of SLE [review]. Nat Rev Rheumatol 2015;12:102–10.
    1. Lee KH, Kronbichler A, Park DD, Park Y, Moon H, Kim H, et al. Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun Rev 2017;16:1160–73.
    1. Bouts YM, Wolthuis DF, Dirkx MF, Pieterse E, Simons EM, van Boekel AM, et al. Apoptosis and NET formation in the pathogenesis of SLE [review]. Autoimmunity 2012;45:597–601.
    1. Mahajan A, Herrmann M, Muñoz LE. Clearance deficiency and cell death pathways: a model for the pathogenesis of SLE [review]. Front Immunol 2016;7:35.
    1. Muñoz LE, Janko C, Schulze C, Schorn C, Sarter K, Schett G, et al. Autoimmunity and chronic inflammation: two clearance‐related steps in the etiopathogenesis of SLE. Autoimmun Rev 2010;10:38–42.
    1. Crow MK. Type I interferon in the pathogenesis of lupus [review]. J Immunol 2014;192:5459–68.
    1. Stone RC, Feng D, Deng J, Singh S, Yang L, Fitzgerald‐Bocarsly P, et al. Interferon regulatory factor 5 activation in monocytes of systemic lupus erythematosus patients is triggered by circulating autoantigens independent of type I interferons. Arthritis Rheum 2012;64:788–98.
    1. Kiefer K, Oropallo MA, Cancro MP, Marshak‐Rothstein A. Role of type I interferons in the activation of autoreactive B cells [review]. Immunol Cell Biol 2012;90:498–504.
    1. Douagi I, Gujer C, Sundling C, Adams WC, Smed‐Sörensen A, Seder RA, et al. Human B cell responses to TLR ligands are differentially modulated by myeloid and plasmacytoid dendritic cells. J Immunol 2009;182:1991–2001.
    1. Cushing L, Winkler A, Jelinsky SA, Lee K, Korver W, Hawtin R, et al. IRAK4 kinase activity controls Toll‐like receptor‐induced inflammation through the transcription factor IRF5 in primary human monocytes. J Biol Chem 2017;292:18689–98.
    1. Colonna M, Trinchieri G, Liu YJ. Plasmacytoid dendritic cells in immunity [review]. Nat Immunol 2004;5:1219–26.
    1. Zhuang H, Szeto C, Han S, Yang L, Reeves WH. Animal models of interferon signature positive lupus [review]. Front Immunol 2015;6:291.
    1. Homer BL, Dower K. 41‐week study of progressive diabetic nephropathy in the ZSF1 fa/faCP rat model. Toxicol Pathol 2018;46:976–7.
    1. Honigberg LA, Smith AM, Sirisawad M, Verner E, Loury D, Chang B, et al. The Bruton tyrosine kinase inhibitor PCI‐32765 blocks B‐cell activation and is efficacious in models of autoimmune disease and B‐cell malignancy. Proc Nat Acad Sci U S A 2010;107:13075–80.
    1. Kim YY, Park KT, Jang SY, Lee KH, Byun JY, Suh KH, et al. HM71224, a selective Bruton's tyrosine kinase inhibitor, attenuates the development of murine lupus. Arthritis Res Ther 2017;19:211.
    1. Chalmers SA, Wen J, Doerner J, Stock A, Cuda CM, Makinde HM, et al. Highly selective inhibition of Bruton's tyrosine kinase attenuates skin and brain disease in murine lupus. Arthritis Res Ther 2018;20:10.
    1. Shi FD, Ljunggren HG, Sarvetnick N. Innate immunity and autoimmunity: from self‐protection to self‐destruction [review]. Trends Immunol 2001;22:97–101.
    1. Waldner H. The role of innate immune responses in autoimmune disease development [review]. Autoimmun Rev 2009;8:400–4.
    1. De Nardo D, Balka KR, Gloria YC, Rao VR, Latz E, Masters SL. Interleukin‐1 receptor–associated kinase 4 (IRAK4) plays a dual role in myddosome formation and Toll‐like receptor signaling. J Biol Chem 2018;293:15195–207.
    1. Pauls E, Nanda SK, Smith H, Toth R, Arthur JS, Cohen P. Two phases of inflammatory mediator production defined by the study of IRAK2 and IRAK1 knock‐in mice. J Immunol 2013;191:2717–30.
    1. Lamba M, Hutmacher MM, Furst DE, Dikranian A, Dowty ME, Conrado D, et al. Model‐informed development and registration of a once‐daily regimen of extended‐release tofacitinib. Clin Pharmacol Ther 2017;101:745–53.
    1. Danto SI, Shojaee N, Singh RS, Manukyan Z, Mancuso J, Peeva E, et al. Efficacy and safety of the selective interleukin‐1 receptor associated kinase 4 inhibitor, PF‐06650833, in patients with active rheumatoid arthritis and inadequate response to methotrexate [abstract]. Arthritis Rheumatol 2019;71 Suppl 10. URL: .
    1. Lazzari E, Jefferies CA. IRF5‐mediated signaling and implications for SLE [review]. Clin Immunol 2014;153:343–52.
    1. Ban T, Sato GR, Tamura T. Regulation and role of the transcription factor IRF5 in innate immune responses and systemic lupus erythematosus [review]. Int Immunol 2018;30:529–36.
    1. Cushing L, Stochaj W, Siegel M, Czerwinski R, Dower K, Wright Q, et al. Interleukin 1/Toll‐like receptor‐induced autophosphorylation activates interleukin 1 receptor‐associated kinase 4 and controls cytokine induction in a cell type‐specific manner. J Biol Chem 2014;289:10865–75.
    1. Otterness IG. The value of C‐reactive protein measurement in rheumatoid arthritis [review]. Semin Arthritis Rheum 1994;24:91–104.
    1. Tishler M, Caspi D, Yaron M. C‐reactive protein levels in patients with rheumatoid arthritis: the impact of therapy. Clin Rheumatol 1985;4:321–4.

Source: PubMed

3
Se inscrever