260
Views
12
CrossRef citations to date
0
Altmetric
Review

Bruton’s Tyrosine Kinase Inhibition for the Treatment of Rheumatoid Arthritis

ORCID Icon, ORCID Icon & ORCID Icon
Pages 333-342 | Published online: 28 Aug 2021

References

  • Ruderman EM, Pope RM. More than just B-cell inhibition. Arthritis Res Ther. 2011;13(4):125. doi:10.1186/ar3439
  • Jones NS, Winter H, Katsumoto TR, et al. Absence of pharmacokinetic interactions between the Bruton’s tyrosine kinase inhibitor fenebrutinib and methotrexate. J Pharmacol Exp Ther. 2019;371(1):202–207. doi:10.1124/jpet.119.257089
  • Satterthwaite AB, Witte ON. The role of Bruton’s tyrosine kinase in B-cell development and function: a genetic perspective. Immunol Rev. 2000;175:120–127. doi:10.1111/j.1600-065X.2000.imr017504.x
  • Brunner C, Müller B, Wirth T. Bruton’s tyrosine kinase is involved in innate and adaptive immunity. Histol Histopathol. 2005;20(3):945–955. doi:10.14670/hh-20.945
  • Park JK, Byun JY, Park JA, et al. HM71224, a novel Bruton’s tyrosine kinase inhibitor, suppresses B cell and monocyte activation and ameliorates arthritis in a mouse model: a potential drug for rheumatoid arthritis. Arthritis Res Ther. 2016;18:91. doi:10.1186/s13075-016-0988-z
  • Liu L, Di Paolo J, Barbosa J, Rong H, Reif K, Wong H. Antiarthritis effect of a novel Bruton’s tyrosine kinase (BTK) inhibitor in rat collagen-induced arthritis and mechanism-based pharmacokinetic/pharmacodynamic modeling: relationships between inhibition of BTK phosphorylation and efficacy. J Pharmacol Exp Ther. 2011;338(1):154–163. doi:10.1124/jpet.111.181545
  • Kelly V, Genovese M. Novel small molecule therapeutics in rheumatoid arthritis. Rheumatology. 2013;52(7):1155–1162. doi:10.1093/rheumatology/kes367
  • Evans EK, Tester R, Aslanian S, et al. Inhibition of Btk with CC-292 provides early pharmacodynamic assessment of activity in mice and humans. J Pharmacol Exp Ther. 2013;346(2):219–228. doi:10.1124/jpet.113.203489
  • Niiro H, Clark EA. Regulation of B-cell fate by antigen-receptor signals. Nat Rev Immunol. 2002;2(12):945–956. doi:10.1038/nri955
  • Rip J, de Bruijn MJW, Appelman MK, Pal Singh S, Hendriks RW, Corneth OBJ. Toll-like receptor signaling drives btk-mediated autoimmune disease. Front Immunol. 2019;10:95. doi:10.3389/fimmu.2019.00095
  • Schafer PH, Kivitz AJ, Ma J, et al. Spebrutinib (CC-292) affects markers of B cell activation, chemotaxis, and osteoclasts in patients with rheumatoid arthritis: Results from a Mechanistic Study. Rheumatol Ther. 2020;7(1):101–119. doi:10.1007/s40744-019-00182-7
  • Wang SP, Iwata S, Nakayamada S, et al. Amplification of IL-21 signalling pathway through Bruton’s tyrosine kinase in human B cell activation. Rheumatology. 2015;54(8):1488–1497. doi:10.1093/rheumatology/keu532
  • Gillooly KM, Pulicicchio C, Pattoli MA, et al. Bruton’s tyrosine kinase inhibitor BMS-986142 in experimental models of rheumatoid arthritis enhances efficacy of agents representing clinical standard-of-care. PLoS One. 2017;12(7):e0181782. doi:10.1371/journal.pone.0181782
  • Shinohara M, Chang BY, Buggy JJ, et al. The orally available Btk inhibitor ibrutinib (PCI-32765) protects against osteoclast-mediated bone loss. Bone. 2014;60:8–15. doi:10.1016/j.bone.2013.11.025
  • Wu J, Liu C, Tsui ST, Liu D. Second-generation inhibitors of Bruton tyrosine kinase. J Hematol Oncol. 2016;9(1):80. doi:10.1186/s13045-016-0313-y
  • Crawford JJ, Johnson AR, Misner DL, et al. Discovery of GDC-0853: a potent, selective, and noncovalent Bruton’s tyrosine kinase inhibitor in early clinical development. J Med Chem. 2018;61(6):2227–2245. doi:10.1021/acs.jmedchem.7b01712
  • Boga SB, Alhassan AB, Liu J, et al. Discovery of 3-morpholino-imidazole[1,5-a]pyrazine BTK inhibitors for rheumatoid arthritis. Bioorg Med Chem Lett. 2017;27(16):3939–3943. doi:10.1016/j.bmcl.2017.03.040
  • Caldwell R, Liu-Bujalski L, Qiu H, et al. Discovery of a novel series of pyridine and pyrimidine carboxamides as potent and selective covalent inhibitors of Btk. Bioorg Med Chem Lett. 2018;28(21):3419–3424. doi:10.1016/j.bmcl.2018.09.033
  • Abdelhameed AS, Attwa MW, Al-Shaklia NS, Kadi AA. A highly sensitive LC-MS/MS method to determine novel Bruton’s tyrosine kinase inhibitor spebrutinib: application to metabolic stability evaluation. R Soc Open Sci. 2019;6(6):190434. doi:10.1098/rsos.190434
  • Ariza Y, Murata M, Ueda Y, Yoshizawa T. Bruton’s tyrosine kinase (Btk) inhibitor tirabrutinib suppresses osteoclastic bone resorption. Bone Rep. 2019;10:100201. doi:10.1016/j.bonr.2019.100201
  • Caldwell RD, Qiu H, Askew BC, et al. Discovery of evobrutinib: an oral, potent, and highly selective, covalent Bruton’s Tyrosine Kinase (BTK) inhibitor for the treatment of immunological diseases. J Med Chem. 2019;62(17):7643–7655. doi:10.1021/acs.jmedchem.9b00794
  • Haselmayer P, Camps M, Liu-Bujalski L, et al. Efficacy and pharmacodynamic modeling of the BTK inhibitor evobrutinib in autoimmune disease models. J Immunol. 2019;202(10):2888–2906. doi:10.4049/jimmunol.1800583
  • Becker A, Martin EC, Mitchell DY, et al. Safety, tolerability, pharmacokinetics, target occupancy, and concentration-QT analysis of the novel BTK inhibitor evobrutinib in healthy volunteers. Clin Transl Sci. 2020;13(2):325–336. doi:10.1111/cts.12713
  • Genovese M, Spindler A, Sagawa A, et al. Safety and efficacy of poseltinib, Bruton’s tyrosine kinase inhibitor, in patients with rheumatoid arthritis: a randomized, double-blind, placebo-controlled, 2-part Phase II Study. J Rheumatol. 2021;48(7):969–976. doi:10.3899/jrheum.200893
  • Liu YT, Ding HH, Lin ZM, et al. A novel tricyclic BTK inhibitor suppresses B cell responses and osteoclastic bone erosion in rheumatoid arthritis. Acta Pharmacol Sin. 2021. doi:10.1038/s41401-020-00578-0
  • Cho H, Lee E, Kwon HA, et al. Discovery of tricyclic pyranochromenone as novel Bruton’s tyrosine kinase inhibitors with in vivo antirheumatic activity. Int J Mol Sci. 2020;21(21):7919. doi:10.3390/ijms21217919
  • Xu D, Kim Y, Postelnek J, et al. RN486, a selective Bruton’s tyrosine kinase inhibitor, abrogates immune hypersensitivity responses and arthritis in rodents. J Pharmacol Exp Ther. 2012;341(1):90–103. doi:10.1124/jpet.111.187740
  • Young WB, Barbosa J, Blomgren P, et al. Potent and selective Bruton’s tyrosine kinase inhibitors: discovery of GDC-0834. Bioorg Med Chem Lett. 2015;25(6):1333–1337. doi:10.1016/j.bmcl.2015.01.032
  • Sodhi JK, Wong S, Kirkpatrick DS, et al. A novel reaction mediated by human aldehyde oxidase: amide hydrolysis of GDC-0834. Drug Metab Dispos. 2015;43(6):908–915. doi:10.1124/dmd.114.061804
  • Saffran DC, Parolini O, Fitch-Hilgenberg ME, et al. Brief report: a point mutation in the SH2 domain of Bruton’s tyrosine kinase in atypical X-linked agammaglobulinemia. N Engl J Med. 1994;330(21):1488–1491. doi:10.1056/NEJM199405263302104
  • Di Paolo JA, Huang T, Balazs M, et al. Specific Btk inhibition suppresses B cell- and myeloid cell-mediated arthritis. Nat Chem Biol. 2011;7(1):41–50. doi:10.1038/nchembio.481
  • ACP-196 versus placebo in subjects with rheumatoid arthritis on background methotrexate. Available from: https://ClinicalTrials.gov/show/NCT02387762. Accessed August 4, 2021.
  • Cohen S, Tuckwell K, Katsumoto TR, et al. Fenebrutinib versus placebo or adalimumab in rheumatoid arthritis: a randomized, double-blind, Phase II trial. Arthritis Rheumatol. 2020;72(9):1435–1446. doi:10.1002/art.41275
  • Study to evaluate safety and pharmacokinetics of GS-4059 (tirabrutinib) in healthy volunteers and participants with Rheumatoid Arthritis (RA). Available from: https://ClinicalTrials.gov/show/NCT02626026. Accessed August 4, 2021.
  • Safety and Efficacy Study of M2951 in participants with rheumatoid arthritis. Available from: https://ClinicalTrials.gov/show/NCT02784106. Accessed August 4, 2021.
  • Phase IIb Study of evobrutinib in subjects with rheumatoid arthritis. Available from: https://ClinicalTrials.gov/show/NCT03233230. Accessed August 4, 2021.
  • Herman AE, Chinn LW, Kotwal SG, et al. Safety, pharmacokinetics, and pharmacodynamics in healthy volunteers treated with GDC-0853, a selective reversible Bruton’s tyrosine kinase inhibitor. Clin Pharmacol Ther. 2018;103(6):1020–1028. doi:10.1002/cpt.1056
  • Liu L, Halladay JS, Shin Y, et al. Significant species difference in amide hydrolysis of GDC-0834, a novel potent and selective Bruton’s tyrosine kinase inhibitor. Drug Metab Dispos. 2011;39(10):1840–1849. doi:10.1124/dmd.111.040840
  • Lorenzo-Vizcaya A, Fasano S, Isenberg DA. Bruton’s tyrosine kinase inhibitors: a new therapeutic target for the treatment of SLE? Immunotargets Ther. 2020;9:105–110. doi:10.2147/itt.S240874
  • Mina-Osorio P, LaStant J, Keirstead N, et al. Suppression of glomerulonephritis in lupus-prone NZB x NZW mice by RN486, a selective inhibitor of Bruton’s tyrosine kinase. Arthritis Rheum. 2013;65(9):2380–2391. doi:10.1002/art.38047
  • Hutcheson J, Vanarsa K, Bashmakov A, et al. Modulating proximal cell signaling by targeting Btk ameliorates humoral autoimmunity and end-organ disease in murine lupus. Arthritis Res Ther. 2012;14(6):R243. doi:10.1186/ar4086
  • Kim YY, Park KT, Jang SY, et al. HM71224, a selective Bruton’s tyrosine kinase inhibitor, attenuates the development of murine lupus. Arthritis Res Ther. 2017;19(1):211. doi:10.1186/s13075-017-1402-1
  • Isenberg D, Furie R, Jones N, et al. Efficacy, safety, and pharmacodynamic effects of the Bruton’s tyrosine kinase inhibitor, fenebrutinib (GDC-0853), in moderate to severe systemic lupus erythematosus: results of a phase 2 randomized controlled trial [abstract]. Arthritis Rheum. 2019;71(10).
  • American Laboratory Products Company. BTK Inhibitors as Cancer Drug Treatments. Available from: https://www.alpco.com/btk-inhibitors-as-cancer-drug-treatments. Accessed August 13, 2021.