Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations

Iain B McInnes, Nicole L Byers, Richard E Higgs, Jonathan Lee, William L Macias, Songqing Na, Robert A Ortmann, Guilherme Rocha, Terence P Rooney, Thomas Wehrman, Xin Zhang, Steven H Zuckerman, Peter C Taylor, Iain B McInnes, Nicole L Byers, Richard E Higgs, Jonathan Lee, William L Macias, Songqing Na, Robert A Ortmann, Guilherme Rocha, Terence P Rooney, Thomas Wehrman, Xin Zhang, Steven H Zuckerman, Peter C Taylor

Abstract

Background: The in vitro pharmacology of baricitinib, upadacitinib, and tofacitinib was evaluated to understand differences among these JAK inhibitors (JAKis) at the cellular level.

Methods: Peripheral blood mononuclear cells from healthy donors were incubated with different JAKis, levels of phosphorylated signal transducer and activator of transcription (pSTAT) were measured following cytokine stimulation, and half maximum inhibitory concentration (IC50) values were calculated in phenotypically gated leukocyte subpopulations. Therapeutic dose relevance of the in vitro analysis was assessed using calculated mean concentration-time profiles over 24 h obtained from JAKi-treated subjects. Time above IC50 and average daily percent inhibition of pSTAT formation were calculated for each JAKi, cytokine, and cell type.

Results: Distinct JAKis displayed different in vitro pharmacologic profiles. For example, tofacitinib and upadacitinib were the most potent inhibitors of the JAK1/3-dependent cytokines tested (interleukin [IL]-2, IL-4, IL-15, and IL-21) with lower IC50 values and increased time above IC50 translating to a greater overall inhibition of STAT signaling during the dosing interval. All JAKis tested inhibited JAK1/2-dependent cytokines (e.g., IL-6 and interferon [IFN]-γ), the JAK1/tyrosine kinase 2 (TYK2)-dependent cytokines IL-10 and IFN-α, the JAK2/2-dependent cytokines IL-3 and granulocyte-macrophage colony-stimulating factor (GM-CSF), and the JAK2/TYK2-dependent cytokine granulocyte colony-stimulating factor (G-CSF), but often to significantly differing degrees.

Conclusions: Different JAKis modulated distinct cytokine pathways to varying degrees, and no agent potently or continuously inhibited an individual cytokine signaling pathway throughout the dosing interval. Notably, baricitinib inhibited JAK1/3 signaling to a lesser extent than upadacitinib and tofacitinib, while upadacitinib, baricitinib, and tofacitinib inhibited the signaling of JAK2/2-dependent cytokines, including GM-CSF and IL-3, as well as the signaling of the JAK2/TYK2-dependent cytokine G-CSF.

Keywords: Baricitinib; Cytokine; Janus kinase; Phosphorylated signal transducer and activator of transcription; Potency; Receptor kinase signaling; Rheumatoid arthritis; Selectivity; Tofacitinib; Upadacitinib.

Conflict of interest statement

IBM has received grants and personal fees from AbbVie, Eli Lilly and Company, Janssen, Novartis, Pfizer, and Roche. NLB, REH, WLM, SN, RAO, GR, TPR, XZ, and SHZ are employees and shareholders of Eli Lilly and Company. JL was an employee and shareholder of Eli Lilly and Company when contributions to this article were made. He is now an employee of PDD4Patients LLC. TW is an employee and shareholder of Primity Bio. PCT has received grants and personal fees from Eli Lilly and Company, Galapagos, and UCB. He has received grants from Abide Therapeutics and Celgene and personal fees from AbbVie, Biogen, GSK, Janssen, Novartis, Pfizer, and Sandoz.

Figures

Fig. 1
Fig. 1
JAKi exposure curves for select cytokines. The number of hours per day JAKi concentrations are above IC50 values is shown for IL-21/pSTAT3 in NK cells (a), IL-3/pSTAT5 in monocytes (b), GM-CSF/pSTAT5 in monocytes (c), IL-6/pSTAT3 in CD4+ T cells (d), and IFN-γ/pSTAT1 in monocytes (e). Panels ae include the average daily percent STAT inhibition. Protein binding was accounted for in the calculation of hours per day above IC50 and average daily percent STAT inhibition, and the IC50 values were corrected for the compound-bound proportion. *p < 0.01, **p < 0.001, ***p < 0.0001 compared to baricitinib 2 mg; †p < 0.01, ††p < 0.001, †††p < 0.0001 compared to baricitinib 4 mg. Bari, baricitinib; BID, twice daily; GM-CSF, granulocyte-macrophage colony-stimulating factor; h, hours; IC50, half maximum inhibitory concentration; IFN, interferon; IL, interleukin; JAK, Janus kinase; JAKi, JAK inhibitor; NK, natural killer; pSTAT, phosphorylated STAT; QD, once daily; STAT, signal transducer and activator of transcription; Tofa, tofacitinib; Upa, upadacitinib

References

    1. Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293.
    1. 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. doi: 10.1136/annrheumdis-2016-210715.
    1. Tanaka Y. Current concepts in the management of rheumatoid arthritis. Korean J Intern Med. 2016;31(2):210–218. doi: 10.3904/kjim.2015.137.
    1. Schwartz DM, Bonelli M, Gadina M, et al. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol. 2016;12(1):25–36. doi: 10.1038/nrrheum.2015.167.
    1. McInnes IB, Buckley CD, Isaacs JD. Cytokines in rheumatoid arthritis - shaping the immunological landscape. Nat Rev Rheumatol. 2016;12(1):63–68. doi: 10.1038/nrrheum.2015.171.
    1. Fridman JS, Scherle PA, Collins R, et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol. 2010;184(9):5298–5307. doi: 10.4049/jimmunol.0902819.
    1. Clark JD, Flanagan ME, Telliez JB. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014;57(12):5023–5038. doi: 10.1021/jm401490p.
    1. Voss J, Graff C, Schwartz A, et al. Pharmacodynamics of a novel Jak1 selective inhibitor in rat arthritis and anemia models and in healthy human subjects [abstract]. Arthritis Rheum. 2013;65(supp 10). . Accessed 30 July 2019.
    1. Genovese MC, Smolen JS, Weinblatt ME, et al. Efficacy and safety of ABT-494, a selective JAK-1 inhibitor, in a phase IIb study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Rheumatol. 2016;68(12):2857–2866. doi: 10.1002/art.39808.
    1. Krutzik PO, Nolan GP. Fluorescent cell barcoding in flow cytometry allows high-throughput drug screening and signaling profiling. Nat Methods. 2006;3(5):361–368. doi: 10.1038/nmeth872.
    1. Zhang X, Chua L, Ernest C, et al. Dose/exposure-response modeling to support dosing recommendation for phase lll development of baricitinib in patients with rheumatoid arthritis. CPT Pharmacometrics Syst Pharmacol. 2017;6:804–813. doi: 10.1002/psp4.12251.
    1. Mohamed ME, Zeng J, Song IH, et al. Pharmacokinetics of ABT-494 with the once-daily extended-release tablet formulation being utilized in the ongoing rheumatoid arthritis phase 3 trials [abstract]. Arthritis Rheumatol. 2016;68(suppl 10). . Accessed 30 July 2019.
    1. Klunder B, Mohamed MF, Othman AA. Population pharmacokinetics of upadacitinib in healthy subjects and subjects with rheumatoid arthritis: analyses of phase I and II clinical trials. Clin Pharmacokinet. 2018;57(8):977–988. doi: 10.1007/s40262-017-0605-6.
    1. Clinical pharmacology and biopharmaceutics review_NDA203214. Secondary clinical pharmacology and biopharmaceutics review_NDA203214. 2012. . Accessed 22 Aug 2017.
    1. Steel Jason C., Waldmann Thomas A., Morris John C. Interleukin-15 biology and its therapeutic implications in cancer. Trends in Pharmacological Sciences. 2012;33(1):35–41. doi: 10.1016/j.tips.2011.09.004.
    1. Tangye SG. Advances in IL-21 biology - enhancing our understanding of human disease. Curr Opin Immunol. 2015;34:107–115. doi: 10.1016/j.coi.2015.02.010.
    1. Hodge JA, Kawabata TT, Krishnaswami S, et al. The mechanism of action of tofacitinib - an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol. 2016;34(2):318–328.
    1. Kremer JM, Emery P, Camp HS, et al. A phase IIb study of ABT-494, a selective JAK-1 inhibitor, in patients with rheumatoid arthritis and an inadequate response to anti-tumor necrosis factor therapy. Arthritis Rheumatol. 2016;68(12):2867–2877. doi: 10.1002/art.39801.
    1. Tanaka Y, McInnes IB, Taylor PC, et al. Characterization and changes of lymphocyte subsets in baricitinib-treated patients with rheumatoid arthritis: an integrated analysis. Arthritis Rheumatol. 2018;70(12):1923–1932. doi: 10.1002/art.40680.
    1. Wicks IP, Roberts AW. Targeting GM-CSF in inflammatory diseases. Nat Rev Rheumatol. 2016;12(1):37–48. doi: 10.1038/nrrheum.2015.161.
    1. Burmester GR, Weinblatt ME, McInnes IB, et al. Efficacy and safety of mavrilimumab in subjects with rheumatoid arthritis. Ann Rheum Dis. 2013;72(9):1445–1452. doi: 10.1136/annrheumdis-2012-202450.
    1. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365(23):2205–2219. doi: 10.1056/NEJMra1004965.
    1. Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-controlled, randomised trial. Lancet. 2008;371(9617):987–997. doi: 10.1016/S0140-6736(08)60453-5.
    1. Castillo P, Kolls JK. IL-10: a paradigm for counterregulatory cytokines. J Immunol. 2016;197(5):1529–1530. doi: 10.4049/jimmunol.1601192.

Source: PubMed

3
Se inscrever