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

Spencer I Danto, Negin Shojaee, Ravi Shankar P Singh, Cheryl Li, Steven A Gilbert, Zorayr Manukyan, Iain Kilty, Spencer I Danto, Negin Shojaee, Ravi Shankar P Singh, Cheryl Li, Steven A Gilbert, Zorayr Manukyan, Iain Kilty

Abstract

Background: PF-06650833 is a potent, selective inhibitor of interleukin-1 receptor-associated kinase 4 (IRAK4). Two randomized, double-blind, sponsor-open phase 1 studies evaluated the safety, pharmacokinetics, and pharmacodynamics of single (SAD) and multiple ascending doses (MAD) of PF-06650833 immediate-release (IR) and modified-release (MR) oral formulations in healthy adult subjects.

Methods: Study 1 (NCT02224651) was a 96-day, placebo-substitution, SAD study of once-daily (QD) oral PF-06650833 IR 1 to 6000 mg and MR 30 to 300 mg in fasted and fed states. Study 2 (NCT02485769) was a 14-day, placebo-controlled, MAD study of PF-06650833 IR 25 to 750 mg twice daily, IR 1000 mg four times per day, IR 330 mg three times per day, and MR 300 mg QD.

Results: PF-06650833 was generally well tolerated, with no dose-limiting treatment-emergent adverse events (TEAEs) identified in either study. TEAEs were generally mild in severity, with headache, gastrointestinal disorders, and acne most commonly reported. No serious AEs or deaths were reported. A maximum tolerated dose was not established in either study. In the SAD study, food intake delayed absorption of IR 30 mg and increased total exposure by 33%. Delayed absorption was achieved with the MR formulation (Tmax of 1 h versus 8 h for IR 100 mg and MR 100 mg formulations, respectively). Food had no effect on total exposure for MR 30 mg, but reduced half-life 1.8-fold and increased Cmax by 62%. In the MAD study, accumulation ranged from 0.9-fold to 1.4-fold for AUCtau and 0.9-fold to 1.3-fold for Cmax. Less than 1% of the dose was recovered unchanged in urine for all dose groups, with renal clearance ranging from 14 to 23 mL/min for IR < 750 mg and MR 300 mg. There was a sustained decrease in serum high-sensitivity C-reactive protein for IR ≥ 250 mg and MR 300 mg. Based on the cholesterol/hydroxycholesterol ratio, no apparent CYP3A induction or inhibition was observed.

Conclusions: PF-06650833, the first IRAK4 inhibitor to enter clinical development, has a favorable safety and pharmacokinetic profile and has shown evidence of pharmacological effect. The data support continued evaluation in human clinical trials for the treatment of rheumatic and autoimmune diseases.

Trial registration: Clinicaltrials.gov, NCT02224651, registered 25 August 2014; NCT02485769, registered 30 June 2015.

Keywords: IRAK4; Pharmacodynamic; Pharmacokinetic.

Conflict of interest statement

All authors are current or past employees of Pfizer Inc. and hold shares in Pfizer Inc.

Figures

Fig. 1
Fig. 1
Design and PF-06650833 final dosing scheme in a study 1 (SAD) and b study 2 (MAD). aPK and PD sampling time was up to 96 h for cohorts 1 and 2. Subjects in cohorts 3 and 4 were followed up to day 21 of the final period to better characterize the terminal phase, given the potentially long elimination half-life based on emerging data. bDose administered after consumption of a high-fat breakfast meal. cAlternate IR formulation. dCohort 3 consisted of only four periods, and cohort 4 consisted of only two periods that were separated by 14 days, in order to maintain the overall predicted exposure in an individual subject to ≤ 28 days. In study 1, within each period, 8 subjects were randomized to receive PF-06650833 and 2 subjects were randomized to receive placebo. All subjects within a cohort received one or more doses of PF-06650833 and/or placebo. Doses were escalated sequentially within each period, based on evaluation of ≥ 48 h of safety and tolerability for all subjects and ≥ 8 h of PK data for at least 6 subjects receiving PF-06650833 and 1 subject receiving placebo. All doses were administered orally under fasting conditions (overnight fast of ≥ 10 h) unless otherwise indicated. In study 2, within each cohort, eight subjects were planned to receive PF-06650833 and 2 subjects were planned to receive placebo. All doses were administered orally under standard (not high-fat) meal, fed conditions. QD doses were 24 h apart, BID doses were 12 h apart, TID doses were 8 h apart, and QID doses were 6 h apart. When dosing in the fed condition, the morning and evening doses were administered within 5 min of completing the standard meal. BID twice daily; IR immediate-release: MAD multiple ascending doses; MR modified-release; PK pharmacokinetics; QD once daily; QID four times per day; SAD single ascending doses; TID three times per day
Fig. 2
Fig. 2
Median plasma concentration-time profile of SAD of PF-06650833 a IR doses ≤ 100 mg, b IR doses > 100 mg, and c MR formulations. All doses were administered orally under fasting conditions (overnight fast of ≥ 10 h) unless otherwise indicated. Fed doses were administered after consumption of a high-fat breakfast meal. Summary statistics were calculated by setting concentration values below the LLOQ to 0. The LLOQ was 0.0500 ng/mL. IR immediate-release; LLOQ lower limit of quantification; MR modified-release; SAD single ascending doses
Fig. 3
Fig. 3
Median plasma concentration-time profile of MAD of PF-06650833 IR and MR formulations at steady state on day 14. Time post-dose refers to the first morning dose on day 14. Day 14 data for cohort 5 (IR 1000 mg QID) were not available due to premature discontinuation of this cohort on day 9. Summary statistics were calculated by setting concentration values BLQ to 0. The LLOQ was 0.0500 ng/mL, except four pre-dose samples with LLOQ of 0.100 ng/mL. All doses were administered orally under fed conditions (standard meal). BID twice daily; BLQ below lower limit of quantification; IR immediate-release; LLOQ lower limit of quantification; MAD multiple ascending doses; MR modified-release; QD once daily; QID four times per day; TID three times per day
Fig. 4
Fig. 4
Geometric mean (90% CI) change from baseline in serum hsCRP following MAD of IR and MR PF-06650833 formulations. Baseline was defined as the last pre-dose measurement taken on day 1. Time post-dose refers to the first morning dose. Values below the LLOQ were set to half of the LLOQ in the calculation. The LLOQ of hsCRP was 0.015 mg/dL. Unplanned readings and early withdrawal readings are excluded. The dosing in 1000 mg QID dose group was stopped by the sponsor after the second dose on day 9, and the subjects had their follow-up visits 2 days (approximately 43 h) and 13 days (approximately 310 h) after the last dose on day 9. BID twice daily; CI confidence interval; D day; h hour; hsCRP high-sensitivity C-reactive protein; IR immediate-release; LLOQ lower limit of quantification; MAD multiple ascending doses

References

    1. Cross M, Smith E, Hoy D, Carmona L, Wolfe F, Vos T, et al. The global burden of rheumatoid arthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis. 2014;73:1316–1322. doi: 10.1136/annrheumdis-2013-204627.
    1. Smith E, Hoy DG, Cross M, Vos T, Naghavi M, Buchbinder R, et al. The global burden of other musculoskeletal disorders: estimates from the Global Burden of Disease 2010 study. Ann Rheum Dis. 2014;73:1462–1469. doi: 10.1136/annrheumdis-2013-204680.
    1. Franklyn K, Hoi A, Nikpour M, Morand EF. The need to define treatment goals for systemic lupus erythematosus. Nat Rev Rheumatol. 2014;10:567–571. doi: 10.1038/nrrheum.2014.118.
    1. Rees F, Doherty M, Grainge MJ, Lanyon P, Zhang W. The worldwide incidence and prevalence of systemic lupus erythematosus: a systematic review of epidemiological studies. Rheumatology. 2017;56:1945–1961. doi: 10.1093/rheumatology/kex260.
    1. Smolen JS, Breedveld FC, Burmester GR, Bykerk V, Dougados M, Emery P, et al. Treating rheumatoid arthritis to target: 2014 update of the recommendations of an international task force. Ann Rheum Dis. 2016;75:3–15. doi: 10.1136/annrheumdis-2015-207524.
    1. van Vollenhoven RF, Mosca M, Bertsias G, Isenberg D, Kuhn A, Lerstrøm K, et al. Treat-to-target in systemic lupus erythematosus: recommendations from an international task force. Ann Rheum Dis. 2014;73:958–967. doi: 10.1136/annrheumdis-2013-205139.
    1. Sokka T, Hetland ML, Mäkinen H, Kautiainen H, Hørslev-Petersen K, Luukkainen RK, et al. Remission and rheumatoid arthritis: data on patients receiving usual care in twenty-four countries. Arthritis Rheum. 2008;58:2642–2651. doi: 10.1002/art.23794.
    1. Khan NA, Sokka T. American College of Rheumatology/European League Against Rheumatism remission criteria achievement in rheumatoid arthritis: data from 32 countries. In: 2011 ACR/ARHP Annual Scientific Meeting Chicago, IL, USA, November 8, 2011. vol. 2462; 2011.
    1. Taylor PC, Moore A, Vasilescu R, Alvir J, Tarallo M. A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: a current perspective. Rheumatol Int. 2016;36:685–695. doi: 10.1007/s00296-015-3415-x.
    1. Kawai T, Akira S. TLR signaling. Cell Death Differ. 2006;13:816–825. doi: 10.1038/sj.cdd.4401850.
    1. Maglione PJ, Simchoni N, Black S, Radigan L, Overbey JR, Bagiella E, et al. IRAK-4 and MyD88 deficiencies impair IgM responses against T-independent bacterial antigens. Blood. 2014;124:3561–3571. doi: 10.1182/blood-2014-07-587824.
    1. Sokolove J, Zhao X, Chandra PE, Robinson WH. Immune complexes containing citrullinated fibrinogen costimulate macrophages via Toll-like receptor 4 and Fcgamma receptor. Arthritis Rheum. 2011;63:53–62. doi: 10.1002/art.30081.
    1. Sakkas LI, Bogdanos DP, Katsiari C, Platsoucas CD. Anti-citrullinated peptides as autoantigens in rheumatoid arthritis—relevance to treatment. Autoimmun Rev. 2014;13:1114–1120. doi: 10.1016/j.autrev.2014.08.012.
    1. Kono DH, Haraldsson MK, Lawson BR, Pollard KM, Koh YT, Du X, et al. Endosomal TLR signaling is required for anti-nucleic acid and rheumatoid factor autoantibodies in lupus. Proc Natl Acad Sci U S A. 2009;106:12061–12066. doi: 10.1073/pnas.0905441106.
    1. Guiducci C, Gong M, Xu Z, Gill M, Chaussabel D, Meeker T, et al. TLR recognition of self nucleic acids hampers glucocorticoid activity in lupus. Nature. 2010;465:937–941. doi: 10.1038/nature09102.
    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-methoxyisoquinoli ne-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;60:5521–5542. doi: 10.1021/acs.jmedchem.7b00231.
    1. Kelly PN, Romero DL, Yang Y, Shaffer AL, 3rd, 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;212:2189–2201. doi: 10.1084/jem.20151074.
    1. Tumey LN, Boschelli DH, Bhagirath N, Shim J, Murphy EA, Goodwin D, et al. Identification and optimization of indolo[2,3-c]quinoline inhibitors of IRAK4. Bioorg Med Chem Lett. 2014;24:2066–2072. doi: 10.1016/j.bmcl.2014.03.056.
    1. McElroy WT, Tan Z, Ho G, Paliwal S, Li G, Seganish WM, et al. Potent and selective amidopyrazole inhibitors of IRAK4 that are efficacious in a rodent model of inflammation. ACS Med Chem Lett. 2015;6:677–682. doi: 10.1021/acsmedchemlett.5b00106.
    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;292:18689–18698. doi: 10.1074/jbc.M117.796912.
    1. Diczfalusy U, Nylén H, Elander P, Bertilsson L. 4β-Hydroxycholesterol, an endogenous marker of CYP3A4/5 activity in humans. Br J Clin Pharmacol. 2011;71:183–189. doi: 10.1111/j.1365-2125.2010.03773.x.

Source: PubMed

3
Se inscrever