Bioequivalence of the pharmacokinetics between tofacitinib aspartate and tofacitinib citrate in healthy subjects

Wonsuk Shin, A-Young Yang, Hyeonji Yun, Doo-Yeoun Cho, Kyung Hee Park, Hyunju Shin, Anhye Kim, Wonsuk Shin, A-Young Yang, Hyeonji Yun, Doo-Yeoun Cho, Kyung Hee Park, Hyunju Shin, Anhye Kim

Abstract

Tofacitinib is an oral disease-modifying anti-rheumatic drug to selectively inhibit Janus kinases. Tofacitinib is a representative small molecule inhibitor that is used to treat many diseases including rheumatoid arthritis and various autoimmune conditions. Unlike biological agents, tofacitinib has several advantages, including the ability to be administered orally and a short half-life. This study aimed to evaluate the bioequivalence of the pharmacokinetics (PK) between tofacitinib aspartate 7.13 mg (test formulation) and tofacitinib citrate 8.08 mg (reference formulation; Xeljanz®) in healthy subjects. A randomized, open-label, single-dose, 2-sequence, 2-period, 2-treatment crossover trial was conducted in 41 healthy volunteers. A total of 5 mg of tofacitinib as the test or the reference formulation was administered, and serial blood samples were collected up to 14 hours after dosing for PK analyses. The plasma concentration of tofacitinib was determined by ultra-performance liquid chromatography-tandem mass spectrometry. A non-compartmental analysis was used to estimate the PK parameters. A total of 35 subjects completed the study and the study drug was well-tolerated. The mean maximum concentration (Cmax) and area under the concentration-time curve from time zero to the time of the last quantifiable concentration (AUClast) for the test formulation were 52.67 ng/mL and 133.86 ng∙h/mL, respectively, and 50.61 ng/mL and 133.49 h∙ng/mL for the reference formulation, respectively. The geometric mean ratios (90% confidence intervals) of the Cmax and AUClast between the 2 formulations were 1.041 (0.944-1.148) and 1.003 (0.968-1.039), respectively. Tofacitinib aspartate exhibited bioequivalent PK profiles to those of the reference formulation.

Trial registration: ClinicalTrials.gov Identifier: NCT04278391.

Keywords: Bioequivalence; Pharmacokinetics; Tofacitinib.

Conflict of interest statement

Conflict of Interest: - Authors: KH Park, and H Shin are the employees of Daewoong Pharmaceutical Co., Ltd. The other authors report no conflicts of interest related to this work. - Reviewers: Nothing to declare - Editors: Nothing to declare

Copyright © 2020 Translational and Clinical Pharmacology.

Figures

Figure 1. Mean ± standard deviation plasma…
Figure 1. Mean ± standard deviation plasma concentration-time profiles of Tofacitinib after single doses of the reference (white triangles) and test formulations (black circles) in healthy subject (A, linear scale; B, log scale).
Figure 2. Individual (A) C max and…
Figure 2. Individual (A) Cmax and (B) AUClast of Tofacitinib after single doses of the reference and test formulations of Tofacitinib in healthy subjects.
Cmax, maximum plasma concentration of drug; AUClast, area under the plasma concentration-time curve from time 0 to the time of the last quantifiable concentration.

References

    1. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet. 2010;376:1094–1108.
    1. Tiippana-Kinnunen T, Kautiainen H, Paimela L, Leirisalo-Repo M. Co-morbidities in Finnish patients with rheumatoid arthritis: 15-year follow-up. Scand J Rheumatol. 2013;42:451–456.
    1. Singh JA, Saag KG, Bridges SL, Jr, Akl EA, Bannuru RR, Sullivan MC, et al. 2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Rheumatol. 2016;68:1–26.
    1. Smolen JS, Landewé R, Bijlsma J, Burmester G, Chatzidionysiou K, Dougados M, et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis. 2017;76:960–977.
    1. Kaito T, Ohshima S, Fujiwara H, Makino T, Yonenobu K, Yoshikawa H. Incidence and risk factors for cervical lesions in patients with rheumatoid arthritis under the current pharmacologic treatment paradigm. Mod Rheumatol. 2017;27:593–597.
    1. Scott LJ. Tofacitinib: a review of its use in adult patients with rheumatoid arthritis. Drugs. 2013;73:857–874.
    1. Harnett J, Curtis JR, Gerber R, Gruben D, Koenig A. Initial experience with tofacitinib in clinical practice: treatment patterns and costs of tofacitinib administered as monotherapy or in combination with conventional synthetic DMARDs in 2 US health care claims databases. Clin Ther. 2016;38:1451–1463.
    1. Tanaka Y. Current concepts in the management of rheumatoid arthritis. Korean J Intern Med. 2016;31:210–218.
    1. Dhillon S. Tofacitinib: a review in rheumatoid arthritis. Drugs. 2017;77:1987–2001.
    1. Fleischmann R. A review of tofacitinib efficacy in rheumatoid arthritis patients who have had an inadequate response or intolerance to methotrexate. Expert Opin Pharmacother. 2017;18:1525–1533.
    1. Pfizer Inc. XELJANZ® (tofacitinib) tablets for oral administration: US prescribing information. [Accessed March 11, 2013]. .
    1. Mease P, Hall S, FitzGerald O, van der Heijde D, Merola JF, Avila-Zapata F, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377:1537–1550.
    1. Sandborn WJ, Ghosh S, Panes J, Vranic I, Su C, Rousell S, et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med. 2012;367:616–624.
    1. Cutolo M, Meroni M. Clinical utility of the oral JAK inhibitor tofacitinib in the treatment of rheumatoid arthritis. J Inflamm Res. 2013;6:129–137.
    1. Lamba M, Wang R, Fletcher T, Alvey C, Kushner J, 4th, 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;56:1362–1371.
    1. Smith G. Bioanalytical method validation: notable points in the 2009 draft EMA Guideline and differences with the 2001 FDA Guidance. Bioanalysis. 2010;2:929–935.
    1. Ministry of Food and Drug Safety. Casebook of approved incrementally modified drug. Cheongju: Ministry of Food and Drug Safety; 2019.
    1. Serajuddin AT. Salt formation to improve drug solubility. Adv Drug Deliv Rev. 2007;59:603–616.
    1. Schmidt J. Feasibility study: fast liquid chromatography-mass spectrometry for the quantification of aspartic acid in an aspartate drug. Anal Bioanal Chem. 2003;377:1120–1123.
    1. Ranade VV, Somberg JC. Bioavailability and pharmacokinetics of magnesium after administration of magnesium salts to humans. Am J Ther. 2001;8:345–357.
    1. Coudray C, Rambeau M, Feillet-Coudray C, Gueux E, Tressol JC, Mazur A, et al. Study of magnesium bioavailability from ten organic and inorganic Mg salts in Mg-depleted rats using a stable isotope approach. Magnes Res. 2005;18:215–223.
    1. Burton ME. Applied pharmacokinetics & pharmacodynamics: principles of therapeutic drug monitoring. Philadelphia (PA): Lippincott Williams & Wilkins; 2006. p. 799.
    1. Fine KD, Santa Ana CA, Fordtran JS. Diagnosis of magnesium-induced diarrhea. N Engl J Med. 1991;324:1012–1017.
    1. Steinberg W. Magnesium-induced diarrhea: more firm data on loose stools. Am J Gastroenterol. 1992;87:675–676.

Source: PubMed

3
구독하다