783
Views
15
CrossRef citations to date
0
Altmetric
Review

PK, PD, and interactions: the new scenario with JAK inhibitors and S1P receptor modulators, two classes of small molecule drugs, in IBD

, , , , , , , & ORCID Icon show all
Pages 797-806 | Received 02 Apr 2020, Accepted 16 Jun 2020, Published online: 01 Jul 2020

References

  • Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009 Nov 19;361(21):2066–2078.
  • de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016 Jan;13(1):13–27.
  • Roda G, Jharap B, Neeraj N, et al. Loss of response to anti-TNFs: definition, epidemiology, and management. Clin Transl Gastroenterol. 2016 Jan 7;7:e135.
  • Gisbert JP, Chaparro M. Predictors of primary response to biologic treatment (anti-TNF, vedolizumab and ustekinumab) in patients with inflammatory bowel disease: from basic science to clinical practice. J Crohns Colitis. 2019 Nov 28:jjz195. DOI:10.1093/ecco-jcc/jjz195.
  • Sabino J, Verstockt B, Vermeire S, et al. New biologics and small molecules in inflammatory bowel disease: an update. Therap Adv Gastroenterol. 2019;12:1756284819853208.
  • Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2018 Dec 23;390(10114):2769–2778.
  • Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015 Dec;12(12):720–727.
  • Paramsothy S, Rosenstein AK, Mehandru S, et al. The current state of the art for biological therapies and new small molecules in inflammatory bowel disease. Mucosal Immunol. 2018 Nov;11(6):1558–1570.
  • Ma C, Battat R, Dulai PS, et al. Innovations in oral therapies for inflammatory bowel disease. Drugs. 2019 Aug;79(12):1321–1335.
  • Rekic D, Reynolds KS, Zhao P, et al. Clinical Drug-Drug Interaction Evaluations to Inform Drug Use and Enable Drug Access. J Pharm Sci. 2017 Sep;106(9):2214–2218.
  • Deng R, Jin F, Prabhu S, et al. Monoclonal antibodies: what are the pharmacokinetic and pharmacodynamic considerations for drug development? Expert Opin Drug Metab Toxicol. 2012 Feb;8(2):141–160.
  • Ordas I, Mould DR, Feagan BG, et al. Anti-TNF monoclonal antibodies in inflammatory bowel disease: pharmacokinetics-based dosing paradigms. Clin Pharmacol Ther. 2012 Apr;91(4):635–646.
  • Ferl GZ, Theil FP, Wong H. Physiologically based pharmacokinetic models of small molecules and therapeutic antibodies: a mini-review on fundamental concepts and applications. Biopharm Drug Dispos. 2016 Mar;37(2):75–92.
  • Posner J, Barrington P, Brier T, et al. Monoclonal Antibodies: past, Present and Future. Handb Exp Pharmacol. 2019;260:81–141.
  • Hemperly A, Sandborn WJ, Vande Casteele N. Clinical pharmacology in adult and pediatric inflammatory bowel disease. Inflamm Bowel Dis. 2018 Nov 29;24(12):2527–2542.
  • Wang W, Wang EQ, Balthasar JP. Monoclonal antibody pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther. 2008 Nov;84(5):548–558.
  • Garg A, Balthasar JP. Physiologically-based pharmacokinetic (PBPK) model to predict IgG tissue kinetics in wild-type and FcRn-knockout mice. J Pharmacokinet Pharmacodyn. 2007 Oct;34(5):687–709.
  • Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov Today. 2006 Jan;11(1–2):81–88.
  • Perez-Jeldres T, Tyler CJ, Boyer JD, et al. Targeting cytokine signaling and lymphocyte traffic via small molecules in inflammatory bowel disease: JAK inhibitors and S1PR agonists. Front Pharmacol. 2019;10:212.
  • Danese S, Fiorino G, Peyrin-Biroulet L. Filgotinib in Crohn’s disease: JAK is back. Gastroenterology. 2017 Aug;153(2):603–605.
  • Mould DR. The pharmacokinetics of biologics: A primer. Dig Dis. 2015;33(Suppl 1):61–69.
  • Ganellin R, Jefferis R, Roberts S. Similarities and Differences in the discovery, development and use of biopharmaceuticals versus small molecule drugs. Introduction to biological and small molecule drug research and development: theory and case studies. 1st ed. Amsterdam, Netherlands: Elsevier; 2013. p. 161–203.
  • Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003 Nov;3(11):900–911.
  • Varyani F, Argyriou K, Phillips F, et al. Profile of tofacitinib in the treatment of ulcerative colitis: an evidence-based review of recent data. Drug Des Devel Ther. 2019;13:4091–4105.
  • Danese S, Grisham M, Hodge J, et al. JAK inhibition using tofacitinib for inflammatory bowel disease treatment: a hub for multiple inflammatory cytokines. Am J Physiol Gastrointest Liver Physiol. 2016 Feb 1;310(3):G155–G162.
  • Danese S, Argollo M, Le Berre C, et al. JAK selectivity for inflammatory bowel disease treatment: does it clinically matter? Gut. 2019 Oct;68(10):1893–1899.
  • Ferrante M, Sabino J. Efficacy of anti-JAK inhibitors in ulcerative colitis. J Crohns Colitis. 2019 Dec 27:jjz202. DOI:10.1093/ecco-jcc/jjz202.
  • Sandborn WJ, Su C, Panes J. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017 Aug 3;377(5):496–497.
  • Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017 May 4;376(18):1723–1736.
  • Panes J, Sandborn WJ, Schreiber S, et al. Tofacitinib for induction and maintenance therapy of Crohn’s disease: results of two phase IIb randomised placebo-controlled trials. Gut. 2017 June;66(6):1049–1059.
  • Lamba M, Wang R, Fletcher T, et al. Extended-release once-daily formulation of tofacitinib: evaluation of pharmacokinetics compared with immediate-release tofacitinib and impact of food. J Clin Pharmacol. 2016 Nov;56(11):1362–1371.
  • Klamerus KJ, Alvey C, Li L, et al. Evaluation of the potential interaction between tofacitinib and drugs that undergo renal tubular secretion using metformin, an in vivo marker of renal organic cation transporter 2. Clin Pharmacol Drug Dev. 2014 Nov;3(6):499–507.
  • Lawendy N, Lamba M, Chan G, et al. The effect of mild and moderate hepatic impairment on the pharmacokinetics of tofacitinib, an orally active Janus kinase inhibitor. Clin Pharmacol Drug Dev. 2014 Nov;3(6):421–427.
  • Krishnaswami S, Chow V, Boy M, et al. Pharmacokinetics of tofacitinib, a janus kinase inhibitor, in patients with impaired renal function and end-stage renal disease. J Clin Pharmacol. 2014 Jan;54(1):46–52.
  • EMA EMA. Xeljanz - summary of product characteristics. 2017 [updated 2020 Mar 3; cited 2020 Mar 14]. Available from: https://www.ema.europa.eu/en/documents/product-information/xeljanz-epar-product-information_en.pdf
  • Gupta P, Chow V, Wang R, et al. Evaluation of the effect of fluconazole and ketoconazole on the pharmacokinetics of tofacitinib in healthy adult subjects. Clin Pharmacol Drug Dev. 2014 Jan;3(1):72–77.
  • Bannwarth B, Kostine M, Poursac N. A pharmacokinetic and clinical assessment of tofacitinib for the treatment of rheumatoid arthritis. Expert Opin Drug Metab Toxicol. 2013 June;9(6):753–761.
  • Pfizer I XELJANZ/XELJANZ XR (Tofacitinib) Product Monograph -drug interactions. 2017 [updated Mar 2019; cited 2020 Mar 14]. Available from: https://www.pfizermedicalinformation.ca/en-ca/xeljanz/drug-interactions#
  • Gupta P, Alvey C, Wang R, et al. Lack of effect of tofacitinib (CP-690,550) on the pharmacokinetics of the CYP3A4 substrate midazolam in healthy volunteers: confirmation of in vitro data. Br J Clin Pharmacol. 2012 July;74(1):109–115.
  • Menon S, Riese R, Wang R, et al. Evaluation of the Effect of Tofacitinib on the Pharmacokinetics of Oral Contraceptive Steroids in Healthy Female Volunteers. Clin Pharmacol Drug Dev. 2016 Sep;5(5):336–342.
  • D’Amico F, Fiorino G, Furfaro F, et al. Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials. Expert Opin Investig Drugs. 2018 July;27(7):595–599.
  • Vermeire S, Schreiber S, Petryka R, et al. Clinical remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet. 2017 Jan 21;389(10066):266–275.
  • Galien R, Vayssière B, de Vos S, et al. Analysis of the JAK1 selectivity of GLPG0634 and its main metabolite in different species, healthy volunteers and rheumatoid arthritis patients (Abstract 478). 2013 ACR/ARHP annual meeting; San Diego (CA); 2013.
  • Labetoulle R, Paul S, Roblin X. Filgotinib for the treatment of Crohn’s disease. Expert Opin Investig Drugs. 2018 Mar;27(3):295–300.
  • Namour F, Diderichsen PM, Cox E, et al. Pharmacokinetics and pharmacokinetic/pharmacodynamic modeling of filgotinib (GLPG0634), a selective JAK1 inhibitor, in support of phase IIB dose selection. Clin Pharmacokinet. 2015 Aug;54(8):859–874.
  • Anderson K, Zheng H, Kotecha M, et al. The relative bioavailability and effects of food and acid-reducing agents on filgotinib tablets in healthy subjects. Clin Pharmacol Drug Dev. 2019 July;8(5):585–594.
  • Sharma S, Xin Y, Gao C, et al. Population pharmacokinetics and exposure‐response of filgotinib in patients with moderate to severe Crohn’s disease. (Poster M-54). American conference of pharmacometrics; Bellevue (WA); 2016.
  • Namour F, Vayssière B, Galien R, et al. Filgotinib (GLPG0634), a selective JAK1 inhibitor, shows similar pharmacokinetics and pharmacodynamics profiles in Japanese and Caucasian healthy volunteers (Abstract 2763). 2015 ACR/ARHP Annual Meeting; San Francisco (CA); 2015.
  • Namour F, Fagard L, Van der Aa A, et al. Influence of age and renal impairment on the steady state pharmacokinetics of filgotinib, a selective JAK1 inhibitor. Br J Clin Pharmacol. 2018 Dec;84(12):2779–2789.
  • Anderson K, Zheng H, Medzihradsky O, et al. THU0117 PHARMACOKINETICS AND SHORT-TERM SAFETY OF FILGOTINIB, A SELECTIVE JANUS KINASE 1 INHIBITOR, IN SUBJECTS WITH MODERATE HEPATIC IMPAIRMENT. Ann Rheum Dis. 2019;78(Suppl 2):331.
  • Namour F, Desrivot J, Van der Aa A, et al. Clinical confirmation that the selective JAK1 inhibitor filgotinib (GLPG0634) has a low liability for drug-drug interactions. Drug Metab Lett. 2016;10(1):38–48.
  • Begley R, Anderson K, Watkins TR, et al. Lack of drug-drug interaction between filgotinib, a JAK-1 selective inhibitor, and a representative hormonal contraceptive medication, levonorgestrel/ethinyl estradiol (Abstract 1459). 2017 ACR/ARHP Annual Meeting; San Diego (CA); 2017.
  • Rogler G. JAK efficacy in Crohn’s disease. J Crohns Colitis. 2019;jjz186.doi:10.1093/ecco-jcc/jjz186 .
  • Sandborn WJ, Feagan BG, Panes J, et al. 874h - safety and efficacy of ABT-494 (Upadacitinib), an oral Jak1 inhibitor, as induction therapy in patients with Crohn’s disease: results from celest. Gastroenterology. 2017 [cited 2017 Apr 1];152(5, Supplement 1):S1308–S1309.
  • Sandborn WJ, Ghosh S, Panés J, et al. Efficacy and safety of upadacitinib as an induction therapy for patients with moderately-to-severely active ulcerative colitis: data from the phase 2B study U-ACHIEVE. United European Gastroenterol J. 2018;6(8 Suppl):A1–A134.
  • Mohamed MF, Jungerwirth S, Asatryan A, et al. Assessment of effect of CYP3A inhibition, CYP induction, OATP1B inhibition, and high-fat meal on pharmacokinetics of the JAK1 inhibitor upadacitinib. Br J Clin Pharmacol. 2017 Oct;83(10):2242–2248.
  • Klunder B, Mittapalli RK, Mohamed MF, et al. Population pharmacokinetics of upadacitinib using the immediate-release and extended-release formulations in healthy subjects and subjects with rheumatoid arthritis: analyses of phase I-III clinical trials. Clin Pharmacokinet. 2019 Aug;58(8):1045–1058.
  • Mohamed MF, Camp HS, Jiang P, et al. Pharmacokinetics, safety and tolerability of ABT-494, a novel selective JAK 1 inhibitor, in healthy volunteers and subjects with rheumatoid arthritis. Clin Pharmacokinet. 2016 Dec;55(12):1547–1558.
  • Mohamed MF, Zeng J, Marroum PJ, et al. Pharmacokinetics of upadacitinib with the clinical regimens of the extended-release formulation utilized in rheumatoid arthritis phase 3 trials. Clin Pharmacol Drug Dev. 2019 Feb;8(2):208–216.
  • Mohamed MF, Feng T, Enejosa JV, et al. Effects of upadacitinib coadministration on the pharmacokinetics of sensitive cytochrome P450 probe substrates: a study with the modified cooperstown 5+1 cocktail. J Clin Pharmacol. 2020 Jan;60(1):86–95.
  • Mohamed MF, Trueman S, Feng T, et al. Characterization of the effect of renal impairment on upadacitinib pharmacokinetics. J Clin Pharmacol. 2019 June;59(6):856–862.
  • Mohamed MF, Trueman S, Feng T, et al. The JAK1 inhibitor upadacitinib has no effect on the pharmacokinetics of levonorgestrel and ethinylestradiol: a study in healthy female subjects. J Clin Pharmacol. 2019 Apr;59(4):510–516.
  • Perez-Jeldres T, Tyler CJ, Boyer JD, et al. Cell trafficking interference in inflammatory bowel disease: therapeutic interventions based on basic pathogenesis concepts. Inflamm Bowel Dis. 2019 Jan 10;25(2):270–282.
  • Biswas S, Bryant RV, Travis S. Interfering with leukocyte trafficking in Crohn’s disease. Best Pract Res Clin Gastroenterol. 2019 Feb - Apr;38–39:101617.
  • Karuppuchamy T, Behrens EH, Gonzalez-Cabrera P, et al. Sphingosine-1-phosphate receptor-1 (S1P1) is expressed by lymphocytes, dendritic cells, and endothelium and modulated during inflammatory bowel disease. Mucosal Immunol. 2017 Jan;10(1):162–171.
  • Parigi TL, Roda G, Argollo M, et al. Is there a role for therapeutic sphingolipids in inflammatory bowel disease? Expert Rev Gastroenterol Hepatol. 2020 Jan;14(1):47–54.
  • Scott FL, Clemons B, Brooks J, et al. Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity. Br J Pharmacol. 2016 June;173(11):1778–1792.
  • Tran JQ, Hartung JP, Peach RJ, et al. Results from the first-in-human study with ozanimod, a novel, selective sphingosine-1-phosphate receptor modulator. J Clin Pharmacol. 2017 Aug;57(8):988–996.
  • Sandborn WJ, Feagan BG, Wolf DC, et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N Engl J Med. 2016 May 5;374(18):1754–1762.
  • Feagan BG, Sandborn WJ, Danese S, et al. Endoscopic and clinical efficacy demonstrated with oral ozanimod in moderately to severely active Crohn’s disease: 669. Am J Gastroenterol. 2017;112:S371.
  • Feagan BG, D’Haens G, Usiskin K, et al. P661 Early histological improvement demonstrated with oral ozanimod in patients with moderately to severely active Crohn’s disease in the STEPSTONE trial. J Crohn’s Colitis. 2019;13(Supplement_1):S450–S450.
  • Juif PE, Kraehenbuehl S, Dingemanse J. Clinical pharmacology, efficacy, and safety aspects of sphingosine-1-phosphate receptor modulators. Expert Opin Drug Metab Toxicol. 2016 Aug;12(8):879–895.
  • Tran JQ, Hartung JP, Tompkins CA, et al. Effects of high- and low-fat meals on the pharmacokinetics of ozanimod, a novel sphingosine-1-phosphate receptor modulator. Clin Pharmacol Drug Dev. 2018 Aug;7(6):634–640.
  • Tran JQ, Hartung JP, Olson AD, et al. Cardiac safety of ozanimod, a novel sphingosine-1-phosphate receptor modulator: results of a thorough QT/QTc study. Clin Pharmacol Drug Dev. 2018 Mar;7(3):263–276.
  • Al-Shamma H, Lehmann-Bruinsma K, Carroll C, et al. The selective sphingosine 1-phosphate receptor modulator etrasimod regulates lymphocyte trafficking and alleviates experimental colitis. J Pharmacol Exp Ther. 2019 June;369(3):311–317.
  • Sandborn WJ, Peyrin-Biroulet L, Zhang J, et al. Efficacy and safety of etrasimod in a phase 2 randomized trial of patients with ulcerative colitis. Gastroenterology. 2020 Feb;158(3):550–561.
  • NHI NLoM. Etrasimod versus placebo as induction therapy in moderately to severely active ulcerative colitis (ELEVATE UC 12). NCT03996369. [cited 2020 Mar 14]. Available from: https://clinicaltrials.gov/ct2/show/NCT03996369
  • NHI NLoM. Etrasimod versus placebo for the treatment of moderately to severely active ulcerative colitis (ELEVATE UC 52). NCT03945188. [cited 2020 Mar 14]. Available from: https://clinicaltrials.gov/ct2/show/NCT03945188
  • NHI NLoM. An extension study for treatment of moderately to severely active ulcerative colitis (ELEVATE UC OLE). NCT03950232. [cited 2020 Mar 14]. Available from: https://clinicaltrials.gov/ct2/show/NCT03950232
  • Lee CA, Acevedo L, Oh DA, et al. P396 Pharmacokinetics and circulating total lymphocyte count pharmacodynamic response from single and multiple oral doses of etrasimod in Japanese and Caucasian healthy male subjects. J Crohn’s Colitis. 2020;14(Supplement_1):S368–S368.
  • Oh DA, Lee CA, Tang Y, et al. Steady-state trough concentrations and their relationship to selected demographic and clinical response measures in etrasimod-treated patients with moderately-to-severely active ulcerative colitis. World congress of gastroenterology; Istanbul (Turkey); 2019.
  • Schreiber S, Morgan M, Christopher R, et al. Etrasimod (APD334), a potent, selective, oral S1P receptor modulator with preclinical autoimmune disease-modifying activity exhibits favorable PK/PD properties in healthy volunteers. (P180). Advances in inflammatory bowel diseases annual meeting; Orlando (FL); 2016.
  • Peyrin-Biroulet L, Morgan M, Christopher R, et al. P-179 Safety, pharmacokinetics and pharmacodynamics of etrasimod (APD334), an oral selective S1P receptor modulator, after dose-escalation, in healthy volunteers. Inflamm Bowel Dis. 2017;23(suppl_1):S60–S61.
  • Peyrin-Biroulet L, Morgan M, Christopher R, et al. Safety, pharmacokinetics and pharmacodynamics of etrasimod (APD334), an oral selective S1P receptor modulator, after single dose-escalation, in healthy volunteers. 2017 [cited 2020 May 27]. Poster. Available from: http://www.arenapharm.com/wp-content/uploads/2016/12/407C1DC3.pdf
  • Lee CA, Oh DA, Tang Y, et al. Mass Balance, metabolic disposition, and pharmacokinetics of [14 C] etrasimod following oral administration to healthy male volunteers. AAPS 2019 PHarmSCi 360; San Antonio (TX); 2019.
  • Lee CA, Oh DA, Tang Y, et al. PII-111 - disposition of [14C]etrasimod following oral administration to healthy male volunteers and role of cyp2c in formation of oxidative metabolites. ASCPT 2020 annual meeting; Huston (TX); 2020.
  • Sun J, Shen X, Dong J, et al. Laquinimod ameliorates spontaneous colitis in interleukin-10-gene-deficient mice with improved barrier function. Int Immunopharmacol. 2015 Dec;29(2):423–432.
  • D’Haens G, Sandborn WJ, Colombel JF, et al. A phase II study of laquinimod in Crohn’s disease. Gut. 2015 Aug;64(8):1227–1235.
  • D’Haens G, Sandborn WJ, Rutgeerts P, et al. P386 pharmacokinetics of laquinimod in patients with active moderate to severe Crohn’s disease. J Crohn’s Colitis. 2014;8(Supplement_1):S226–S226.
  • Thone J, Linker RA. Laquinimod in the treatment of multiple sclerosis: a review of the data so far. Drug Des Devel Ther. 2016;10:1111–1118.
  • Ziemssen T, Tumani H, Sehr T, et al. Safety and in vivo immune assessment of escalating doses of oral laquinimod in patients with RRMS. J Neuroinflammation. 2017 Aug 31;14(1):172.
  • Fernandez O. Oral laquinimod treatment in multiple sclerosis. Neurologia. 2011 Mar;26(2):111–117.
  • Tuvesson H, Hallin I, Persson R, et al. Cytochrome P450 3A4 is the major enzyme responsible for the metabolism of laquinimod, a novel immunomodulator. Drug Metab Dispos. 2005 June;33(6):866–872.
  • Elgart A, Zur AA, Mimrod D, et al. The effect of laquinimod, a novel immuno-modulator in development to treat Huntington disease, on the pharmacokinetics of ethinylestradiol and levonorgestrel in healthy young women. Eur J Clin Pharmacol. 2019 Jan;75(1):41–49.
  • EMA EMA. Refusal of the marketing authorisation for Nerventra (Laquinimod). Outcome of re-examination 2014. [updated 2014 Sep 1; cited 2020 Mar 14]. Available from: https://www.ema.europa.eu/en/documents/assessment-report/nerventra-epar-refusal-public-assessment-report_en.pdf

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.