Population pharmacokinetics-pharmacodynamics of vedolizumab in patients with ulcerative colitis and Crohn's disease

M Rosario, N L Dirks, M R Gastonguay, A A Fasanmade, T Wyant, A Parikh, W J Sandborn, B G Feagan, W Reinisch, I Fox, M Rosario, N L Dirks, M R Gastonguay, A A Fasanmade, T Wyant, A Parikh, W J Sandborn, B G Feagan, W Reinisch, I Fox

Abstract

Background: Vedolizumab, an anti-α(4)β(7) integrin monoclonal antibody (mAb), is indicated for treating patients with moderately to severely active ulcerative colitis (UC) and Crohn's disease (CD). As higher therapeutic mAb concentrations have been associated with greater efficacy in inflammatory bowel disease, understanding determinants of vedolizumab clearance may help to optimise dosing.

Aims: To characterise vedolizumab pharmacokinetics in patients with UC and CD, to identify clinically relevant determinants of vedolizumab clearance, and to describe the pharmacokinetic-pharmacodynamic relationship using population modelling.

Methods: Data from a phase 1 healthy volunteer study, a phase 2 UC study, and 3 phase 3 UC/CD studies were included. Population pharmacokinetic analysis for repeated measures was conducted using nonlinear mixed effects modelling. Results from the base model, developed using extensive phase 1 and 2 data, were used to develop the full covariate model, which was fit to sparse phase 3 data.

Results: Vedolizumab pharmacokinetics was described by a 2-compartment model with parallel linear and nonlinear elimination. Using reference covariate values, linear elimination half-life of vedolizumab was 25.5 days; linear clearance (CL(L)) was 0.159 L/day for UC and 0.155 L/day for CD; central compartment volume of distribution (V(c)) was 3.19 L; and peripheral compartment volume of distribution was 1.66 L. Interindividual variabilities (%CV) were 35% for CLL and 19% for V(c); residual variance was 24%. Only extreme albumin and body weight values were identified as potential clinically important predictors of CL(L).

Conclusions: Population pharmacokinetic parameters were similar in patients with moderately to severely active UC and CD. This analysis supports use of vedolizumab fixed dosing in these patients. Clinicaltrials.gov Identifiers: NCT01177228; NCT00783718 (GEMINI 1); NCT00783692 (GEMINI 2); NCT01224171 (GEMINI 3).

© 2015 Takeda Pharmaceuticals International Co published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Median (interquartile range) of observed vedolizumab trough serum concentration vs. nominal sampling time in patients with UC (GEMINI 1) and patients with CD (GEMINI 2) during maintenance treatment with placebo or vedolizumab 300 mg every 4 weeks (Q4W) or every 8 weeks (Q8W). All patients (including those in the placebo group) received 2 doses of vedolizumab 300 mg during induction (at weeks 0 and 2). The median value is shown as a point and the interquartile range is represented by a vertical bar.
Figure 2
Figure 2
Diagrammatic representation of the population pharmacokinetic model of vedolizumab. Conc, vedolizumab concentration; Km, concentration at half‐maximum elimination rate; Vmax, maximum elimination rate.
Figure 3
Figure 3
Distribution of individual vedolizumab linear clearance (CLL) estimates from the final population pharmacokinetic model in patients with UC and patients with CD.
Figure 4
Figure 4
Goodness‐of‐fit plots: observed vedolizumab serum concentration vs. population and individual predicted vedolizumab concentration from the final population pharmacokinetic model. Values are shown as points with a dashed pink loess trend line through the data. A line of identity (solid black) is shown for reference.
Figure 5
Figure 5
Effect of covariates on vedolizumab linear clearance (CLL). Each point and line represent the median and 95% credible interval, respectively, of the Bayesian posterior distribution of normalised samples of vedolizumab CLL adjusted for the covariate. The reference individual weighs 70 kg; is 40 years old; has an albumin level of 4 g/dL, a faecal calprotectin level of 700 mg/kg, a CDAI score of 300 (for patient with CD), a partial Mayo score of 6 (for patient with UC), and no concomitant therapy use; and is ADA negative and TNF‐α antagonist therapy naïve. Albumin: 2.7, 3.2, 3.7, 4.2 and 4.7 g/dL represent the 6th, 18th, 70th, 85th, and 98.5th percentiles, respectively, of baseline albumin levels for patients in GEMINI 1, 2, and 3. Weight: 40, 60, 80, 100, and 120 kg represent the 1.5th, 30th, 71st, 92nd and 98th percentiles, respectively, of baseline weight values for patients in GEMINI 1, 2, and 3. The vertical black line is drawn at the reference point estimate, and the shaded region is ± 25% of the reference point estimate chosen to represent an uncertainty range of clinical unimportance.
Figure 6
Figure 6
Individual vedolizumab linear clearance (CLL) estimates by Mayo endoscopic subscore at the end of induction (week 6) for patients with UC (GEMINI 1). Midlines represent medians. Box limits represent 25th and 75th percentiles. Whiskers (error bars) represent highest and lowest points within 1.5 × interquartile range. Individual points represent outliers.
Figure 7
Figure 7
Receptor (α4β7) saturation plot: observed MAdCAM‐1 vs. observed vedolizumab serum concentration in patients with UC (GEMINI 1) and patients with CD (GEMINI 2). Baseline data are included.
Figure 8
Figure 8
Observed MAdCAM‐1 vs. time for patients with UC (GEMINI 1) and patients with CD (GEMINI 2). Upper left panel: patients received placebo during induction and maintenance; upper right panel: patients received 2 doses of vedolizumab 300 mg during induction and vedolizumab 300 mg every 4 weeks (Q4W) during maintenance; lower left panel: patients received 2 doses of vedolizumab 300 mg during induction and vedolizumab 300 mg every 8 weeks (Q8W) during maintenance; lower right panel: patients received 2 doses of vedolizumab 300 mg during induction and placebo during maintenance.

References

    1. Soler D, Chapman T, Yang LL, Wyant T, Egan R, Fedyk ER. The binding specificity and selective antagonism of vedolizumab, an anti‐alpha4beta7 integrin therapeutic antibody in development for inflammatory bowel diseases. J Pharmacol Exp Ther 2009; 330: 864–75.
    1. Paul S, Moreau AC, Del Tedesco E, et al Pharmacokinetics of adalimumab in inflammatory bowel diseases: a systematic review and meta‐analysis. Inflamm Bowel Dis 2014; 20: 1288–95.
    1. Ordas I, Mould DR, Feagan BG, Sandborn WJ. Anti‐TNF Monoclonal Antibodies in Inflammatory Bowel Disease: Pharmacokinetics‐Based Dosing Paradigms. Clin Pharmacol Ther 2012; 91: 635–46.
    1. Colombel JF, Feagan BG, Sandborn WJ, Van Assche G, Robinson AM. Therapeutic drug monitoring of biologics for inflammatory bowel disease. Inflamm Bowel Dis 2012a; 18: 349–58.
    1. Feagan BG, Greenberg GR, Wild G, et al Treatment of active Crohn's disease with MLN0002, a humanized antibody to the alpha4beta7 integrin. Clin Gastroenterol Hepatol 2008; 6: 1370–7.
    1. Feagan BG, Greenberg GR, Wild G, et al Treatment of ulcerative colitis with a humanized antibody to the alpha4beta7 integrin. N Engl J Med 2005; 352: 2499–507.
    1. Parikh A, Leach T, Wyant T, et al Vedolizumab for the treatment of active ulcerative colitis: a randomized controlled phase 2 dose‐ranging study. Inflamm Bowel Dis 2012; 18: 1470–9.
    1. Feagan BG, Rutgeerts P, Sands BE, et al Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013; 369: 699–710.
    1. Sandborn WJ, Feagan BG, Rutgeerts P, et al Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013; 369: 711–21.
    1. Sands BE, Feagan BG, Rutgeerts P, et al Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology 2014; 147: 618–27.
    1. Rosario M, French J, Dirks N, Milton A, Fox I, Gastonguay M, eds. Exposure‐response relationship during vedolizumab induction therapy in adults with ulcerative colitis. 9th Congress of the European Crohn's and Colitis Organisation; 20–22 February 2014; Copenhagen, Denmark: J Crohns Colitis, 2014a.
    1. Rosario M, French J, Dirks N, Milton A, Fox I, Gastonguay M, eds. Exposure‐response relationship of vedolizumab after 6 weeks of treatment in adults with Crohn's disease. 9th Congress of the European Crohn's and Colitis Organisation; 20–22 February 2014; Copenhagen, Denmark: J Crohns Colitis, 2014b.
    1. Beal SL, Sheiner LB, Boeckmann AJ, Bauer RJ, eds. NONMEM Users Guide Part I‐VIII. Hanover, MD: ICON Development Solutions, 1989–2012.
    1. Gastonguay M, ed. A full model estimation approach for covariate effects: Inference based on clinical importance and estimation precision. American Association of Pharmaceutical Scientists Annual Meeting and Exposition; 7–11 November 2004; Baltimore, MD: AAPS Journal, 2004.
    1. Gibiansky L, Gibiansky E. Target‐mediated drug disposition model: relationships with indirect response models and application to population PK‐PD analysis. J Pharmacokinet Pharmacodyn 2009; 36: 341–51.
    1. Tabrizi MA, Tseng CM, Roskos LK. Elimination mechanisms of therapeutic monoclonal antibodies. Drug Discov Today 2006; 11: 81–8.
    1. Dirks NL, Nolting A, Kovar A, Meibohm B. Population pharmacokinetics of cetuximab in patients with squamous cell carcinoma of the head and neck. J Clin Pharmacol 2008; 48: 267–78.
    1. Frey N, Grange S, Woodworth T. Population pharmacokinetic analysis of tocilizumab in patients with rheumatoid arthritis. J Clin Pharmacol 2010; 50: 754–66.
    1. Ng CM, Joshi A, Dedrick RL, Garovoy MR, Bauer RJ. Pharmacokinetic‐pharmacodynamic‐efficacy analysis of efalizumab in patients with moderate to severe psoriasis. Pharm Res 2005; 22: 1088–100.
    1. Xu Z, Seitz K, Fasanmade A, et al Population pharmacokinetics of infliximab in patients with ankylosing spondylitis. J Clin Pharmacol 2008; 48: 681–95.
    1. Fasanmade AA, Adedokun OJ, Ford J, et al Population pharmacokinetic analysis of infliximab in patients with ulcerative colitis. Eur J Clin Pharmacol 2009; 65: 1211–28.
    1. Zhou H, Jang H, Fleischmann RM, et al Pharmacokinetics and safety of golimumab, a fully human anti‐TNF‐alpha monoclonal antibody, in subjects with rheumatoid arthritis. J Clin Pharmacol 2007; 47: 383–96.
    1. Colombel JF, Feagan B, Sandborn W, Assche Gv, Robinson A. Therapeutic drug monitoring of biologics for inflammatory bowel disease. Inflamm Bowel Dis 2012; 18: 349–58.
    1. Nestorov I. Clinical pharmacokinetics of TNF antagonists: how do they differ? Semin Arthritis Rheum 2005; 5(Suppl. 1): 12–8.
    1. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet 2010; 49: 633–59.
    1. Fasanmade AA, Adedokun OJ, Olson A, Strauss R, Davis HM. Serum albumin concentration: a predictive factor of infliximab pharmacokinetics and clinical response in patients with ulcerative colitis. Int J Clin Pharmacol Ther 2010; 48: 297–308.
    1. Brandse JF, van der Kleij D, Wolbink GJ, et al The pharmacokinetics of infliximab induction therapy in patients with moderate to severe ulcerative colitis. Gastroenterology 2014; 146(5 Suppl. 1): S134.
    1. Van Moerkercke W, Ackaert C, Compernolle G, et al High infliximab levels are associated with mucosal healing in Crohn's disease. Gastroenterology 2010; 138(Suppl. 1): S60.
    1. Roblin X, Marotte H, Rinaudo M, et al Association between pharmacokinetics of adalimumab and mucosal healing in patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol 2014; 12: 80–4.
    1. Colombel JF, Sandborn WJ, Allez M, et al Association between plasma concentrations of certolizumab pegol and endoscopic outcomes of patients with Crohn's disease. Clin Gastroenterol Hepatol 2014; 12: 423–31.
    1. Wade JR, Parker G, Kosutic G, et al Population pharmacokinetic analysis of certolizumab pegol in patients with Crohn's disease. J Clin Pharmacol 2015. [Epub ahead of print].
    1. Velagapudi RB, Noertersheuser P, Awni W, et al , eds. Effect of methotrexate (MTX) coadministration on the pharmacokinetics (PK) of adalimumab (HUMIRA (TM), Abbott) following a single intravenous (iv) injection. American College of Rheumatology 67th Annual Meeting; 23–28 October 2003; Orlando, FL: Arthritis Rheum, 2003.
    1. Chow A, Earp J, Gupta M, et al Utility of population pharmacokinetic modeling in the assessment of therapeutic protein‐drug interactions. J Clin Pharmacol 2014; 54: 593–601.
    1. Earp J, Fang L, Ma L, et al Assessing labeling claims for drug interactions using a population PK approach: Vedolizumab. J Pharmacokinet Pharmacodyn 2014; 41: S39.

Source: PubMed

3
Se inscrever