Modeling and Experimental Studies of Obeticholic Acid Exposure and the Impact of Cirrhosis Stage

J E Edwards, C LaCerte, T Peyret, N H Gosselin, J F Marier, A F Hofmann, D Shapiro, J E Edwards, C LaCerte, T Peyret, N H Gosselin, J F Marier, A F Hofmann, D Shapiro

Abstract

Obeticholic acid (OCA), a semisynthetic bile acid, is a selective and potent farnesoid X receptor (FXR) agonist in development for the treatment of chronic nonviral liver diseases. Physiologic pharmacokinetic models have been previously used to describe the absorption, distribution, metabolism, and excretion (ADME) of bile acids. OCA plasma levels were measured in healthy volunteers and cirrhotic subjects. A physiologic pharmacokinetic model was developed to quantitatively describe the ADME of OCA in patients with and without hepatic impairment. There was good agreement between predicted and observed increases in systemic OCA exposure in subjects with mild, moderate, and severe hepatic impairment, which were 1.4-, 8-, and 13-fold relative to healthy volunteers. Predicted liver exposure for subjects with mild, moderate, and severe hepatic impairment were increased only 1.1-, 1.5-, and 1.7-fold. In subjects with cirrhosis, OCA exposure in the liver, the primary site of pharmacological activity along with the intestine, is increased marginally (∼2-fold).

Trial registration: ClinicalTrials.gov NCT01904539 NCT01933503.

© 2016 The Authors. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Physiologic Pharmacokinetic Model Diagram. Similar to other bile acids, OCA is conjugated to glycine and taurine. OCA and its conjugates undergo enterohepatic recirculation. Accounting for differences in hepatic impairment is done by modifying the different flow rates “f,” transport rates “t,” and biotransformation rates “b” at different points throughout the model.
Figure 2
Figure 2
Visual Predictive Check of the Physiologic Pharmacokinetic Model in Healthy Volunteers With Normal Hepatic Function. Blue circles represent observed data points. Solid red line represents median of observed data. Dashed red lines represent the 5th and 95th percentiles of observed data. Solid black line represents predicted median. Gray band represents the 90% prediction interval.
Figure 3
Figure 3
Visual Predictive Check of the Physiologic Pharmacokinetic Model in Subjects With Normal and Impaired Hepatic Function. Blue circles represent observed data points. Solid red line represents median of observed data. Dashed red lines represent the 5th and 95th percentiles of observed data. Solid black line represents predicted median. Gray band represents the 90% prediction interval.
Figure 4
Figure 4
External Validation of the Physiologic Pharmacokinetic Model. (a) Healthy volunteers with normal hepatic function. (b) Subjects with impaired hepatic function. Blue circles represent observed data points. Solid red line represents median of observed data. Dashed red lines represent the 5th and 95th percentiles of observed data. Solid black line represents predicted median. Gray band represents the 90% prediction interval.
Figure 5
Figure 5
Plasma OCA Concentrations are a Poor Surrogate for Liver OCA Concentrations. Systemic exposure of OCA was predicted to be 1.4‐, 8.0‐, and 13‐fold greater in subjects with mild, moderate, and severe hepatic impairment, respectively, based on Child‐Pugh score, than in healthy volunteers, which is consistent with the observed results. Liver exposure of OCA was predicted to be 1.1‐, 1.5‐, and 1.7‐fold greater in subjects with mild, moderate, and severe hepatic impairment, respectively, than in healthy volunteers. n = 8 for observed values. Predicted values were estimated using a 200 replicate simulation from the physiologic pharmacokinetic model of OCA (n = 1,600). Boxplot whiskers represent 1st to 99th percentile.

References

    1. Hirschfield, G.M. et al Efficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acid. Gastroenterology 148, 751–761 (2015). English.
    1. Walters, J.R. , Johnston, I.M. , Nolan, J.D. , Vassie, C. , Pruzanski, M.E. & Shapiro, D.A. The response of patients with bile acid diarrhoea to the farnesoid X receptor agonist obeticholic acid. Aliment. Pharmacol. Ther. 41, 54–64 (2015). PubMed PMID: 25329562.
    1. Mudaliar, S. et al Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 145, 574–582 e1 (2013). PubMed PMID: 23727264.
    1. Neuschwander‐Tetri, B.A. et al Farnesoid X nuclear receptor ligand obeticholic acid for non‐cirrhotic, non‐alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo‐controlled trial. Lancet 385, 956–965 (2015).
    1. Pellicciari, R. et al 6alpha‐ethyl‐chenodeoxycholic acid (6‐ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. J. Med. Chem. 45, 3569–3572 (2002). PubMed PMID: 12166927. Epub 2002/08/09. eng.
    1. Schuppan, D. , Schattenberg, J.M. Non‐alcoholic steatohepatitis: pathogenesis and novel therapeutic approaches. J. Gastroenterol. Hepatol. 28 (Suppl 1), 68–76 (2013). PubMed PMID: 23855299.
    1. Fischer, S. , Beuers, U. , Spengler, U. , Zwiebel, F.M. , Koebe, H.G. Hepatic levels of bile acids in end‐stage chronic cholestatic liver disease. Clin. Chim. Acta 251, 173–186 (1996). PubMed PMID: 8862472.
    1. Ostrow, J. Metabolism of bile salts in cholestasis in humans In Hepatic transport and bile secretion. (eds. Tavoloni N., Berk P. 673–712 (Raven Press, New York, 1993).
    1. Martell, M. , Coll, M. , Ezkurdia, N. , Raurell, I. , Genesca, J. Physiopathology of splanchnic vasodilation in portal hypertension. World J. Hepatol. 2, 208–220 (2010). PubMed PMID: 21160999. Pubmed Central PMCID: 2999290.
    1. Ohkubo, H. , Okuda, K. , Iida, S. , Ohnishi, K. , Ikawa, S. , Makino, I. Role of portal and splenic vein shunts and impaired hepatic extraction in the elevated serum bile acids in liver cirrhosis. Gastroenterology 86, 514–520 (1984).
    1. Johnson, T.N. , Boussery, K. , Rowland‐Yeo, K. , Tucker, G.T. , Rostami‐Hodjegan, A. A semi‐mechanistic model to predict the effects of liver cirrhosis on drug clearance. Clin. Pharmacokinet. 49, 189–206 (2010). PubMed PMID: 20170207.
    1. Eipel, C. , Abshagen, K. , Vollmar, B. Regulation of hepatic blood flow: the hepatic arterial buffer response revisited. World J. Gastroenterol. WJG 16, 6046–6057 (2010). PubMed PMID: 21182219. Pubmed Central PMCID: 3012579.
    1. Garbutt, J.T. , Lack, L. , Tyor, M.P. Physiological basis of alterations in the relative conjugation of bile acids with glycine and taurine. Am. J. Clin. Nutr. 24, 218–228 (1971). PubMed PMID: 5545849.
    1. Harada, T. Studies on biliary bile acid metabolism in hepato‐biliary diseases. Gastroenterol. Jap. 10, 174–178 (1975). PubMed PMID: 1234096.
    1. Turnberg, L.A. , Grahame, G. Bile salt secretion in cirrhosis of the liver. Gut 11, 126–133 (1970). PubMed PMID: 5441881. Pubmed Central PMCID: 1411355.
    1. Murphy, G.M. , Ross, A. , Billing, B.H. Serum bile acids in primary biliary cirrhosis. Gut 13, 201–206 (1972). PubMed PMID: 5024725. Pubmed Central PMCID: 1412131.
    1. Neale, G. , Lewis, B. , Weaver, V. , Panveliwalla, D. Serum bile acids in liver disease. Gut 12, 145–152 (1971). PubMed PMID: 5548561. Pubmed Central PMCID: 1411536.
    1. Molino, G. , Hofmann, A.F. , Cravetto, C. , Belforte, G. , Bona, B. Simulation of the metabolism and enterohepatic circulation of endogenous chenodeoxycholic acid in man using a physiological pharmacokinetic model. Eur. J. Clin. Investig. 16, 397–414 (1986). PubMed PMID: 3100308. Epub 1986/10/01. eng.
    1. Hofmann, A.F. , Molino, G. , Milanese, M. , Belforte, G. Description and simulation of a physiological pharmacokinetic model for the metabolism and enterohepatic circulation of bile acids in man. Cholic acid in Healthy Man. J. Clin. Investig. 71, 1003–1022 (1983). PubMed PMID: 6682120. Pubmed Central PMCID: 436958.
    1. Hofmann, A.F. , Cravetto, C. , Molino, G. , Belforte, G. , Bona, B. Simulation of the metabolism and enterohepatic circulation of endogenous deoxycholic acid in humans using a physiologic pharmacokinetic model for bile acid metabolism. Gastroenterology 93, 693–709 (1987). PubMed PMID: 3623017.
    1. Beal, S.L. Ways to fit a PK model with some data below the quantification limit. J. Pharmacokinet. Pharmacodynam. 28, 481–504 (2001). PubMed PMID: 11768292.
    1. Steiner, C. et al Bile acid metabolites in serum: intraindividual variation and associations with coronary heart disease, metabolic syndrome and diabetes mellitus. PLoS One 6, e25006 (2011). PubMed PMID: 22110577. Pubmed Central PMCID: 3215718.
    1. Bathena, S.P. et al Urinary bile acids as biomarkers for liver diseases I. Stability of the baseline profile in healthy subjects. Toxicol. Sci. 143, 296–307 (2015). PubMed PMID: 25344562.
    1. Danielsson, H. , Eneroth, P. , Hellström, K. , Lindstedt, S. , Sjövall, J. On the turnover and excretory products of cholic and chenodeoxycholic acid in man. J. Biol. Chem. 238, 2299–2304 (1963). PubMed PMID: 14024949.
    1. Pedersen, L. , Arnfred, T. Kinetics and pool size of chenodeoxycholic acid in cholesterol gallstone patients. Scand. J. Gastroenterol. 10, 557–560 (1975). PubMed PMID: 1153952.
    1. Kew, M.C. , Varma, R.R. , Santos, H.A.D. , Scheuer, P.J. Portal hypertension in primary biliary cirrhosis. Gut 12, 830–834 (1971). PubMed PMID: PMC1411882.
    1. Duché, M. , Fabre, M. , Kretzschmar, B. , Serinet, M.‐O. , Gauthier, F. , Chardot, C. Prognostic Value of Portal Pressure at the Time of Kasai Operation in Patients With Biliary Atresia. J. Pediatr. Gastroenterol. Nutr. 43, (2006):640‐5. PubMed PMID: 00005176‐200611000‐00017.

Source: PubMed

3
Prenumerera