A randomized trial of obeticholic acid monotherapy in patients with primary biliary cholangitis

Kris V Kowdley, Velimir Luketic, Roger Chapman, Gideon M Hirschfield, Raoul Poupon, Christoph Schramm, Catherine Vincent, Christian Rust, Albert Parés, Andrew Mason, Hanns-Ulrich Marschall, David Shapiro, Luciano Adorini, Cathi Sciacca, Tessa Beecher-Jones, Olaf Böhm, Richard Pencek, David Jones, Obeticholic Acid PBC Monotherapy Study Group, Kris V Kowdley, Velimir Luketic, Roger Chapman, Gideon M Hirschfield, Raoul Poupon, Christoph Schramm, Catherine Vincent, Christian Rust, Albert Parés, Andrew Mason, Hanns-Ulrich Marschall, David Shapiro, Luciano Adorini, Cathi Sciacca, Tessa Beecher-Jones, Olaf Böhm, Richard Pencek, David Jones, Obeticholic Acid PBC Monotherapy Study Group

Abstract

Obeticholic acid (OCA), a potent farnesoid X receptor agonist, was studied as monotherapy in an international, randomized, double-blind, placebo-controlled phase 2 study in patients with primary biliary cholangitis who were then followed for up to 6 years. The goals of the study were to assess the benefit of OCA in the absence of ursodeoxycholic acid, which is relevant for patients who are intolerant of ursodeoxycholic acid and at higher risk of disease progression. Patients were randomized and dosed with placebo (n = 23), OCA 10 mg (n = 20), or OCA 50 mg (n = 16) given as monotherapy once daily for 3 months (1 randomized patient withdrew prior to dosing). The primary endpoint was the percent change in alkaline phosphatase from baseline to the end of the double-blind phase of the study. Secondary and exploratory endpoints included change from baseline to month 3/early termination in markers of cholestasis, hepatocellular injury, and farnesoid X receptor activation. Efficacy and safety continue to be monitored through an ongoing 6-year open-label extension (N = 28). Alkaline phosphatase was reduced in both OCA groups (median% [Q1, Q3], OCA 10 mg -53.9% [-62.5, -29.3], OCA 50 mg -37.2% [-54.8, -24.6]) compared to placebo (-0.8% [-6.4, 8.7]; P < 0.0001) at the end of the study, with similar reductions observed through 6 years of open-label extension treatment. OCA improved many secondary and exploratory endpoints (including γ-glutamyl transpeptidase, alanine aminotransferase, conjugated bilirubin, and immunoglobulin M). Pruritus was the most common adverse event; 15% (OCA 10 mg) and 38% (OCA 50 mg) discontinued due to pruritus.

Conclusion: OCA monotherapy significantly improved alkaline phosphatase and other biochemical markers predictive of improved long-term clinical outcomes. Pruritus increased dose-dependently with OCA treatment. Biochemical improvements were observed through 6 years of open-label extension treatment. (Hepatology 2018;67:1890-1902).

© 2017 The Authors. Hepatology published by Wiley Periodicals, Inc. on behalf of American Association for the Study of Liver Diseases.

Figures

Figure 1
Figure 1
OCA reduces ALP. (A) Percent change in ALP at EOS. (B) ALP change from baseline. Data are median (Q1, Q3). **P < 0.01, ***P < 0.0001; P values determined by pairwise comparisons using a two‐sided Wilcoxon‐Mann‐Whitney test comparing changes in each OCA group with the placebo group. Note: the primary endpoint includes ET values or last observation carried forward values. ALP change from baseline values at month 3 include the ET visit data for patients who discontinued where available. Abbreviation: BL, baseline.
Figure 2
Figure 2
Effect of OCA on liver biochemistries. (A) GGT change from baseline. (B) AST change from baseline. (C) ALT change from baseline. (D) Conjugated bilirubin change from baseline. Data are median (Q1, Q3). Change from baseline values at month 3 include the ET visit data for patients who discontinued. *P < 0.05, **P < 0.01, ***P < 0.0001; P values determined by pairwise comparisons using a two‐sided Wilcoxon‐Mann‐Whitney test comparing changes in each OCA group with the placebo group. Abbreviation: BL, baseline.
Figure 3
Figure 3
Markers of FXR activation. (A) FGF‐19 change from baseline to month 3/ET. Baseline FGF‐19 was 77.5 (27.6, 85.5), 109.7 (86.3, 228.3), and 74.7 (48.8, 144.3) for placebo, OCA 10 mg, and OCA 50 mg groups, respectively. (B) C4 change from baseline to month 3/ET. Baseline C4 was 18.2 (8.9, 22.3), 8.6 (4.3, 19.0), and 9.1 (6.5, 27.4) for placebo, OCA 10 mg and OCA 50 mg groups, respectively. Data are median (Q1, Q3). Change from baseline values at month 3 include the ET visit data for patients who discontinued. *P < 0.05; P values determined by pairwise comparisons using a two‐sided Wilcoxon‐Mann‐Whitney test comparing changes in each OCA group with the placebo group.
Figure 4
Figure 4
Changes in liver biochemistry over 6 years of OLE. (A) ALP change from baseline. (B) GGT change from baseline. (C) AST change from baseline. (D) ALT change from baseline. (E) Conjugated bilirubin change from baseline. For all plots, values presented are median (Q1, Q3) of all patients participating in the trial at any point regardless of dose or group. Dashed line represents the median change from baseline at the end of the double‐blind phase. Off‐Rx refers to the study period from month 3 and initiating OCA during the OLE. Abbreviation: BL, baseline.

References

    1. Hirschfield GM, Gershwin ME. The immunobiology and pathophysiology of primary biliary cirrhosis. Annu Rev Pathol 2013;8:303‐330.
    1. Beuers U, Gershwin ME, Gish RG, Invernizzi P, Jones DEJ, Lindor K, et al. Changing nomenclature for PBC: from “cirrhosis” to “cholangitis.” Hepatology 2015;62:1620‐1622.
    1. European Association for the Study of the Liver . EASL clinical practice guidelines: management of cholestatic liver diseases. J Hepatol 2009;51:237‐267.
    1. Lindor KD, Gershwin ME, Poupon R, Kaplan M, Bergasa NV, Heathcote EJ. Primary biliary cirrhosis: AASLD practice guidelines. Hepatology 2009;50:291‐308.
    1. Lammers WJ, Kowdley KV, van Buuren HR. Predicting outcome in primary biliary cirrhosis. Ann Hepatol 2014;13:316‐326.
    1. Corpechot C, Chazouilleres O, Poupon R. Early primary biliary cirrhosis: biochemical response to treatment and prediction of long‐term outcome. J Hepatol 2011;55:1361‐1367.
    1. Lammers WJ, van Buuren HR, Hirschfield GM, Janssen HL, Invernizzi P, Mason AL, et al. Levels of alkaline phosphatase and bilirubin are surrogate end points of outcomes of patients with primary biliary cirrhosis: an international follow‐up study. Gastroenterology 2014;147:1338‐1349.
    1. Hirschfield GM, Mason A, Luketic V, Lindor K, Gordon S, Mayo M, et al. Efficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acid. Gastroenterology 2015;148:751‐761.
    1. Hempfling W, Dilger K, Beuers U. Systematic review: ursodeoxycholic acid—adverse effects and drug interactions. Aliment Pharmacol Ther 2003;18:963‐972.
    1. Pellicciari R, Fiorucci S, Camaioni E, Clerici C, Costantino G, Maloney PR, et al. 6Alpha‐ethyl‐chenodeoxycholic acid (6‐ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. J Med Chem 2002;45:3569‐3572.
    1. Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev 2009;89:147‐191.
    1. Kliewer SA, Mangelsdorf DJ. Bile acids as hormones: the FXR‐FGF15/19 pathway. Dig Dis 2015;33:327‐331.
    1. Beuers U, Trauner M, Jansen P, Poupon R. New paradigms in the treatment of hepatic cholestasis: from UDCA to FXR, PXR and beyond. J Hepatol 2015;62(Suppl.):S25‐S37.
    1. Fiorucci S, Antonelli E, Rizzo G, Renga B, Mencarelli A, Riccardi L, et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology 2004;127:1497‐1512.
    1. Nevens F, Andreone P, Mazzella G, Strasser SI, Bowlus C, Invernizzi P, et al. A placebo‐controlled trial of obeticholic acid in primary biliary cholangitis. N Engl J Med 2016;375:631‐643.
    1. Carbone M, Sharp SJ, Flack S, Paximadas D, Spiess K, Adgey C, et al. The UK‐PBC risk scores: derivation and validation of a scoring system for long‐term prediction of end‐stage liver disease in primary biliary cirrhosis. Hepatology 2015;63:930‐950.
    1. Lammers WJ, Hirschfield GM, Corpechot C, Nevens F, Lindor KD, Janssen HL, et al. Development and validation of a scoring system to predict outcomes of patients with primary biliary cirrhosis receiving ursodeoxycholic acid therapy. Gastroenterology 2015;149:1804‐1812.
    1. Lubsen J, Kirwan BA. Combined endpoints: can we use them? Stat Med 2002;21:2959‐2970.
    1. Angulo P, Dickson ER, Therneau TM, Jorgensen RA, Smith C, DeSotel CK, et al. Comparison of three doses of ursodeoxycholic acid in the treatment of primary biliary cirrhosis: a randomized trial. J Hepatol 1999;30:830‐835.
    1. Poupon RE, Balkau B, Eschwege E, Poupon R. A multicenter, controlled trial of ursodiol for the treatment of primary biliary cirrhosis. UDCA‐PBC Study Group. N Engl J Med 1991;324:1548‐1554.
    1. Lindor KD, Dickson ER, Baldus WP, Jorgensen RA, Ludwig J, Murtaugh PA, et al. Ursodeoxycholic acid in the treatment of primary biliary cirrhosis. Gastroenterology 1994;106:1284‐1290.
    1. Heathcote EJ, Cauch‐Dudek K, Walker V, Bailey RJ, Blendis LM, Ghent CN, et al. The Canadian Multicenter Double‐blind Randomized Controlled Trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology 1994;19:1149‐1156.
    1. Pares A, Caballeria L, Rodes J, Bruguera M, Rodrigo L, Garcia‐Plaza A, et al. Long‐term effects of ursodeoxycholic acid in primary biliary cirrhosis: results of a double‐blind controlled multicentric trial. UDCA‐Cooperative Group from the Spanish Association for the Study of the Liver. J Hepatol 2000;32:561‐566.
    1. Marschall HU, Wagner M, Zollner G, Fickert P, Silbert D, Gustafsson U, et al. Combined rifampicin and ursodeoxycholic acid treatment does not amplify rifampicin effects on hepatic detoxification and transport systems in humans. Digestion 2012;86:244‐249.
    1. Liu H, Norman GL, Shums Z, Worman HJ, Krawitt EL, Bizzaro N, et al. PBC screen: an IgG/IgA dual isotype ELISA detecting multiple mitochondrial and nuclear autoantibodies specific for primary biliary cirrhosis. J Autoimmun 2010;35:436‐442.
    1. Webb GJ, Siminovitch KA, Hirschfield GM. The immunogenetics of primary biliary cirrhosis: a comprehensive review. J Autoimmun 2015;64:42‐52.
    1. Selmi C, Meroni PL, Gershwin ME. Primary biliary cirrhosis and Sjogren's syndrome: autoimmune epithelitis. J Autoimmun 2012;39:34‐42.
    1. Poupon R. Liver alkaline phosphatase: a missing link between choleresis and biliary inflammation. Hepatology 2015;61:2080‐2090.
    1. Talwalkar JA, Souto E, Jorgensen RA, Lindor KD. Natural history of pruritus in primary biliary cirrhosis. Clin Gastroenterol Hepatol 2003;1:297‐302.
    1. Carey EJ, Ali AH, Lindor KD. Primary biliary cirrhosis. Lancet 2015;386:1565‐1575.
    1. Sorokin A, Brown JL, Thompson PD. Primary biliary cirrhosis, hyperlipidemia, and atherosclerotic risk: a systematic review. Atherosclerosis 2007;194:293‐299.
    1. Chao F, Gong W, Zheng Y, Li Y, Huang G, Gao M, et al. Upregulation of scavenger receptor class B type I expression by activation of FXR in hepatocyte. Atherosclerosis 2010;213:443‐448.

Source: PubMed

3
Prenumerera