The Nonsteroidal Farnesoid X Receptor Agonist Cilofexor (GS-9674) Improves Markers of Cholestasis and Liver Injury in Patients With Primary Sclerosing Cholangitis

Michael Trauner, Aliya Gulamhusein, Bilal Hameed, Stephen Caldwell, Mitchell L Shiffman, Charles Landis, Bertus Eksteen, Kosh Agarwal, Andrew Muir, Simon Rushbrook, Xiaomin Lu, Jun Xu, Jen-Chieh Chuang, Andrew N Billin, Georgia Li, Chuhan Chung, G Mani Subramanian, Robert P Myers, Christopher L Bowlus, Kris V Kowdley, Michael Trauner, Aliya Gulamhusein, Bilal Hameed, Stephen Caldwell, Mitchell L Shiffman, Charles Landis, Bertus Eksteen, Kosh Agarwal, Andrew Muir, Simon Rushbrook, Xiaomin Lu, Jun Xu, Jen-Chieh Chuang, Andrew N Billin, Georgia Li, Chuhan Chung, G Mani Subramanian, Robert P Myers, Christopher L Bowlus, Kris V Kowdley

Abstract

Primary sclerosing cholangitis (PSC) represents a major unmet medical need. In a phase II double-blind, placebo-controlled study, we tested the safety and efficacy of cilofexor (formerly GS-9674), a nonsteroidal farnesoid X receptor agonist in patients without cirrhosis with large-duct PSC. Patients were randomized to receive cilofexor 100 mg (n = 22), 30 mg (n = 20), or placebo (n = 10) orally once daily for 12 weeks. All patients had serum alkaline phosphatase (ALP) > 1.67 × upper limit of normal and total bilirubin ≤ 2 mg/dL at baseline. Safety, tolerability, pharmacodynamic effects of cilofexor (serum C4 [7α-hydroxy-4-cholesten-3-one] and bile acids), and changes in liver biochemistry and serum fibrosis markers were evaluated. Overall, 52 patients were randomized (median age 43 years, 58% male, 60% with inflammatory bowel disease, 46% on ursodeoxycholic acid). Baseline median serum ALP and bilirubin were 348 U/L (interquartile range 288-439) and 0.7 mg/dL (0.5-1.0), respectively. Dose-dependent reductions in liver biochemistry were observed. At week 12, cilofexor 100 mg led to significant reductions in serum ALP (median reduction -21%; P = 0.029 versus placebo), gamma-glutamyl transferase (-30%; P < 0.001), alanine aminotransferase (ALT) (-49%; P = 0.009), and aspartate aminotransferase (-42%; P = 0.019). Cilofexor reduced serum C4 compared with placebo; reductions in bile acids were greatest with 100 mg. Relative reductions in ALP were similar between ursodeoxycholic acid-treated and untreated patients. At week 12, cilofexor-treated patients with a 25% or more relative reduction in ALP had greater reductions in serum alanine aminotransferase, aspartate aminotransferase, gamma-glutamyl transferase, tissue inhibitor of metalloproteinase 1, C-reactive protein, and bile acids than nonresponders. Adverse events were similar between cilofexor and placebo-treated patients. Rates of grade 2 or 3 pruritus were 14% with 100 mg, 20% with 30 mg, and 40% with placebo. Conclusion: In this 12-week, randomized, placebo-controlled study, cilofexor was well tolerated and led to significant improvements in liver biochemistries and markers of cholestasis in patients with PSC.

Trial registration: ClinicalTrials.gov NCT02943460.

© 2019 The Authors. Hepatology published by Wiley Periodicals, Inc., on behalf of American Association for the Study of Liver Diseases.

Figures

Figure 1
Figure 1
Cilofexor improves serum ALP in patients with PSC. (A) Median (IQR) serum ALP between baseline and week 12 of the double‐blind phase of the study. (B) Median absolute change in serum ALP from baseline to week 12 of therapy. P values versus placebo are according to Wilcoxon rank‐sum test. (C) Median (IQR) change in serum ALP relative to the ULN between baseline and week 12 of therapy. (D) Median relative (percentage) change in serum ALP from baseline to week 12 of therapy (overall and according to UDCA treatment). P values versus placebo are according to Wilcoxon rank‐sum test.
Figure 2
Figure 2
Effect of cilofexor on liver biochemistry and markers of fibrosis and bile acid homeostasis. (A) Cilofexor 100 mg leads to improvement in serum GGT, ALT, AST, and TIMP‐1 compared with placebo. P values for cilofexor 100 mg versus placebo are according to Wilcoxon rank‐sum test. (B) Cilofexor leads to reductions in C4, total bile acids, primary bile acids, and secondary bile acids compared with placebo. Total bile acids by enzymatic assay and bile acid species by LC/MS‐MS. UDCA and its conjugates have been removed from secondary bile acids. P values for cilofexor 100 mg versus placebo are according to Wilcoxon rank‐sum test. Abbreviations: BA, bile acid; HA, hyaluronic acid.
Figure 3
Figure 3
Cilofexor 100 mg lowers serum bile acids, but does not alter bile acid composition. (A) Absolute levels of serum bile acid species before and after 12 weeks of treatment with cilofexor 100 mg daily. UDCA and its conjugates have been removed. (B) Relative contribution of serum bile acid species before and after 12 weeks of treatment with cilofexor 100 mg daily. UDCA and its conjugates have been removed. Abbreviations: CA, cholic acid; CDCA, chenodeoxycholic acid; DCA, deoxycholic acid; GCA, glycocholic acid; GCDCA: glycochenodeoxycholic acid; GDCA, glycolithocholic acid; GLCA, glycolithocholic acid; LCA, lithocholic acid; TCA: taurocholic acid; TCDCA, taurochenodeoxycholic acid; TDCA, taurodeoxycholic acid; TLCA, taurolithocholic acid.
Figure 4
Figure 4
Relative changes in liver biochemistry, markers of fibrosis, and other biomarkers according to achievement of an ALP response, defined as at least a 25% relative reduction in serum ALP from baseline to week 12, in cilofexor‐treated patients. Responder subgroup includes 7 patients treated with cilofexor 100 mg and 1 patient treated with cilofexor 30 mg. P values are according to Wilcoxon rank‐sum test. Abbreviations: BA, bile acid; HA, hyaluronic acid.

References

    1. Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med 2016;375:1161‐1170.
    1. Chapman RW. Primary sclerosing cholangitis: what is the role of ursodeoxycholic acid in therapy for PSC? Nat Rev Gastroenterol Hepatol 2010;7:74‐75.
    1. Lindor KD, Kowdley KV, Luketic VA, Harrison ME, McCashland T, Befeler AS, et al. High‐dose ursodeoxycholic acid for the treatment of primary sclerosing cholangitis. Hepatology 2009;50:808‐814.
    1. Olsson R, Boberg KM, de Muckadell OS, Lindgren S, Hultcrantz R, Folvik G, et al. High‐dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5‐year multicenter, randomized, controlled study. Gastroenterology 2005;129:1464‐1472.
    1. Beuers U, Spengler U, Kruis W, Aydemir U, Wiebecke B, Heldwein W, et al. Ursodeoxycholic acid for treatment of primary sclerosing cholangitis: a placebo‐controlled trial. Hepatology 1992;16:707‐714.
    1. de Vries EM, Wang J, Leeflang MM, Boonstra K, Weersma RK, Beuers UH, et al. Alkaline phosphatase at diagnosis of primary sclerosing cholangitis and 1 year later: evaluation of prognostic value. Liver Int 2016;36:1867‐1875.
    1. Lindstrom L, Hultcrantz R, Boberg KM, Friis‐Liby I, Bergquist A. Association between reduced levels of alkaline phosphatase and survival times of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2013;11:841‐846.
    1. Rupp C, Rossler A, Halibasic E, Sauer P, Weiss KH, Friedrich K, et al. Reduction in alkaline phosphatase is associated with longer survival in primary sclerosing cholangitis, independent of dominant stenosis. Aliment Pharmacol Ther 2014;40:1292‐1301.
    1. Al Mamari S, Djordjevic J, Halliday JS, Chapman RW. Improvement of serum alkaline phosphatase to <1.5 upper limit of normal predicts better outcome and reduced risk of cholangiocarcinoma in primary sclerosing cholangitis. J Hepatol 2013;58:329‐334.
    1. Chiang JY. Bile acids: regulation of synthesis. J Lipid Res 2009;50:1955‐1966.
    1. Chiang JY, Kimmel R, Weinberger C, Stroup D. Farnesoid X receptor responds to bile acids and represses cholesterol 7alpha‐hydroxylase gene (CYP7A1) transcription. J Biol Chem 2000;275:10918‐10924.
    1. Inagaki T, Choi M, Moschetta A, Peng L, Cummins CL, McDonald JG, et al. Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis. Cell Metab 2005;2:217‐225.
    1. Kliewer SA, Mangelsdorf DJ. Fibroblast growth factor 21: from pharmacology to physiology. Am J Clin Nutr 2010;91:254S‐257S.
    1. Song KH, Li T, Owsley E, Strom S, Chiang JY. Bile acids activate fibroblast growth factor 19 signaling in human hepatocytes to inhibit cholesterol 7alpha‐hydroxylase gene expression. Hepatology 2009;49:297‐305.
    1. Schwabl P, Budas G, Hambruch E, Supper P, Burnet M, Lileset J, et al. The FXR agonist GS‐9674 reduces fibrosis and portal hypertension in a rat model of NASH [Abstract]. J Hepatol 2018;68:S471‐S472.
    1. Muir AJ, Taghipour M, Hassanzadeh E, Sahni VA, Sainani N, Shiffman ML, et al. Radiologic progression in primary sclerosing cholangitis by two‐dimensional magnetic resonance cholangiopancreatography: prospective data from a randomized controlled trial [Abstract]. Hepatology 2017;66(Suppl 1):156A.
    1. Vesterhus M, Hov JR, Holm A, Schrumpf E, Nygård S, Godang K, et al. Enhanced liver fibrosis score predicts transplant‐free survival in primary sclerosing cholangitis. Hepatology 2015;62:188‐197.
    1. Djedjos CS, Kirby BJ, Billin A, Gosink JJ, Song Q, Srihari R, et al. Pharmacodynamic effects of the oral, nonsteroidal farnesoid X receptor agonist GS‐9674 in healthy volunteers [Abstract]. Hepatology 2016;63(Suppl 1):543A.
    1. Al‐Khaifi A, Rudling M, Angelin B. An FXR agonist reduces bile acid synthesis independently of increases in FGF19 in healthy volunteers. Gastroenterology 2018;155:1012‐1016.
    1. Ponsioen CY, Chapman RW, Chazouilleres O, Hirschfield GM, Karlsen TH, Lohse AW, et al. Surrogate endpoints for clinical trials in primary sclerosing cholangitis: review and results from an International PSC Study Group consensus process. Hepatology 2016;63:1357‐1367.
    1. Ponsioen CY, Lindor KD, Mehta R, Dimick‐Santos L. Design and endpoints for clinical trials in primary sclerosing cholangitis. Hepatology 2018;68:1174‐1188.
    1. Hirschfield GM, Mason A, Luketic V, Lindor K, Gordon SC, 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. Stanich PP, Bjornsson E, Gossard AA, Enders F, Jorgensen R, Lindor KD. Alkaline phosphatase normalization is associated with better prognosis in primary sclerosing cholangitis. Dig Liver Dis 2011;43:309‐313.
    1. Neuschwander‐Tetri BA, Loomba R, Sanyal AJ, Lavine JE, Van Natta ML, Abdelmalek MF, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non‐cirrhotic, non‐alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo‐controlled trial. Lancet 2015;385:956‐965.

Source: PubMed

3
订阅