A randomised, double-blind, placebo-controlled phase 1 study of the safety, tolerability and pharmacodynamics of volixibat in overweight and obese but otherwise healthy adults: implications for treatment of non-alcoholic steatohepatitis

Melissa Palmer, Lee Jennings, Debra G Silberg, Caleb Bliss, Patrick Martin, Melissa Palmer, Lee Jennings, Debra G Silberg, Caleb Bliss, Patrick Martin

Abstract

Background: Accumulation of toxic free cholesterol in hepatocytes may cause hepatic inflammation and fibrosis. Volixibat inhibits bile acid reuptake via the apical sodium bile acid transporter located on the luminal surface of the ileum. The resulting increase in bile acid synthesis from cholesterol could be beneficial in patients with non-alcoholic steatohepatitis. This adaptive dose-finding study investigated the safety, tolerability, pharmacodynamics, and pharmacokinetics of volixibat.

Methods: Overweight and obese adults were randomised 3:1 to double-blind volixibat or placebo, respectively, for 12 days. Volixibat was initiated at a once-daily dose of 20 mg, 40 mg or 80 mg. Based on the assessment of predefined safety events, volixibat dosing was either escalated or reduced. Other dose regimens (titrations and twice-daily dosing) were also evaluated. Assessments included safety, tolerability, stool hardness, faecal bile acid (FBA) excretion, and serum levels of 7α-hydroxy-4-cholesten-3-one (C4) and lipids.

Results: All 84 randomised participants (volixibat, 63; placebo, 21) completed the study, with no serious adverse events at doses of up to 80 mg per day (maximum assessed dose). The median number of daily bowel evacuations increased from 1 (range 0-4) to 2 (0-8) during volixibat treatment, and stool was looser with volixibat than placebo. Volixibat was minimally absorbed; serum levels were rarely quantifiable at any dose or sampling time point, thereby precluding pharmacokinetic analyses. Mean daily FBA excretion was 930.61 μmol (standard deviation [SD] 468.965) with volixibat and 224.75 μmol (195.403) with placebo; effects were maximal at volixibat doses ≥20 mg/day. Mean serum C4 concentrations at day 12 were 98.767 ng/mL (standard deviation, 61.5841) with volixibat and 16.497 ng/mL (12.9150) with placebo. Total and low-density lipoprotein cholesterol levels decreased in the volixibat group, with median changes of - 0.70 mmol/L (range - 2.8 to 0.4) and - 0.6990 mmol/L (- 3.341 to 0.570), respectively.

Conclusions: This study indicates that maximal inhibition of bile acid reabsorption, as assessed by FBA excretion, occurs at volixibat doses of ≥20 mg/day in obese and overweight adults, without appreciable change in gastrointestinal tolerability. These findings guided dose selection for an ongoing phase 2 study in patients with non-alcoholic steatohepatitis.

Trial registration: ClinicalTrials.gov identifier: NCT02287779 (registration first received 6 November 2014).

Keywords: Apical sodium-dependent bile acid transporter (ASBT); Cholesterol; LUM002; Non-alcoholic fatty liver disease; Non-alcoholic steatohepatitis; Obesity; SHP626; Volixibat.

Conflict of interest statement

Ethics approval and consent to participate

The study was conducted in accordance with International Conference for Harmonisation guidelines for Good Clinical Practice, the principles of the Declaration of Helsinki, and other applicable local ethical and legal requirements. The study protocol was approved by an independent institutional review board (Crescent City Institutional Review Board) and regulatory agency before initiation. Each participant provided written informed consent before commencing any study-specific procedures.

Consent for publication

Not applicable.

Competing interests

All authors are employees of Shire and may own stock or stock options. This study was funded by Shire Development LLC. Volixibat is a Shire investigational medical product. Shire develops and markets treatments for gastrointestinal and metabolic diseases, including NASH.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Study design. Each cohort consisted of 12 participants (volixibat, n = 9; placebo, n = 3); details are shown for the volixibat arm only. Light-grey boxes indicate dose regimen options that were not undertaken. Bold text indicates alterations to planned doses. Cohorts 4 and onwards were each initiated after reviewing results from previous cohorts.*Study days are approximate. Cohort 2 treatment was to begin at least 4 days after cohort 1 treatment; cohort 3 treatment was to start after completion of treatment in cohort 2.†Changed from 80 mg q.d. following review of data from cohorts 1 and 2 to a descending dose titration of 80–40–20 mg q.d. ‡Results from cohorts 1–3 triggered the use of intermediate and reduced doses in cohorts 4 and 5, instead of increased doses. ¶Treatment of an optional second b.i.d. dose cohort was not undertaken. §Treatment of an optional second q.d. or b.i.d. dose titration cohort was not undertaken. AE, adverse event; b.i.d., twice daily; FBA, faecal bile acid; q.d., once daily
Fig. 2
Fig. 2
Mean daily faecal bile acid excretion (a) at baseline and days 11–12, and (b) change from baseline to days 11–12. Baseline was days − 2 and − 1 for this data set. Data are from the pharmacodynamic analysis set. b.i.d., twice daily; CI, confidence interval; q.d., once daily; SD, standard deviation
Fig. 3
Fig. 3
Absolute serum 7α-hydroxy-4-cholesten-3-one (C4) concentration at (a) baseline and 13 h after dosing on day 12, and (b) change from baseline to 13 h after dosing on day 12. Baseline was the last observation before the first dose of study drug. Data are from the pharmacodynamic analysis set. b.i.d., twice daily; CI, confidence interval; q.d., once daily; SD, standard deviation
Fig. 4
Fig. 4
Daily frequency of bowel movements. Baseline was day − 1 for this data set. Squares represent medians; shaded bars represent ranges. Data are from the pharmacodynamic analysis set. b.i.d., twice daily; q.d., once daily

References

    1. Chalasani N, Younossi Z, Lavine JE, Diehl AM, Brunt EM, Cusi K, Charlton M, Sanyal AJ. The diagnosis and management of non-alcoholic fatty liver disease: practice guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology. 2012;55(6):2005–2023. doi: 10.1002/hep.25762.
    1. Masuoka HC, Chalasani N. Nonalcoholic fatty liver disease: an emerging threat to obese and diabetic individuals. Ann N Y Acad Sci. 2013;1281:106–122. doi: 10.1111/nyas.12016.
    1. Rinella ME. Nonalcoholic fatty liver disease: a systematic review. JAMA. 2015;313(22):2263–2273. doi: 10.1001/jama.2015.5370.
    1. Musso G, Gambino R, Cassader M. Cholesterol metabolism and the pathogenesis of non-alcoholic steatohepatitis. Prog Lipid Res. 2013;52(1):175–191. doi: 10.1016/j.plipres.2012.11.002.
    1. Williams CD, Stengel J, Asike MI, Torres DM, Shaw J, Contreras M, Landt CL, Harrison SA. Prevalence of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis among a largely middle-aged population utilizing ultrasound and liver biopsy: a prospective study. Gastroenterology. 2011;140(1):124–131. doi: 10.1053/j.gastro.2010.09.038.
    1. Zezos P, Renner EL. Liver transplantation and non-alcoholic fatty liver disease. World J Gastroenterol. 2014;20(42):15532–15538. doi: 10.3748/wjg.v20.i42.15532.
    1. Bhala N, Jouness RI, Bugianesi E. Epidemiology and natural history of patients with NAFLD. Curr Pharm Des. 2013;19(29):5169–5176. doi: 10.2174/13816128113199990336.
    1. Hyysalo J, Mannisto VT, Zhou Y, Arola J, Karja V, Leivonen M, Juuti A, Jaser N, Lallukka S, Kakela P, et al. A population-based study on the prevalence of NASH using scores validated against liver histology. J Hepatol. 2014;60(4):839–846. doi: 10.1016/j.jhep.2013.12.009.
    1. Vernon G, Baranova A, Younossi ZM. Systematic review: the epidemiology and natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in adults. Aliment Pharmacol Ther. 2011;34(3):274–285. doi: 10.1111/j.1365-2036.2011.04724.x.
    1. Ekstedt M, Franzen LE, Mathiesen UL, Thorelius L, Holmqvist M, Bodemar G, Kechagias S. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology. 2006;44(4):865–873. doi: 10.1002/hep.21327.
    1. Wong VW, Wong GL, Choi PC, Chan AW, Li MK, Chan HY, Chim AM, Yu J, Sung JJ, Chan HL. Disease progression of non-alcoholic fatty liver disease: a prospective study with paired liver biopsies at 3 years. Gut. 2010;59(7):969–974. doi: 10.1136/gut.2009.205088.
    1. Fassio E, Alvarez E, Dominguez N, Landeira G, Longo C. Natural history of nonalcoholic steatohepatitis: a longitudinal study of repeat liver biopsies. Hepatology. 2004;40(4):820–826.
    1. Adams LA, Sanderson S, Lindor KD, Angulo P. The histological course of nonalcoholic fatty liver disease: a longitudinal study of 103 patients with sequential liver biopsies. J Hepatol. 2005;42(1):132–138. doi: 10.1016/j.jhep.2004.09.012.
    1. McPherson S, Hardy T, Henderson E, Burt AD, Day CP, Anstee QM. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: implications for prognosis and clinical management. J Hepatol. 2015;62(5):1148–1155. doi: 10.1016/j.jhep.2014.11.034.
    1. Angulo P. Nonalcoholic fatty liver disease. N Engl J Med. 2002;346(16):1221–1231. doi: 10.1056/NEJMra011775.
    1. Wong RJ, Aguilar M, Cheung R, Perumpail RB, Harrison SA, Younossi ZM, Ahmed A. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology. 2015;148(3):547–555. doi: 10.1053/j.gastro.2014.11.039.
    1. Banini BA. Nonalcoholic steatohepatitis (NASH) has surpassed hepatitis C as the leading etiology for listing for liver transplant: implications for NASH in children and young adults. In: American College of Gastroenterology Annual Scientific Meeting (Congress Abstract 46). 2016. Accessed 17 Nov 2017.
    1. European Association for the Study of the Liver European Association for the Study of diabetes, European Association for the Study of obesity: EASL-EASD-EASO clinical practice guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388–1402. doi: 10.1016/j.jhep.2015.11.004.
    1. Kassirer JP, Angell M. Losing weight – an ill-fated new Year's resolution. N Engl J Med. 1998;338(1):52–54. doi: 10.1056/NEJM199801013380109.
    1. Hallsworth K, Avery L, Trenell MI. Targeting lifestyle behavior change in adults with NAFLD during a 20-min consultation: summary of the dietary and exercise literature. Curr Gastroenterol Rep. 2016;18(3):11. doi: 10.1007/s11894-016-0485-1.
    1. Keller B, Dorenbaum A, Wynne D, Gedulin B, Setchell K, Olek E, Levin N, Kennedy C. Effect of apical sodium-dependent bile acid transporter (ASBT) inhibition on serum and fecal bile acids in healthy volunteers (congress abstract 55). Falk Symposium. 2014;194
    1. Shneider BL. Intestinal bile acid transport: biology, physiology, and pathophysiology. J Pediatr Gastroenterol Nutr. 2001;32(4):407–417. doi: 10.1097/00005176-200104000-00002.
    1. Halilbasic E, Claudel T, Trauner M. Bile acid transporters and regulatory nuclear receptors in the liver and beyond. J Hepatol. 2013;58(1):155–168. doi: 10.1016/j.jhep.2012.08.002.
    1. Hylemon PB, Zhou H, Pandak WM, Ren S, Gil G, Dent P. Bile acids as regulatory molecules. J Lipid Res. 2009;50(8):1509–1520. doi: 10.1194/jlr.R900007-JLR200.
    1. Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368(9548):1696–1705. doi: 10.1016/S0140-6736(06)69705-5.
    1. Dunning BE, Foley JE, Ahren B. Alpha cell function in health and disease: influence of glucagon-like peptide-1. Diabetologia. 2005;48(9):1700–1713. doi: 10.1007/s00125-005-1878-0.
    1. Gutzwiller JP, Goke B, Drewe J, Hildebrand P, Ketterer S, Handschin D, Winterhalder R, Conen D, Beglinger C. Glucagon-like peptide-1: a potent regulator of food intake in humans. Gut. 1999;44(1):81–86. doi: 10.1136/gut.44.1.81.
    1. Cyphert HA, Ge X, Kohan AB, Salati LM, Zhang Y, Hillgartner FB. Activation of the farnesoid X receptor induces hepatic expression and secretion of fibroblast growth factor 21. J Biol Chem. 2012;287(30):25123–25138. doi: 10.1074/jbc.M112.375907.
    1. Pournaras DJ, Glicksman C, Vincent RP, Kuganolipava S, Alaghband-Zadeh J, Mahon D, Bekker JH, Ghatei MA, Bloom SR, Walters JR, et al. The role of bile after roux-en-Y gastric bypass in promoting weight loss and improving glycaemic control. Endocrinology. 2012;153(8):3613–3619. doi: 10.1210/en.2011-2145.
    1. Lambert G, Amar MJ, Guo G, Brewer HB, Jr, Gonzalez FJ, Sinal CJ. The farnesoid X-receptor is an essential regulator of cholesterol homeostasis. J Biol Chem. 2003;278(4):2563–2570. doi: 10.1074/jbc.M209525200.
    1. Modica S, Gadaleta RM, Moschetta A. Deciphering the nuclear bile acid receptor FXR paradigm. Nucl Recept Signal. 2010;8:e005.
    1. Halilbasic E, Baghdasaryan A, Trauner M. Nuclear receptors as drug targets in cholestatic liver diseases. Clin Liver Dis. 2013;17(2):161–189. doi: 10.1016/j.cld.2012.12.001.
    1. Chen L, Yao X, Young A, McNulty J, Anderson D, Liu Y, Nystrom C, Croom D, Ross S, Collins J, et al. Inhibition of apical sodium-dependent bile acid transporter as a novel treatment for diabetes. Am J Physiol Endocrinol Metab. 2012;302(1):E68–E76. doi: 10.1152/ajpendo.00323.2011.
    1. Gedulin B. Apical sodium-dependent bile transport inhibitors (ASBTi) exhibit potent antidiabetic activity in ZDF rats. 49th EASD Annual Meeting.
    1. West KL, Zern TL, Butteiger DN, Keller BT, Fernandez ML. SC-435, an ileal apical sodium co-dependent bile acid transporter (ASBT) inhibitor lowers plasma cholesterol and reduces atherosclerosis in Guinea pigs. Atherosclerosis. 2003;171(2):201–210. doi: 10.1016/j.atherosclerosis.2003.08.019.
    1. Tiessen RG, Kennedy C, Keller B, Levin N, Acevedo L, Gedulin C, van Vliet A, Dorenbaum A, Palmer M. Randomized controlled trial: safety, tolerability, pharmacokinetics and pharmacodynamics of apical sodium-dependent bile acid transporter inhibition with volixibat in healthy adults and patients with type 2 diabetes mellitus. BMC Gasteroenterology. 2017; [ms submitted]
    1. Tiessen RG, Kennedy C, Keller BT, Levin N, Acevedo L, Wynne D, Gedulin B, van Vliet A, Olek E, Dorenbaum A: LUM002 positive metabolic profile shown after administration of 10mg for 28 days in type 2 diabetes mellitus patients leading to potential treatment for patients with nonalcoholic steatohepatitis (NASH). Hepatology 2014, 60(S1):629A.
    1. Siebers N, Palmer M, Silberg DG, Jennings L, Bliss C, Martin PT. Absorption, Distribution, Metabolism, and excretion of [14C]-Volixibat in healthy men: phase 1 open-label study. Eur J Drug Metab Pharmacokinet. 2017; 10.1007/s13318-017-0429-7. [Epub ahead of print]
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol. 1997;32(9):920–924. doi: 10.3109/00365529709011203.
    1. Chiang DJ, Pritchard MT, Nagy LE. Obesity, diabetes mellitus, and liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2011;300(5):G697–G702. doi: 10.1152/ajpgi.00426.2010.
    1. Tremont SJ, Lee LF, Huang HC, Keller BT, Banerjee SC, Both SR, Carpenter AJ, Wang CC, Garland DJ, Huang W, et al. Discovery of potent, nonsystemic apical sodium-codependent bile acid transporter inhibitors (part 1) J Med Chem. 2005;48(18):5837–5852. doi: 10.1021/jm040215+.
    1. Wilcox C, Turner J, Green J. Systematic review: the management of chronic diarrhoea due to bile acid malabsorption. Aliment Pharmacol Ther. 2014;39(9):923–939. doi: 10.1111/apt.12684.
    1. Chalasani N. Statins and hepatotoxicity: focus on patients with fatty liver. Hepatology. 2005;41(4):690–695. doi: 10.1002/hep.20671.
    1. Herzog E, Pragst I, Waelchli M, Gille A, Schenk S, Mueller-Cohrs J, Diditchenko S, Zanoni P, Cuchel M, Seubert A, et al. Reconstituted high-density lipoprotein can elevate plasma alanine aminotransferase by transient depletion of hepatic cholesterol: role of the phospholipid component. J Appl Toxicol. 2016;36(8):1038–1047. doi: 10.1002/jat.3264.
    1. Rao A, Kosters A, Mells JE, Zhang W, Setchell KD, Amanso AM, Wynn GM, Xu T, Keller BT, Yin H, et al. Inhibition of ileal bile acid uptake protects against nonalcoholic fatty liver disease in high-fat diet-fed mice. Sci Transl Med. 2016;8(357):357ra122. doi: 10.1126/scitranslmed.aaf4823.

Source: PubMed

3
Subscribe