PRIMIS: design of a pivotal, randomized, phase 3 study evaluating the safety and efficacy of the nonsteroidal farnesoid X receptor agonist cilofexor in noncirrhotic patients with primary sclerosing cholangitis

Michael Trauner, Chuhan Chung, Kate Sterling, Xiangyu Liu, Xiaomin Lu, Jun Xu, Clare Tempany-Afdhal, Zachary D Goodman, Martti Färkkilä, Atsushi Tanaka, Palak Trivedi, Kris V Kowdley, Christopher L Bowlus, Cynthia Levy, Robert P Myers, Michael Trauner, Chuhan Chung, Kate Sterling, Xiangyu Liu, Xiaomin Lu, Jun Xu, Clare Tempany-Afdhal, Zachary D Goodman, Martti Färkkilä, Atsushi Tanaka, Palak Trivedi, Kris V Kowdley, Christopher L Bowlus, Cynthia Levy, Robert P Myers

Abstract

Background: Primary sclerosing cholangitis (PSC) is a chronic progressive liver disease leading to biliary fibrosis and cirrhosis. Cilofexor is a nonsteroidal farnesoid X receptor agonist that demonstrated significant improvements in liver biochemistry and markers of cholestasis in patients with PSC in a phase 2 study. We describe here the rationale, design, and implementation of the phase 3 PRIMIS trial, the largest placebo-controlled trial in PSC.

Methods: Adults with large-duct PSC without cirrhosis are randomized 2:1 to receive oral cilofexor 100 mg once daily or placebo for up to 96 weeks during the blinded phase. Patients completing the blinded phase are eligible to receive open-label cilofexor 100 mg daily for up to 96 weeks. The primary objective is to evaluate whether cilofexor reduces the risk of fibrosis progression compared with placebo. Liver biopsy is performed at screening and Week 96 of the blinded phase for histologic assessment of fibrosis. The primary endpoint-chosen in conjunction with guidance from the U.S. Food and Drug Administration-is the proportion of patients with ≥ 1-stage increase in fibrosis according to Ludwig histologic classification at week 96. Secondary objectives include evaluation of changes in liver biochemistry, serum bile acids, liver fibrosis assessed by noninvasive methods, health-related quality of life, and safety of cilofexor.

Conclusion: The phase 3 PRIMIS study is the largest randomized, double-blind, placebo-controlled trial in PSC to date and will allow for robust evaluation of the efficacy and safety of cilofexor in noncirrhotic patients with large-duct PSC.

Trial registration: ClinicalTrials.gov NCT03890120; registered 26/03/2019.

Keywords: Farnesoid X receptor; Liver fibrosis; Primary sclerosing cholangitis.

Conflict of interest statement

MT consults, is on the speakers’ bureau, and has received grants from Gilead, Falk, Intercept, and MSD; consults and has received grants from Albireo; consults for BiomX, Boehringer Ingelheim, Genfit, Janssen, Novartis, Phenex, Pliant, Regulus, and Shire; is on the speakers’ bureau for BMS and Roche; and has received grants from Alnylam, CymaBay, Takeda, and UltraGenyx. CC, KS, X Liu, X Lu, and JX are employed by and own stock in Gilead. CT-A receives funding from Gilead; serves as a medical advisor to Profound Medical and Promaxo; and is an independent contractor for Medscape. ZDG has no personal conflicts of interest with respect to this work; his institution currently receives funding to support his research from Gilead, BMS, CymaBay, Eiger, Inventiva, MSD, NGM, and Novartis. MF has received grant support from Gilead; and is participating in a clinical trial of norUDCA in a study sponsored by Falk. AT consults for Gilead, EA Pharma, and GSK; and has received grants from AbbVie and Chugai. PT is on the speakers’ bureau of Falk and Intercept; consults and has received grants from Gilead, BMS, Falk, Intercept, LifeArc, Medical Research Foundation, National Institute of Health Research, Perspectum, and Wellcome Trust; and is an advisor for CymaBay, Falk, Intercept, and Pliant. KVK has received grant support from Gilead, 89bio, BMS, Celgene, Corcept, CymaBay, Enanta, Genfit, GSK, Hanmi, HighTide, Intercept, Madrigal, Metacrine, Mirum, NGM, Pfizer, Pliant, Protagonist, Terns, and Viking; serves as a consultant and on advisory boards for Gilead, 89bio, CymaBay, Enanta; Genfit, HighTide, Inipharm, Intercept, Madrigal, Mirum, NGM, and Pfizer, and Enanta; and is on speakers’ bureaus for Gilead, AbbVie, and Intercept. CLB advises and has received grants from Gilead, CymaBay, Eli Lilly, and Intercept; advises BiomX, Parvus, Patara, and Pliant; and has received grants from Arena, BMS, Genkyotex, GSK, and Takeda. CL advises and has received grants from Gilead, Cara, CymaBay, Genfit, Genkyotex, GSK, Intercept, Mirum, Pliant, and Target RWE; advises Escient and Teva; and has received grants from Alnylam, Mitsubishi, NGM, Novartis, and Zydus. RPM was formerly employed by Gilead.

© 2023. The Author(s).

Figures

Fig. 1
Fig. 1
PRIMIS study design. PSC, primary sclerosing cholangitis; UDCA, ursodeoxycholic acid
Fig. 2
Fig. 2
Nonworsening of fibrosis at Weeks 48 and 96 among noncirrhotic patients in the phase 2 simtuzumab primary sclerosing cholangitis (PSC) study was associated with significantly reduced rate of PSC-related events. p values by Fisher’s exact test
Fig. 3
Fig. 3
Greater fibrosis burden at baseline, as defined by Ludwig fibrosis stage or hepatic collagen content by morphometry, was associated with a significantly increased risk of disease progression among noncirrhotic patients with PSC in the phase 2 simtuzumab study. Disease progression was defined by progression to cirrhosis (F4), ascending cholangitis, hepatic decompensation, liver transplantation, or death. [15] p values by log-rank test

References

    1. Chapman MH, Thorburn D, Hirschfield GM, et al. British Society of Gastroenterology and UK-PSC guidelines for the diagnosis and management of primary sclerosing cholangitis. Gut. 2019;68:1356–1378. doi: 10.1136/gutjnl-2018-317993.
    1. Chapman R, Fevery J, Kalloo A, et al. Diagnosis and management of primary sclerosing cholangitis. Hepatology. 2010;51:660–678. doi: 10.1002/hep.23294.
    1. Hirschfield GM, Karlsen TH, Lindor KD, Adams DH. Primary sclerosing cholangitis. Lancet. 2013;382:1587–99. doi: 10.1016/S0140-6736(13)60096-3.
    1. Trivedi PJ, Bowlus CL, Yimam KK, et al. Epidemiology, natural history, and outcomes of primary sclerosing cholangitis: a systematic review of population-based studies. Clin Gastroenterol Hepatol. 2021;S1542–3565(21):00919–928.
    1. Boonstra K, Beuers U, Ponsioen CY. Epidemiology of primary sclerosing cholangitis and primary biliary cirrhosis: a systematic review. J Hepatol. 2012;56:1181–1188. doi: 10.1016/j.jhep.2011.10.025.
    1. Chapman RW. Update on primary sclerosing cholangitis. Clin Liver Dis. 2017;9:107–110. doi: 10.1002/cld.633.
    1. Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med. 2016;375:1161–1170. doi: 10.1056/NEJMra1506330.
    1. Takakura WR, Tabibian JH, Bowlus CL. The evolution of natural history of primary sclerosing cholangitis. Curr Opin Gastroenterol. 2017;33:71–77. doi: 10.1097/MOG.0000000000000333.
    1. Karlsen TH, Folseraas T, Thorburn D, Vesterhus M. Primary sclerosing cholangitis–a comprehensive review. J Hepatol. 2017;67:1298–1323. doi: 10.1016/j.jhep.2017.07.022.
    1. Kuo A, Gomel R, Safer R, et al. Characteristics and outcomes reported by patients with primary sclerosing cholangitis through an online registry. Clin Gastroenterol Hepatol. 2019;17:1372–1378. doi: 10.1016/j.cgh.2018.04.047.
    1. Walmsley M, Langford A, Thorburn D, et al. Clinical need in PSC and clinically meaningful change: what is important to patients? March 3, 2016. Available at: . Accessed 22 Nov 2021.
    1. Younossi ZM, Afendy A, Stepanova M, et al. Development and validation of a primary sclerosing cholangitis-specific patient-reported outcomes instrument: the PSC PRO. Hepatology. 2018;68:155–165. doi: 10.1002/hep.29664.
    1. Fung BM, Lindor KD, Tabibian JH. Cancer risk in primary sclerosing cholangitis: epidemiology, prevention, and surveillance strategies. World J Gastroenterol. 2019;25:659–671. doi: 10.3748/wjg.v25.i6.659.
    1. Tabibian JH, Ali AH, Lindor KD. Primary sclerosing cholangitis, part 2: cancer risk, prevention, and surveillance. Gastroenterol Hepatol. 2018;14:427–432.
    1. Muir AJ, Levy C, Janssen HLA, et al. Simtuzumab for primary sclerosing cholangitis: phase 2 study results with insights on the natural history of the disease. Hepatology. 2019;69:684–698. doi: 10.1002/hep.30237.
    1. Krones E, Marschall H-U, Fickert P. Future medical treatment of PSC. Curr Hepatology Rep. 2019;18:96–106. doi: 10.1007/s11901-019-00454-4.
    1. Tabibian JH, Ali AH, Lindor KD. Primary sclerosing cholangitis, part 1: epidemiology, etiopathogenesis, clinical features, and treatment. Gastroenterol Hepatol. 2018;14:293–304.
    1. Henson JB, Patel YA, King LY, et al. Outcomes of liver retransplantation in patients with primary sclerosing cholangitis. Liver Transpl. 2017;23:769–780. doi: 10.1002/lt.24703.
    1. Steenstraten IC, Sebib Korkmaz K, Trivedi PJ, et al. Systematic review with meta-analysis: risk factors for recurrent primary sclerosing cholangitis after liver transplantation. Aliment Pharmacol Ther. 2019;49:636–643. doi: 10.1111/apt.15148.
    1. Arndtz K, Hirschfield GM. Quality of life and primary sclerosing cholangitis: the business of defining what counts. Hepatology. 2018;68:16–18. doi: 10.1002/hep.29775.
    1. Ranieri V, McKay K, Walmsley M, et al. Primary sclerosing cholangitis and psychological wellbeing: a scoping review. Semin Liver Dis. 2019;39:104–110. doi: 10.1055/s-0038-1676099.
    1. Walmsley M, Leburgue A, Thorburn D, et al. Identifying research priorities in primary sclerosing cholangitis: driving clinically meaningful change from the patients’ perspective. J Hepatol. 2019;70:e412–e413. doi: 10.1016/S0618-8278(19)30812-6.
    1. Tabibian JH, Bowlus CL. Primary sclerosing cholangitis: a review and update. Liver Res. 2017;1:221–230. doi: 10.1016/j.livres.2017.12.002.
    1. Vesterhus M, Karlsen TH. Emerging therapies in primary sclerosing cholangitis: pathophysiological basis and clinical opportunities. J Gastroenterol. 2020;55:588–614. doi: 10.1007/s00535-020-01681-z.
    1. Chiang JY. Bile acids: regulation of synthesis. J Lipid Res. 2009;50:1955–1966. doi: 10.1194/jlr.R900010-JLR200.
    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. doi: 10.1074/jbc.275.15.10918.
    1. Trauner M, Gulamhusein A, Hameed B, et al. The nonsteroidal farnesoid X receptor agonist cilofexor (GS-9674) improves markers of cholestasis and liver injury in patients with primary sclerosing cholangitis. Hepatology. 2019;70:788–801. doi: 10.1002/hep.30509.
    1. Schwabl P, Hambruch E, Budas GR, et al. The non-steroidal FXR agonist cilofexor improves portal hypertension and reduces hepatic fibrosis in a rat NASH model. Biomedicines. 2021;9:60. doi: 10.3390/biomedicines9010060.
    1. Sroda N, Fuchs CD, Suriben R, et al. Cilofexor reduces fibrosis and improves measures of liver function in the Mdr2 knockout mouse model of biliary fibrosis. Presented at The Liver Meeting® (AASLD), 2021; poster 1254.
    1. Ludwig J, Dickson ER, McDonald GS. Staging of chronic nonsuppurative destructive cholangitis (syndrome of primary biliary cirrhosis) Virchows Arch A Pathol Anat Histol. 1978;379:103–112. doi: 10.1007/BF00432479.
    1. Patel K, Harrison SA, Elkhashab M, et al. Cilofexor, a nonsteroidal FXR agonist, in patients with non-cirrhotic NASH: a phase 2 randomized controlled trial. Hepatology. 2020;72:58–71. doi: 10.1002/hep.31205.
    1. Djedjos CS, Kirby BJ, Billin A, 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. Kirby BJ, Djedjos CS, Birkeback J, et al. Evaluation of the safety and pharmacokinetics of the oral, nonsteroidal farnesoid x receptor agonist GS-9674 in healthy volunteers. Presented at The Liver Meeting (AASLD); 2016; Boston, MA; poster 1140.
    1. Fleming TR. Surrogate endpoints and FDA's accelerated approval process. Health Aff (Millwood) 2005;24:67–78. doi: 10.1377/hlthaff.24.1.67.
    1. Ponsioen CY. Endpoints in the design of clinical trials for primary sclerosing cholangitis. Biochim Biophys Acta Mol Basis Dis. 2018;1864:1410–1414. doi: 10.1016/j.bbadis.2017.08.015.
    1. Ponsioen CY, Chapman RW, Chazouillères O, 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. doi: 10.1002/hep.28256.
    1. Trivedi PJ, Muir AJ, Levy C, et al. Inter- and intra-individual variation, and limited prognostic utility, of serum alkaline phosphatase in a trial of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol. 2021;19:1248–1257. doi: 10.1016/j.cgh.2020.07.032.
    1. de Vries EMG, de Krijger M, Färkkilä M, et al. Validation of the prognostic value of histologic scoring systems in primary sclerosing cholangitis: an international cohort study. Hepatology. 2017;65:907–919. doi: 10.1002/hep.28963.
    1. Ruiz A, Lemoinne S, Carrat F, et al. Radiologic course of primary sclerosing cholangitis: assessment by three-dimensional magnetic resonance cholangiography and predictive features of progression. Hepatology. 2014;59:242–250. doi: 10.1002/hep.26620.
    1. Olsson R, Boberg KM, de Muckadell OS, et al. High-dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5-year multicenter, randomized, controlled study. Gastroenterology. 2005;129:1464–1472. doi: 10.1053/j.gastro.2005.08.017.
    1. European Association for the Study of the Liver EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol. 2009;51:237–267. doi: 10.1016/j.jhep.2009.04.009.
    1. Corpechot C, Gaouar F, El Naggar A, et al. Baseline values and changes in liver stiffness measured by transient elastography are associated with severity of fibrosis and outcomes of patients with primary sclerosing cholangitis. Gastroenterology. 2014;146:970–979. doi: 10.1053/j.gastro.2013.12.030.
    1. Vesterhus M, Hov JR, Holm A, et al. Enhanced liver fibrosis score predicts transplant-free survival in primary sclerosing cholangitis. Hepatology. 2015;62:188–197. doi: 10.1002/hep.27825.
    1. Loomba R, Noureddin M, Kowdley KV, et al. Combination therapies including cilofexor and firsocostat for bridging fibrosis and cirrhosis attributable to NASH. Hepatology. 2021;73:625–643. doi: 10.1002/hep.31622.

Source: PubMed

3
S'abonner