BMS-986263 in patients with advanced hepatic fibrosis: 36-week results from a randomized, placebo-controlled phase 2 trial

Eric J Lawitz, Diane E Shevell, Giridhar S Tirucherai, Shuyan Du, Warner Chen, Uma Kavita, Angie Coste, Fred Poordad, Morten Karsdal, Mette Nielsen, Zachary Goodman, Edgar D Charles, Eric J Lawitz, Diane E Shevell, Giridhar S Tirucherai, Shuyan Du, Warner Chen, Uma Kavita, Angie Coste, Fred Poordad, Morten Karsdal, Mette Nielsen, Zachary Goodman, Edgar D Charles

Abstract

Background and aims: Hepatic fibrosis secondary to HCV infection can lead to cirrhosis and hepatic decompensation. Sustained virologic response (SVR) is possible with direct-acting antiviral drug regimens; however, patients with advanced fibrosis have an increased risk for HCC. Heat shock protein 47 (HSP47), a key collagen chaperone, has been implicated in fibrosis development. We evaluated the efficacy and safety of BMS-986263, a lipid nanoparticle delivering small interfering RNA designed to degrade HSP47 mRNA, for the treatment of advanced fibrosis.

Approach and results: NCT03420768 was a Phase 2, randomized (1:1:2), placebo-controlled trial conducted at a hepatology clinic in the United States. Patients with HCV-SVR (for ≥ 1 year) and advanced fibrosis received once-weekly i.v. infusions of placebo or BMS-986263 (45 or 90 mg) for 12 weeks. The primary endpoint was ≥ 1 METAVIR stage improvement at Week 12; key secondary endpoints included Ishak score improvement, pharmacokinetics, fibrosis biomarkers, and safety. All 61 patients completed treatment, and 2/15 (13%, placebo), 3/18 (17%, 45 mg), and 6/28 (21%, 90 mg) had METAVIR improvements of ≥ 1 stage at Week 12. Five patients in the 90-mg arm had Ishak improvements by ≥ 2 stages. BMS-986263 plasma concentrations increased in a generally dose-proportional fashion between BMS-986263 doses, with no notable accumulation with weekly dosing. All adverse events (AEs) were mild or moderate in intensity; most treatment-related AEs were infusion-related reactions in the BMS-986263 arms. At baseline, collagen levels were low, indicating low levels of fibrogenesis in these patients.

Conclusions: In patients with HCV-SVR, BMS-986263 administration was generally well tolerated through Week 36 and resulted in METAVIR and Ishak score improvements. Further evaluation of BMS-986263 in patients with active fibrogenesis is warranted.

Conflict of interest statement

Dr. Lawitz consults for and received grants from Metacrine, Boehringer Ingelheim, and Bristol Myers Squibb. He received grants from 89Bio, AbbVie, Akcea, Arena, Celgene, Conatus, Durect, Enanta, Enyo, Galectin, Galmed, Genfit, Gilead, Hanmi, Intercept, Madrigal, Novartis, Novo Nordisk, Octeta, and Zydus. Dr. Shevell was an employee of Bristol Myers Squibb when the study was performed and owns company stock. Dr. Tirucherai is employed by, owns stock in, and holds intellectual property rights with Bristol Myers Squibb. Dr. Du is employed by and owns stock in Bristol Myers Squibb. Dr. Karsdal is employed by and owns stock in Nordic Bioscience. Dr. Nielsen is employed by and owns stock in Nordic Bioscience. M. Karsdal and M. Nielsen are among the original inventors and patent holders of assays to measure PRO‐C3 and C3M. Dr. Charles is employed by, owns stock in, and holds intellectual property rights with Bristol Myers Squibb. W. Chen was an employee of Bristol Myers Squibb at the time of this study and may hold company stock. U. Kavita was an employee of Bristol Myers Squibb at the time of this study and may hold company stock. A. Coste has no disclosures to report. F. Poordad has participated in speakers’ bureaus for and received grants from Gilead Sciences and Intercept Pharmaceuticals. Z. Goodman has received grants from Alexion Pharmaceuticals, Allergan, Conatus Pharmaceuticals, Exalenz Bioscience, Galactin Therapeutics, Gilead Sciences, and Intercept Pharmaceuticals.

© 2021 Bristol Myers Squibb. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.

Figures

FIGURE 1
FIGURE 1
NCT03420768 study design. Eligible patients were randomized 1:1:2 to receive placebo or BMS‐986263 (45 mg or 90 mg) i.v. QW for 12 weeks. aLiver biopsies were performed within 8 weeks prior to or during the screening period
FIGURE 2
FIGURE 2
METAVIR stage change from baseline to Week 12. Patients with ≥ 1 METAVIR stage improvement at Week 12 stratified by (A) arm or (B) baseline fibrosis stage. aOR and difference in response rates are shown for each arm compared with placebo. bOne F2 patient had an inadequate Week 12 biopsy specimen and was considered to have no fibrosis improvement
FIGURE 3
FIGURE 3
Ishak score change from baseline to Week 12. Patients with ≥ 2 Ishak score improvement at Week 12 stratified by study arm and baseline fibrosis stage. aOne F2 patient had an inadequate Week 12 biopsy specimen and was excluded from the analysis
FIGURE 4
FIGURE 4
Liver HSP47 mRNA and protein change from baseline to Week 12. (A) Percentage of patients per arm with reduced HSP47 mRNA at Week 12. (B,C) Median percentage change in HSP47 mRNA (B) and HSP47 protein (C). aMissing data, n = 1. bMissing data, n = 2
FIGURE 5
FIGURE 5
Geometric mean BMS‐986263 plasma concentration. Figure shows the mean concentration time profile following administration of all infusions in the study. PK samples were collected at preinfusion, midinfusion, and postinfusion at Weeks 0, 1, 4, 6, and 11. Trough PK samples were collected preinfusion at Weeks 2, 5, 7, and 8. Samples during a dosing interval were collected at 72 h postinfusion following the doses at Weeks 0 and 6. Additional postinfusion PK samples were collected at 1 and 3 weeks following the Week 11 dose

References

    1. Weiskirchen R, Weiskirchen S, Tacke F. Recent advances in understanding liver fibrosis: bridging basic science and individualized treatment concepts. F1000Res 2018;7:921.
    1. Vilar‐Gomez E, Calzadilla‐Bertot L, Wai‐Sun Wong V, Castellanos M, Aller‐de la Fuente R, Metwally M, et al. Fibrosis severity as a determinant of cause‐specific mortality in patients with advanced nonalcoholic fatty liver disease: a multi‐national cohort study. Gastroenterology. 2018;155:443–57.e17.
    1. Dulai PS, Singh S, Patel J, Soni M, Prokop LJ, Younossi Z, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta‐analysis. Hepatology. 2017;65:1557–65.
    1. Xu F, Moorman AC, Tong X, Gordon SC, Rupp LB, Lu M, et al. All‐cause mortality and progression risks to hepatic decompensation and hepatocellular carcinoma in patients infected with hepatitis C virus. Clin Infect Dis. 2016;62:289–97.
    1. Curry MP, Tapper EB, Bacon B, Dieterich D, Flamm SL, Guest L, et al. Effectiveness of 8‐ or 12‐weeks of ledipasvir and sofosbuvir in real‐world treatment‐naive, genotype 1 hepatitis C infected patients. Aliment Pharmacol Ther. 2017;46:540–8.
    1. American Association for the Study of Liver Diseases , Infectious Diseases Society of America . When and in whom to initiate HCV therapy. HCV guidance: recommendations for testing, managing, and treating hepatitis C. Available at: . Accessed 25 Nov 2020.
    1. American Association for the Study of Liver Diseases, Infectious Diseases Society of America . Monitoring patients who are starting HCV treatment, are on treatment, or have completed therapy. HCV guidance: recommendations for testing, managing, and treating hepatitis C. Available at: . Accessed 25 Nov 2020.
    1. Karsdal MA, Daniels SJ, Holm Nielsen S, Bager C, Rasmussen DGK, Loomba R, et al. Collagen biology and non‐invasive biomarkers of liver fibrosis. Liver Int. 2020;40:736–50.
    1. Ito S, Nagata K. Roles of the endoplasmic reticulum‐resident, collagen‐specific molecular chaperone Hsp47 in vertebrate cells and human disease. J Biol Chem. 2019;294:2133–41.
    1. Roderfeld M. Matrix metalloproteinase functions in hepatic injury and fibrosis. Matrix Biol. 2018;68–69:452–62.
    1. Parsons CJ, Bradford BU, Pan CQ, Cheung E, Schauer M, Knorr A, et al. Antifibrotic effects of a tissue inhibitor of metalloproteinase‐1 antibody on established liver fibrosis in rats. Hepatology. 2004;40:1106–15.
    1. Sato Y, Murase K, Kato J, Kobune M, Sato T, Kawano Y, et al. Resolution of liver cirrhosis using vitamin A–coupled liposomes to deliver siRNA against a collagen‐specific chaperone. Nat Biotechnol. 2008;26:431–42.
    1. Lawitz E, Balabanska RI, Charles ED, Friedman SL, Gutierrez JA, Niitsu Y, et al. Clinical phase 1b/2 study results for safety, pharmacokinetics, and efficacy of ND‐L02‐s0201, a novel targeted lipid nanoparticle (LNP) delivering HSP47 siRNA for the treatment of patients with advanced liver fibrosis [Abstract]. Hepatology. 2017;66:237A.
    1. Soule B, Tirucherai G, Kavita U, Kundu S, Christian R. Safety, tolerability, and pharmacokinetics of BMS‐986263/NDL02‐s0201, a novel targeted lipid nanoparticle delivering HSP47 siRNA, in healthy participants: a randomised, placebo‐controlled, double‐blind, phase 1 study [Abstract]. J Hepatol. 2018;68:S112.
    1. Masuda H, Fukumoto M, Hirayoshi K, Nagata K. Coexpression of the collagen‐binding stress protein HSP47 gene and the alpha 1(I) and alpha 1(III) collagen genes in carbon tetrachloride‐induced rat liver fibrosis. J Clin Invest. 1994;94:2481–8.
    1. Razzaque MS, Hossain MA, Kohno S, Taguchi T. Bleomycin‐induced pulmonary fibrosis in rat is associated with increased expression of collagen‐binding heat shock protein (HSP) 47. Virchows Arch. 1998;432:455–60.
    1. Honzawa Y, Nakase H, Shiokawa M, Yoshino T, Imaeda H, Matsuura M, et al. Involvement of interleukin‐17A‐induced expression of heat shock protein 47 in intestinal fibrosis in Crohn's disease. Gut. 2014;63:1902–12.
    1. Friedman SL, Neuschwander‐Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med. 2018;24:908–22.
    1. Friedman SL. Targeting siRNA to arrest fibrosis. Nat Biotechnol. 2008;26:399–400.
    1. Tam C, Wong JH, Cheung RCF, Zuo T, Ng TB. Therapeutic potentials of short interfering RNAs. Appl Microbiol Biotechnol. 2017;101:7091–111.
    1. Arthur MJ. Reversibility of liver fibrosis and cirrhosis following treatment for hepatitis C. Gastroenterology. 2002;122:1525–8.
    1. Soule B, Charles ED, Tirucherai G, Roodt F, Kavita U, Revankar R, et al. Safety, pharmacokinetics, and immunogenicity of BMS‐986263/ND‐L02‐s0201 in healthy Japanese and non‐Japanese participants. [Abstract] Hepatology. 2018;68:739A.
    1. Sakamoto N, Ogawa K, Suda G, Morikawa K, Sho T, Nakai M, et al. Clinical phase 1b study results for safety, pharmacokinetics and efficacy of ND‐L02‐s0201, a novel targeted lipid nanoparticle delivering HSP47 SIRNA for the treatment of Japanese patients with advanced liver fibrosis. [Abstract] J Hepatol. 2018;68:S242.
    1. Wang Y, Vincent R, Yang J, Asgharpour A, Liang X, Idowu MO, et al. Dual‐photon microscopy‐based quantitation of fibrosis‐related parameters (q‐FP) to model disease progression in steatohepatitis. Hepatology. 2017;65:1891–903.
    1. Kavita U, Miller W, Ji QC, Pillutla RC. A fit‐for‐purpose method for the detection of human antibodies to surface‐exposed components of BMS‐986263, a lipid nanoparticle–based drug product containing a siRNA drug substance. AAPS J. 2019;21:92.
    1. Lee YA, Wallace MC, Friedman SL. Pathobiology of liver fibrosis: a translational success story. Gut. 2015;64:830–41.
    1. George SL, Bacon BR, Brunt EM, Mihindukulasuriya KL, Hoffmann J, Di Bisceglie AM. Clinical, virologic, histologic, and biochemical outcomes after successful HCV therapy: a 5‐year follow‐up of 150 patients. Hepatology. 2009;49:729–38.
    1. Ioannou GN, Beste LA, Green PK, Singal AG, Tapper EB, Waljee AK, et al. Increased risk for hepatocellular carcinoma persists up to 10 years after HCV eradication in patients with baseline cirrhosis or high FIB‐4 scores. Gastroenterology. 2019;157:1264–78.e64.
    1. Chen S‐H, Peng C‐Y, Chiang I‐P, Lai H‐C, Lee C‐J, Su W‐P, et al. Comparison of collagen proportionate areas in liver fibrosis quantification between chronic hepatitis B and C. Medicine (Baltimore). 2016;95:e4736.
    1. Harrison SA, Abdelmalek MF, Caldwell S, Shiffman ML, Diehl AM, Ghalib R, et al. Simtuzumab is ineffective for patients with bridging fibrosis or compensated cirrhosis caused by nonalcoholic steatohepatitis. Gastroenterology. 2018;155:1140–53.
    1. Pan JJ, Bao F, Du E, Skillin C, Frenette CT, Waalen J, et al. Morphometry confirms fibrosis regression from sustained virologic response to direct‐acting antivirals for hepatitis C. Hepatol Commun. 2018;2:1320–30.
    1. Daniels SJ, Erhardtsen E, Kehlet SN, Leeming DJ, Shevell D, Rasmussen DGK, et al. Establishing the stability, precision, potential influencing factors and reference ranges of a biomarker of liver fibrosis, PRO‐C3, in healthy subjects, obese, and NAFLD patients [Abstract]. Hepatology. 2020;72:911A.
    1. Tachi Y, Hirai T, Miyata A, Ohara K, Iida T, Ishizu Y, et al. Progressive fibrosis significantly correlates with hepatocellular carcinoma in patients with a sustained virological response. Hepatol Res. 2015;45:238–46.
    1. Kanwal F, Kramer J, Asch SM, Chayanupatkul M, Cao Y, El‐Serag HB. Risk of hepatocellular cancer in HCV patients treated with direct‐acting antiviral agents. Gastroenterology. 2017;153:996–1005.e1.

Source: PubMed

3
Předplatit