Clinical Study of Single-Stranded Oligonucleotide RO7062931 in Healthy Volunteers and Patients With Chronic Hepatitis B

Edward Gane, Man-Fung Yuen, Dong Joon Kim, Henry Lik-Yuen Chan, Bernadette Surujbally, Vedran Pavlovic, Sudip Das, Miriam Triyatni, Remi Kazma, Joseph F Grippo, Simon Buatois, Annabelle Lemenuel-Diot, Ben-Fillippo Krippendorff, Henrik Mueller, Yuchen Zhang, Hyung Joon Kim, Apinya Leerapun, Tien Huey Lim, Young-Suk Lim, Tawesak Tanwandee, Won Kim, Wendy Cheng, Tsung-Hui Hu, Cynthia Wat, Edward Gane, Man-Fung Yuen, Dong Joon Kim, Henry Lik-Yuen Chan, Bernadette Surujbally, Vedran Pavlovic, Sudip Das, Miriam Triyatni, Remi Kazma, Joseph F Grippo, Simon Buatois, Annabelle Lemenuel-Diot, Ben-Fillippo Krippendorff, Henrik Mueller, Yuchen Zhang, Hyung Joon Kim, Apinya Leerapun, Tien Huey Lim, Young-Suk Lim, Tawesak Tanwandee, Won Kim, Wendy Cheng, Tsung-Hui Hu, Cynthia Wat

Abstract

Background and aims: RO7062931 is an N-acetylgalactosamine (GalNAc)-conjugated single-stranded locked nucleic acid oligonucleotide complementary to HBV RNA. GalNAc conjugation targets the liver through the asialoglycoprotein receptor (ASGPR). This two-part phase 1 study evaluated the safety, pharmacokinetics, and pharmacodynamics of RO7062931 in healthy volunteers and patients with chronic hepatitis B (CHB) who were virologically suppressed.

Approach and results: Part 1 was a single ascending dose study in healthy volunteers randomized to receive a single RO7062931 dose (0.1-4.0 mg/kg), or placebo. Part 2 was a multiple ascending dose study in patients with CHB randomized to receive RO7062931 at 0.5, 1.5, or 3.0 mg/kg or placebo every month for a total of 2 doses (Part 2a) or RO7062931 at 3.0 mg/kg every 2 weeks, 3.0 mg/kg every week (QW), or 4.0 mg/kg QW or placebo for a total of 3-5 doses (Part 2b). Sixty healthy volunteers and 59 patients received RO7062931 or placebo. The majority of adverse events (AEs) reported were mild in intensity. Common AEs included self-limiting injection site reactions and influenza-like illness. Supradose-proportional increases in RO7062931 plasma exposure and urinary excretion occurred at doses ≥3.0 mg/kg. In patients with CHB, RO7062931 resulted in dose-dependent and time-dependent reduction in HBsAg versus placebo. The greatest HBsAg declines from baseline were achieved with the 3.0 mg/kg QW dose regimen (mean nadir ~0.5 log10 IU/mL) independent of HBeAg status.

Conclusions: RO7062931 is safe and well tolerated at doses up to 4.0 mg/kg QW. Supradose-proportional exposure at doses of 3.0-4.0 mg/kg was indicative of partial saturation of the ASGPR-mediated liver uptake system. Dose-dependent declines in HBsAg demonstrated target engagement with RO7062931.

Trial registration: ClinicalTrials.gov NCT03038113.

© 2021 Roche Products Ltd. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.

Figures

FIG. 1
FIG. 1
Study design. Part 1: single ascending dose cohorts in healthy volunteers. Parts 2a and 2b: multiple‐dose cohorts in patients with CHB.
FIG. 2
FIG. 2
Volunteer and patient flow through the study for (A) Part 1 single ascending dose in healthy volunteers and (B) Part 2 multiple dosing in patients with CHB.
FIG. 3
FIG. 3
Mean change from baseline in select (hepatic, hematologic, renal) safety laboratory parameters in patients with CHB receiving placebo or RO7062931: (A) ALT (U/L), (B) aspartate aminotransferase (U/L), (C) activated partial thromboplastin time (sec), (D) platelets (109/L), (E) creatinine (μmol/L), and (F) blood urea nitrogen (mmol/L). Abbreviations: Fu, follow‐up; Q1M, every 1 month.
FIG. 4
FIG. 4
Plasma and urine RO7072931 PK. Plasma concentration‐time profiles for RO7062931 over (A) 24 hours and (B) 168 hours after single doses (0.1‐4.0 mg/kg) in healthy volunteers; (C) mean plasma AUC0‐inf versus total milligram dose in healthy volunteers; the dashed line is a linear regression line of the observed AUC at the doses of 0.1, 0.3, and 1 mg/kg in HVs. The orange line is a loess regression line of the observed AUC in HV. (D) Mean % dose eliminated in urine in the first 8 hours postdose in healthy volunteers and patients with CHB. Error bars indicate standard deviation. Abbreviations: AUC0‐inf, area under concentration‐time curve extrapolated to infinity; HV, healthy volunteers.
FIG. 5
FIG. 5
Mean log10 (IU/mL) change in HBsAg from baseline in patients with CHB receiving placebo or (A) RO7062931 0.5‐3.0 mg/kg QM or (B) RO7062931 3.0 mg/kg QW or Q2W or 4.0 mg/kg QW. (C) RO7062931 3.0 mg/kg QW according to HBeAg status. Error bars indicate standard error; (D) maximum log10 change from baseline in HBsAg in patients with CHB receiving RO7062931 (0.5‐3.0 mg/kg QM; 3.0 mg/kg QW or Q2W, or 4.0 mg/kg QW) or placebo. Error bars indicate 95% confidence intervals.

References

    1. World Health Organization . Global hepatitis report 2017. . Accessed September 16, 2020.
    1. Yuen MF, Chen DS, Dusheiko GM, Janssen HL, Lau DT, Locarnini SA, et al. Hepatitis B virus infection. Nat Rev Dis Primer 2018;4:18035.
    1. Anderson RT, Choi HS, Lenz O, Peters MG, Janssen HL, Mishra P, et al. Association between seroclearance of hepatitis B surface antigen and long‐term clinical outcomes of patients with chronic hepatitis B virus infection: systematic review and meta‐analysis. Clin Gastroenterol Hepatol 2021;19:463‐472.
    1. European Association for the Study of the Liver . EASL 2017 Clinical Practice Guidelines on the management of hepatitis B virus infection. J Hepatol 2017;67:370‐398.
    1. Zoulim F, Durantel D. Antiviral therapies and prospects for a cure of chronic hepatitis B. Cold Spring Harb Perspect Med 2015;5:a021501.
    1. Lee HM, Banini BA. Updates on chronic HBV: current challenges and future goals. Curr Treat Options Gastroenterol 2019;17:271‐291.
    1. Lok AS, Zoulim F, Dusheiko G, Ghany MG. Hepatitis B cure: from discovery to regulatory approval. J Hepatol 2017;67:847‐861.
    1. Ye B, Liu X, Li X, Kong H, Tian L, Chen Y. T‐cell exhaustion in chronic hepatitis B infection: current knowledge and clinical significance. Cell Death Dis 2015;6:e1694.
    1. Seto WK, Lo YR, Pawlotsky JM, Yuen MF. Chronic hepatitis B virus infection. Lancet 2018;392:2313‐2324.
    1. Fisicaro P, Barili V, Rossi M, Montali I, Vecchi A, Acerbi G, et al. Pathogenetic mechanisms of T cell dysfunction in chronic HBV infection and related therapeutic approaches. Front Immunol 2020;11:849.
    1. Lebossé F, Testoni B, Fresquet J, Facchetti F, Galmozzi E, Fournier M, et al. Intrahepatic innate immune response pathways are downregulated in untreated chronic hepatitis B. J Hepatol 2017;66:897‐909.
    1. Kondo Y, Ninomiya M, Kakazu E, Kimura O, Shimosegawa T. Hepatitis B surface antigen could contribute to the immunopathogenesis of hepatitis B virus infection. ISRN Gastroenterol 2013;2013:935295.
    1. Hu B, Zhong L, Weng Y, Peng L, Huang Y, Zhao Y, et al. Therapeutic siRNA: state of the art. Signal Transduct Target Ther 2020;5:101.
    1. Javanbakht H, Mueller H, Walther J, Zhou X, Lopez A, Pattupara T, et al. Liver‐targeted anti‐HBV single‐stranded oligonucleotides with locked nucleic acid potently reduce HBV gene expression in vivo. Mol Ther Nucleic Acids 2018;11:441‐454.
    1. Schluep T, Lickliter J, Hamilton J, Lewis DL, Lai CL, Lau JY, et al. Safety, tolerability, and pharmacokinetics of ARC‐520 injection, an RNA interference‐based therapeutic for the treatment of chronic hepatitis B virus infection, in healthy volunteers. Clin Pharmacol Drug Dev 2017;6:350‐362.
    1. Thi EP, Dhillon AP, Ardzinski A, Bidirici‐Ertekin L, Cobarrubias KD, Cuconati A, et al. ARB‐1740, a RNA interference therapeutic for chronic hepatitis B infection. ACS Infect Dis 2019;5:725‐737.
    1. Yuen MF, Schiefke I, Yoon JH, Ahn SH, Heo J, Kim JH, et al. RNA interference therapy with ARC‐520 results in prolonged hepatitis B surface antigen response in patients with chronic hepatitis B infection. Hepatology 2020;72:19‐31.
    1. Gish RG, Yuen MF, Chan HL, Given BD, Lai CL, Locarnini SA, et al. Synthetic RNAi triggers and their use in chronic hepatitis B therapies with curative intent. Antiviral Res 2015;121:97‐108.
    1. Dicerna . Corporate overview: maximizing the impact of RNAi on medicine. . Accessed November 2, 2020.
    1. Gane E, Lim YS, Tangkijvanich P, O’Beirne J, Lim TH, Bakardjiev A, et al. Preliminary safety and antiviral activity of VIR‐2218, an X‐targeting HBV RNAi therapeutic, in chronic hepatitis B patients. J Hepatol 2020;73:S50‐S51.
    1. Arbutus Biopharma . AB‐729 (GalNAc‐RNAi). . Accessed November 2, 2020.
    1. Billioud G, Kruse RL, Carrillo M, Whitten‐Bauer C, Gao D, Kim A, et al. In vivo reduction of hepatitis B virus antigenemia and viremia by antisense oligonucleotides. J Hepatol 2016;64:781‐789.
    1. Yuen MF, Heo J, Jang JW, Yoon JH, Kweon YO, Park SJ, et al. Hepatitis B virus surface antigen inhibition with GSK3228836 (ISIS 505358) in chronic hepatitis B patients on stable nucleos(t)ide analogue regimen and in nucleos(t)ide analogue‐naive patients: a phase 2a, randomized, double‐blind, placebo‐controlled study. J Hepatol 2020;73:S49‐S50.
    1. Tanowitz M, Hettrick L, Revenko A, Kinberger GA, Prakash TP, Seth PP. Asialoglycoprotein receptor 1 mediates productive uptake of N‐acetylgalactosamine‐conjugated and unconjugated phosphorothioate antisense oligonucleotides into liver hepatocytes. Nucleic Acids Res 2017;45:12388‐12400.
    1. Huang Y. Preclinical and clinical advances of GalNAc‐decorated nucleic acid therapeutics. Mol Ther Nucleic Acids 2017;6:116‐132.
    1. Bon C, Hofer T, Bousquet‐Mélou A, Davies MR, Krippendorff BF. Capacity limits of asialoglycoprotein receptor‐mediated liver targeting. mAbs 2017;9:1360‐1369.
    1. Chang ML, Liaw YF. Hepatitis B flares in chronic hepatitis B: pathogenesis, natural course, and management. J Hepatol 2014;61:1407‐1417.
    1. Heo NY. Is alanine aminotransferase flare‐up in nucleos(t)ide analogue treatment of chronic hepatitis B a promising, rather than a devastating, sign? Clin Mol Hepatol 2017;23:125‐127.
    1. Liao H, Liu Y, Li X, Wang J, Chen X, Zou J, et al. Monitoring of serum HBV RNA, HBcrAg, HBsAg and anti‐HBc levels in patients during long‐term nucleoside/nucleotide analogue therapy. Antivir Ther 2019;24:105‐115.
    1. Rong F, Jie P, Qing X, Deming T, Min X, Junqi N, et al.; Chronic Hepatitis B Study Consortium . Combining hepatitis B virus RNA and hepatitis B core‐related antigen: guidance for safely stopping nucleos(t)ide analogues in hepatitis B e antigen‐positive patients with chronic hepatitis B. J Infect Dis 2020;222:611‐618.
    1. Crooke ST, Witztum JL, Bennett CF, Baker BF. RNA‐targeted therapeutics. Cell Metab 2018;27:714‐739.
    1. Gane EJ, Locarnini S, Lim TH, Strasser S, Sievert W, Cheng W, et al. Dose response with the RNA interference therapy JNJ‐3989 combined with nucleos(t)ide analogue treatment in expanded cohorts of patients with chronic hepatitis B. Hepatology 2019;70:434A (Abstract 696).
    1. Gane E, Locarnini S, Lim TH, Strasser S, Sievert W, Cheng W, et al. Short‐term treatment with RNA interference therapy, JNJ‐3989, results in sustained hepatitis B surface antigen supression in patients with chronic hepatitis B receiving nucleos(t)ide analogue treatment. J Hepatol 2020;73:S20 (Abstract GS10).
    1. Yuen MF, Heo J, Kumada H, Suzuki F, Suzuki Y, Xie Q, et al. Results after 12 weeks treatment of multiple doses of GSK3389404 in chronic hepatitis B (CHB) subjects on stable nucleos(t)ide therapy in a phase 2a double‐blind, placebo‐controlled study. Hepatology 2019;70:433A (Abstract 695).
    1. Wooddell CI, Yuen MF, Chan HY, Gish RG, Locarnini SA, Chavez D, et al. RNAi‐based treatment of chronically infected patients and chimpanzees reveals that integrated hepatitis B virus DNA is a source of HBsAg. Sci Transl Med 2017;9:eaan0241.
    1. Geary RS. Antisense oligonucleotide pharmacokinetics and metabolism. Expert Opin Drug Metab Toxicol 2009;5:381‐391.
    1. Wang Y, Yu RZ, Henry S, Geary RS. Pharmacokinetics and clinical pharmacology considerations of GalNAc3‐conjugated antisense oligonucleotides. Expert Opin Drug Metab Toxicol 2019;15:475‐485.

Source: PubMed

3
Abonnieren