Clinical and Preclinical Single-Dose Pharmacokinetics of VIR-2218, an RNAi Therapeutic Targeting HBV Infection

Sneha V Gupta, Marie C Fanget, Christopher MacLauchlin, Valerie A Clausen, Jing Li, Daniel Cloutier, Ling Shen, Gabriel J Robbie, Erik Mogalian, Sneha V Gupta, Marie C Fanget, Christopher MacLauchlin, Valerie A Clausen, Jing Li, Daniel Cloutier, Ling Shen, Gabriel J Robbie, Erik Mogalian

Abstract

Background and objective: VIR-2218 is an investigational N-acetylgalactosamine-conjugated RNA interference therapeutic in development for chronic hepatitis B virus (HBV) infection. VIR-2218 was designed to silence HBV transcripts across all genotypes and uses Enhanced Stabilization Chemistry Plus (ESC+) technology. This study was designed to evaluate the single-dose pharmacokinetics of VIR-2218 in preclinical species and healthy volunteers.

Methods: Preclinically, a single subcutaneous dose of VIR-2218 (10 mg/kg) was administered to rats and nonhuman primates (NHPs), and the pharmacokinetics were assessed in plasma, urine, and liver using standard noncompartmental analysis (NCA) methods. Clinically, healthy volunteers were randomized (6:2 active:placebo) to receive a single subcutaneous dose of VIR-2218 (50-900 mg) or placebo. Pharmacokinetics were similarly assessed within human plasma and urine using NCA methods.

Results: In rats and NHPs, VIR-2218 was stable in plasma and was converted to AS(N-1)3'VIR-2218, the most prominent circulating metabolite, at < 10% plasma exposure compared with parent. VIR-2218 rapidly distributed to the liver, reaching peak liver concentrations within 7 and 24 h in rats and NHPs, respectively. In humans, VIR-2218 was rapidly absorbed, with a median time to peak plasma concentration (tmax) of 4-7 h, and had a short median plasma half-life of 2-5 h. Plasma exposures for area under the plasma concentration-time curve up to 12 h (AUC0-12) and mean maximum concentrations (Cmax) increased in a slightly greater-than-dose-proportional manner across the dose range studied. Interindividual pharmacokinetic variability was low to moderate, with a percent coefficient of variation of < 32% for AUC and < 43% for Cmax. A portion of VIR-2218 was converted to an active metabolite, AS(N-1)3'VIR-2218, with a median tmax of 6-10 h, both of which declined below the lower limit of quantification in plasma within 48 h. The pharmacokinetic profile of AS(N-1)3'VIR-2218 was similar to that of VIR-2218, with plasma AUC0-12 and Cmax values ≤ 12% of VIR-2218. VIR-2218 and AS(N-1)3'VIR-2218 were detectable in urine through the last measured time point, with approximately 17-48% of the administered dose recovered in urine as unchanged VIR-2218 over 0-24 h postdose. Based on pharmacokinetics in preclinical species, VIR-2218 localizes to the liver and likely exhibits prolonged hepatic exposure. Overall, no severe or serious adverse events or discontinuations due to adverse events were observed within the dose range evaluated for VIR-2218 in healthy volunteers (Vir Biotechnology, Inc., unpublished data).

Conclusions: VIR-2218 showed favorable pharmacokinetics in healthy volunteers supportive of subcutaneous dosing and continued development in patients with chronic HBV infection.

Clinical trial registration no: NCT03672188, September 14, 2018.

Trial registration: ClinicalTrials.gov NCT03672188 NCT00657007 NCT00071487 NCT01345253 NCT01516450 NCT00410384 NCT00424476 NCT02880852 NCT01583530.

Conflict of interest statement

Sneha V. Gupta, Marie C. Fanget, Daniel Cloutier, Ling Shen, and Erik Mogalian are or were employees of Vir Biotechnology, Inc. and may hold Vir stocks or options. Christopher MacLauchlin, Valerie A. Clausen, Jing Li, and Gabriel J. Robbie are or were employees of Alnylam Pharmaceuticals, Inc. and may hold Alnylam stocks or options.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Study design for VIR-2218 administration in healthy volunteers. Phase I randomized, double-blind, placebo-controlled, single-ascending-dose study in healthy adults who received a single ascending dose of subcutaneous VIR-2218 or placebo. PK, pharmacokinetic; SC, subcutaneous
Fig. 2
Fig. 2
Plasma concentration vs. time profiles for VIR-2218 and AS(N-1)3’VIR-2218 after a single subcutaneous dose in healthy volunteers. For plotting purposes, BLQ values in subjects were included as 0. Subjects with all BLQ values were excluded. BLQ, below limit of quantitation; SD, standard deviation
Fig. 3
Fig. 3
Plasma VIR-2218 dose proportionality from 50 to 900 mg after single subcutaneous VIR-2218 administration. VIR-2218 dose vs. a AUC0–12, b AUC∞, and cCmax on a logarithmic scale. The estimated slope (β) or proportionality coefficients were 1.19 (90% CI 1.10–1.28), 1.24 (90% CI 1.16–1.31), and 1.16 (90% CI 1.06–1.26) for AUC0–12, AUC∞, and Cmax, respectively. Crosses denote individual pharmacokinetic parameters, and the estimated slope is the proportionality coefficient. The shaded area represents the 90% CIs around the regression line. AUC0–12h, area under the plasma concentration–time curve from time 0 to 12 h; AUC∞, AUC extrapolated to infinity; CI, confidence interval; Cmax, maximum concentration; PK, pharmacokinetics
Fig. 4
Fig. 4
Urine concentration vs. time profiles for VIR-2218 and AS(N-1)3’VIR-2218. For plotting purposes, the BLQ value in subjects was included as 0. Subjects with all BLQ values were excluded. BLQ, below limit of quantitation; SD, standard deviation

References

    1. Trepo C. A brief history of hepatitis milestones. Liver Int. 2014;34(Suppl 1):29–37. doi: 10.1111/liv.12409.
    1. Schweitzer A, Horn J, Mikolajczyk RT, Krause G, Ott JJ. Estimations of worldwide prevalence of chronic hepatitis B virus infection: a systematic review of data published between 1965 and 2013. Lancet. 2015;386(10003):1546–1555. doi: 10.1016/S0140-6736(15)61412-X.
    1. Stanaway JD, Flaxman AD, Naghavi M, Fitzmaurice C, Vos T, Abubakar I, et al. The global burden of viral hepatitis from 1990 to 2013: findings from the Global Burden of Disease Study 2013. Lancet. 2016;388(10049):1081–1088. doi: 10.1016/S0140-6736(16)30579-7.
    1. Nelson NP, Easterbrook PJ, McMahon BJ. Epidemiology of hepatitis B virus Infection and impact of vaccination on disease. Clin Liver Dis. 2016;20(4):607–628. doi: 10.1016/j.cld.2016.06.006.
    1. Kramvis A. Genotypes and genetic variability of hepatitis B virus. Intervirology. 2014;57(3–4):141–150. doi: 10.1159/000360947.
    1. Protzer U, Maini MK, Knolle PA. Living in the liver: hepatic infections. Nat Rev Immunol. 2012;12(3):201–213. doi: 10.1038/nri3169.
    1. Liang TJ, Block TM, McMahon BJ, Ghany MG, Urban S, Guo JT, et al. Present and future therapies of hepatitis B: from discovery to cure. Hepatology. 2015;62(6):1893–1908. doi: 10.1002/hep.28025.
    1. Scaglione SJ, Lok AS. Effectiveness of hepatitis B treatment in clinical practice. Gastroenterology. 2012;142(6):1360–1368. doi: 10.1053/j.gastro.2012.01.044.
    1. Konerman MA, Lok AS. Interferon treatment for hepatitis B. Clin Liver Dis. 2016;20(4):645–665. doi: 10.1016/j.cld.2016.06.002.
    1. Agrawal N, Dasaradhi PV, Mohmmed A, Malhotra P, Bhatnagar RK, Mukherjee SK. RNA interference: biology, mechanism, and applications. Microbiol Mol Biol Rev. 2003;67(4):657–685. doi: 10.1128/mmbr.67.4.657-685.2003.
    1. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806–811. doi: 10.1038/35888.
    1. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M, et al. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature. 2004;432(7014):173–178. doi: 10.1038/nature03121.
    1. Nair JK, Attarwala H, Sehgal A, Wang Q, Aluri K, Zhang X, et al. Impact of enhanced metabolic stability on pharmacokinetics and pharmacodynamics of GalNAc-siRNA conjugates. Nucleic Acids Res. 2017;45(19):10969–10977. doi: 10.1093/nar/gkx818.
    1. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, et al. Multivalent N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. 2014;136(49):16958–16961. doi: 10.1021/ja505986a.
    1. Agarwal S, Simon AR, Goel V, Habtemariam BA, Clausen VA, Kim JB, et al. Pharmacokinetics and pharmacodynamics of the small interfering ribonucleic acid, givosiran, in patients with acute hepatic porphyria. Clin Pharmacol Ther. 2020;108(1):63–72. doi: 10.1002/cpt.1802.
    1. Habtemariam BA, Karsten V, Attarwala H, Goel V, Melch M, Clausen VA, et al. Single-dose pharmacokinetics and pharmacodynamics of transthyretin targeting N-acetylgalactosamine-small interfering ribonucleic acid conjugate, vutrisiran, in healthy subjects. Clin Pharmacol Ther. 2021;109(2):372–382. doi: 10.1002/cpt.1974.
    1. Schwartz AL, Rup D, Lodish HF. Difficulties in the quantification of asialoglycoprotein receptors on the rat hepatocyte. J Biol Chem. 1980;255(19):9033–9036. doi: 10.1016/S0021-9258(19)70522-5.
    1. Willoughby JLS, Chan A, Sehgal A, Butler JS, Nair JK, Racie T, et al. Evaluation of galNAc-siRNA conjugate activity in pre-clinical animal models with reduced asialoglycoprotein receptor expression. Mol Ther. 2018;26(1):105–114. doi: 10.1016/j.ymthe.2017.08.019.
    1. Smith BP, Vandenhende FR, DeSante KA, Farid NA, Welch PA, Callaghan JT, et al. Confidence interval criteria for assessment of dose proportionality. Pharm Res. 2000;17(10):1278–1283. doi: 10.1023/a:1026451721686.
    1. Manoharan M. RNA interference and chemically modified small interfering RNAs. Curr Opin Chem Biol. 2004;8(6):570–579. doi: 10.1016/j.cbpa.2004.10.007.
    1. Badri P, Jiang X, Borodovsky A, Najafian N, Kim J, Clausen VA, et al. Pharmacokinetic and pharmacodynamic properties of cemdisiran, an RNAi therapeutic targeting complement component 5, in healthy subjects and Patients with Paroxysmal Nocturnal Hemoglobinuria. Clin Pharmacokinet. 2020 doi: 10.1007/s40262-020-00940-9.

Source: PubMed

3
Abonneren