Safety, tolerability, and pharmacokinetics of AL-335 in healthy volunteers and hepatitis C virus-infected subjects

Matthew W McClure, Elina Berliba, Tengiz Tsertsvadze, Adrian Streinu-Cercel, Leen Vijgen, Béatrice Astruc, Alain Patat, Christopher Westland, Sushmita Chanda, Qingling Zhang, Thomas N Kakuda, Jennifer Vuong, Nick Khorlin, Leonid Beigelman, Lawrence M Blatt, John Fry, Matthew W McClure, Elina Berliba, Tengiz Tsertsvadze, Adrian Streinu-Cercel, Leen Vijgen, Béatrice Astruc, Alain Patat, Christopher Westland, Sushmita Chanda, Qingling Zhang, Thomas N Kakuda, Jennifer Vuong, Nick Khorlin, Leonid Beigelman, Lawrence M Blatt, John Fry

Abstract

Background: The nucleotide analog AL-335 is a pangenotypic hepatitis C virus (HCV) nonstructural protein (NS)5B inhibitor being evaluated as treatment for chronic HCV infection.

Methods: This three-part randomized, double-blind study evaluated the safety and pharmacokinetics of single and multiple ascending oral doses of AL-335. Healthy volunteers (HVs) received single doses of AL-335 (100-1,200 mg) or placebo in a fasted or fed (400 mg) state. Non-cirrhotic subjects (HCV genotype [GT]1-4) and GT1-infected subjects with Child Pugh A cirrhosis received multiple doses of AL-335 (400, 800, 1,200 mg) or placebo once daily (QD) for 7 days.

Results: Forty-eight HVs and 64 subjects with HCV GT1-4 were randomized and received treatment. AL-335 was well tolerated in HVs and HCV-infected subjects with/without cirrhosis. AL-335 was rapidly absorbed and converted to the metabolites ALS-022399 and ALS-022227. ALS-022227 exposure increased less than dose-proportionally and was unaffected by food, while AL-335 and ALS-022399 exposure increased with food by 85% and 50%, respectively, in HVs. Rapid and dose-dependent reductions in HCV-RNA were observed in GT1-infected subjects. In non-cirrhotic, GT1-4-infected subjects receiving AL-335 800 mg QD, potent antiviral activity was observed, regardless of genotype (mean maximum reductions in HCV-RNA of 4.0-4.8 log10 IU/mL). The same dose in GT1-infected cirrhotic subjects resulted in a 3.5 log10 IU/mL mean maximum reduction in HCV-RNA.

Conclusions: AL-335 was well tolerated when administered as single and multiple doses, with an acceptable pharmacokinetic profile. The drug also demonstrated potent antiviral activity in HCV GT1-4-infected subjects, including GT1-infected subjects with cirrhosis.

Trial registration: ClinicalTrials.gov NCT02339207.

Conflict of interest statement

The authors have read the journal's policy and the authors of this manuscript have the following competing interests: MWM and LB were employees of Alios BioPharma Inc., part of the Janssen Pharmaceutical Companies, at the time of the study. EB has been the Principal Investigator in clinical trials on HCV sponsored by Alios BioPharma Inc., part of the Janssen Pharmaceutical Companies, Atea Pharmaceuticals, and Janssen. TT has a project-based contractual relationship with ARENSIA. AS-C has been the Principal Investigator in clinical trials on HCV sponsored by AbbVie, Alios BioPharma Inc., part of the Janssen Pharmaceutical Companies, Boehringer Ingelheim, Janssen, and Merck Sharp & Dohme. LV is an employee of Janssen and a shareholder of Johnson & Johnson. BA and AP are employees of Biotrial. CW, TNK, JV, and NK are employees of Alios BioPharma Inc., part of the Janssen Pharmaceutical Companies, and are shareholders of Johnson & Johnson. SC, QZ, LMB, and JF were employees of Alios BioPharma Inc., part of the Janssen Pharmaceutical Companies, at the time of the study, and are shareholders of Johnson & Johnson. There are no patents, products in development or marketed products associated with this research to declare. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1
Fig 1
(A) Study design and (B) subject disposition.aAll randomized subjects in the GT4–6 cohort were infected with GT4; b8/48 subjects (from AL-335 400 mg tablet cohort) were enrolled in Part I and then re-dosed in fed state. ECG = electrocardiogram, fasted = in fasted state, fed = in fed state, GT = genotype, I/E = inclusion/exclusion, MAD = multiple ascending doses, QD = once daily, SAD = single ascending doses.
Fig 2
Fig 2
(A) Mean plasma concentration of ALS-022227 after SAD in Part I/II (top) and after MAD in Part III on Days 1 and 7 (bottom) in healthy volunteers; (B) Mean plasma concentration of AL-335, ALS-022399, and ALS-022227 after multiple doses of AL-335 administered as 800 mg QD in HCV GT1-infected subjects without cirrhosis (Part III, cohort 2). fed = in fed state, GT = genotype, HCV = hepatitis C virus, MAD = multiple ascending doses, QD = once daily, SAD = single ascending doses.
Fig 3. Mean (±SEM) HCV-RNA change from…
Fig 3. Mean (±SEM) HCV-RNA change from baseline (log10 IU/mL) over time following multiple oral administrations of AL-335 (tablet form) under fed conditions (Part III, Days 1–7; pharmacodynamic set).
Values below the LLOQ (aBaseline is defined as the average of Day -2 and Day 1 pre-dose values; bAll randomized subjects in the GT4–6 cohort were infected with GT4. GT = genotype, HCV = hepatitis C virus, LLOQ = lower limit of quantification, MAD = multiple ascending doses, QD = once daily, RNA = ribonucleic acid, SEM = standard error of the mean.
Fig 4. Mean maximal change from baseline…
Fig 4. Mean maximal change from baseline in HCV-RNA per cohort between Day 1 and Day 10 in Part III.
aAll randomized subjects in the HCV GT4–6 cohort were infected with GT4. Baseline is defined as the average of Day -2 and Day 1 pre-dose values. comp = compensated, GT = genotype, HCV = hepatitis C virus, RNA = ribonucleic acid, SD = standard deviation.

References

    1. World Health Organization. Hepatitis C–Fact Sheet 164. 2017. Available from:
    1. European Association for the Study of the Liver. EASL recommendations on treatment of hepatitis C 2016. J Hepatol. 2017;66(1):153–94. 10.1016/j.jhep.2016.09.001
    1. American Association for the Study of Liver Diseases, Infectious Diseases Society of America. HCV guidance: recommendations for testing, managing and treating hepatitis C. 2017. Available from:
    1. Bang CS, Song IH. Impact of antiviral therapy on hepatocellular carcinoma and mortality in patients with chronic hepatitis C: systematic review and meta-analysis. BMC Gastroenterol. 2017;17(1):46 10.1186/s12876-017-0606-9
    1. Thrift AP, El-Serag HS, Kanwal F. Global epidemiology and burden of HCV infection and HCV-related disease. Nat Rev Gastroenterol Hepatol. 2017;14(2):122–32. 10.1038/nrgastro.2016.176
    1. Falade-Nwulia O, Suarez-Cuervo C, Nelson DR, Fried MW, Segal JB, Sulkowski MS. Oral direct-acting agent therapy for hepatitis C virus infection: A systematic review. Ann Intern Med. 2017;166(9):637–48. 10.7326/M16-2575
    1. Bidell MR, McLaughlin M, Faragon J, Morse C, Patel N. Desirable characteristics of hepatitis C treatment regimens: A review of what we have and what we need. Infect Dis Ther. 2016;5(3):299–312. 10.1007/s40121-016-0118-x
    1. Bobeck DR, Schinazi RF, Coats SJ. Advances in nucleoside monophosphate prodrugs as anti-HCV agents. Antivir Ther. 2010;15(7):935–50. 10.3851/IMP1667
    1. Coats SJ, Garnier-Amblard EC, Amblard F, Ehteshami M, Amiralaei S, Zhang H, et al. Chutes and ladders in hepatitis C nucleoside drug development. Antiviral Res. 2014;102:119–47. 10.1016/j.antiviral.2013.11.008
    1. Jordheim LP, Durantel D, Zoulim F, Dumontet C. Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat Rev Drug Discov. 2013;12(6):447–64. 10.1038/nrd4010
    1. Tan H, Shaw K, Jekel A, Deval J, Jin Z, Fung A, et al. Preclinical characterization of AL-335, a potent uridine based nucleoside polymerase inhibitor for the treatment of chronic hepatitis C. J Hepatol. 2015;S577–S8.
    1. Vijgen L, Fevery B, Jekle A, Shaw K, Tan H, Lenz O, et al. In vitro virology profile of the 3 direct-acting antiviral combination of AL-335, odalasvir and simeprevir. J Hepatol. 2017;S533–S4.
    1. Lund KC, Peterson LL, Wallace KB. Absence of a universal mechanism of mitochondrial toxicity by nucleoside analogs. Antimicrob Agents Chemother. 2007;51(7):2531–9. 10.1128/AAC.00039-07
    1. Food and Drug Administration. Guidance for industry: Food-effect bioavailability and fed bioequivalence studies. 2002. Available from:
    1. Venuto CS, Talal AH. Intrahepatic sampling for the elucidation of antiviral clinical pharmacology. Clin Pharmacol Drug Dev. 2017;6(2):169–75. 10.1002/cpdd.311
    1. Lawitz EJ, Rodriguez-Torres M, Denning J, Mathias A, Mo H, Gao B, et al. All-oral therapy with nucleotide inhibitors sofosbuvir and GS-0938 for 14 days in treatment-naive genotype 1 hepatitis C (nuclear). J Viral Hepat. 2013;20(10):699–707. 10.1111/jvh.12091
    1. Karchava M, Waldenstrom J, Parker M, Hallack R, Sharvadze L, Gatserelia L, et al. High incidence of the hepatitis C virus recombinant 2k/1b in Georgia: Recommendations for testing and treatment. Hepatol Res. 2015;45(13):1292–8. 10.1111/hepr.12505
    1. Kalinina O, Norder H, Mukomolov S, Magnius LO. A natural intergenotypic recombinant of hepatitis C virus identified in St. Petersburg. J Virol. 2002;76(8):4034–43. 10.1128/JVI.76.8.4034-4043.2002
    1. Svarovskaia ES, Dvory-Sobol H, Parkin N, Hebner C, Gontcharova V, Martin R, et al. Infrequent development of resistance in genotype 1–6 hepatitis C virus-infected subjects treated with sofosbuvir in phase 2 and 3 clinical trials. Clin Infect Dis. 2014;59(12):1666–74. 10.1093/cid/ciu697

Source: PubMed

3
订阅