Elsevier

European Journal of Medicinal Chemistry

Volume 164, 15 February 2019, Pages 179-192
European Journal of Medicinal Chemistry

Research paper
5-Aminothiophene-2,4-dicarboxamide analogues as hepatitis B virus capsid assembly effectors

https://doi.org/10.1016/j.ejmech.2018.12.047Get rights and content

Highlights

  • Validation and optimization of an ATDC analogue as a CAE hit.

  • SAR from a μM hit led to 6 CAEs active at nM.

  • SPR led to improved metabolic stability and oral bioavailability.

  • Molecular modeling study revealed a binding mode.

  • The best CAEs from this series are better than reported CAEs.

Abstract

Chronic hepatitis B virus (HBV) infection represents a major health threat. Current FDA-approved drugs do not cure HBV. Targeting HBV core protein (Cp) provides an attractive approach toward HBV inhibition and possibly infection cure. We have previously identified and characterized a 5-amino-3-methylthiophene-2,4-dicarboxamide (ATDC) compound as a structurally novel hit for capsid assembly effectors (CAEs). We report herein hit validation through studies on absorption, distribution, metabolism and excretion (ADME) properties and pharmacokinetics (PK), and hit optimization via analogue synthesis aiming to probe the structure-activity relationship (SAR) and structure-property relationship (SPR). In the end, these medicinal chemistry efforts led to the identification of multiple analogues strongly binding to Cp, potently inhibiting HBV replication in nanomolar range without cytotoxicity, and exhibiting good oral bioavailability (F). Two of our analogues, 19o (EC50 = 0.11 μM, CC50 > 100 μM, F = 25%) and 19k (EC50 = 0.31 μM, CC50 > 100 μM, F = 46%), displayed overall lead profiles superior to reported CAEs 710 used in our studies.

Introduction

HBV chronically infects an estimated 250 million people worldwide and remains a major health threat [1]. Despite a successful vaccine, there are still around 1 million people newly infected each year. Chronic HBV infection is often associated with severe liver diseases which typically progress through fibrosis, cirrhosis and eventually develop into hepatocellular carcinoma (HCC) [2]. It is estimated that HBV-associated liver diseases result in approximately 660,000 deaths annually. Current FDA-approved HBV drugs include the immunomodulatory agent pegylated interferon alpha (IFN-α) [3] and direct acting nucleos(t)ide analogues (NAs) which target reverse transcriptase (RT) of the HBV P protein complex [4]. These NAs are based on all four endogenous nucleosides with distinct approaches with respect to mimicking the ribose moiety (Fig. 1), including β-L-nucleosides [5,6] lamivudine (1, 3 TC, analogue of dC) and telbivudine (2, LdT, analogue of T), the carbocyclic nucleoside entecavir (3, ETV, analogue of dG) [7], the acyclic nucleoside phosphonate (ANPs) [8] adefovir (4, ADV, analogue of dA) and two forms of tenofovir (TFV, analogue of dA): tenofovir disoproxil fumarate (5, TDF) and tenofovir alafenamide (6, TAF). While NAs are generally well tolerated [9] and largely effective [10,11] in suppressing viral load, they do not cure HBV infection presumably because they do not eliminate HBV covalently closed circular DNA (cccDNA) which is the main viral reservoir for persistent infection [12]. HBV cure calls for novel antiviral approaches to completely eliminate or functionally inactivate cccDNA [[13], [14], [15]]. Otherwise, clinical management of chronic HBV infection requires lifetime treatment with current drugs.

The HBV replication cycle [16] entails a critical step where Cp dimers are assembled into functional viral capsids which encapsulate both viral pregenomic RNA (pgRNA) and polymerase complex for active reverse transcription to generate genomic, partially double-stranded relaxed circular DNA (RC-DNA). Interestingly, RC-DNA-containing mature nucleocapsids are either enveloped then secreted as new infectious virions, or enter the nucleus to replenish the cccDNA pool, constituting the intracellular amplification pathway of cccDNA. Disrupting the proper assembly of capsid can therefore inhibit HBV by blocking the production of infectious viruses and depleting cccDNA replenishment, and potentially contribute to HBV cure [17]. Therefore, targeting HBV Cp represents an attractive approach for treating chronic HBV infection. A few chemotypes have been reported as potent CAEs (Fig. 2) [14,18,19]. Amongst these, the heteroaryldihydropyrimidine (HAP) [[20], [21], [22], [23], [24]] chemotype as represented by compound 7 and the sulfamoylbenzamide (SBA) chemotype [25,26] as represented by compound 8 are particularly well studied with analogues in clinical development [[27], [28], [29]]. Mechanistically, all known CAEs accelerate capsid assembly, albeit with different assembled products. The HAP series constitutes a class of its own by inducing misassembly to form aberrant nonfunctional capsid particles, whereas SBA and other chemotypes, such as phenylpropenamide (9) [30] and NZ-4 (10) [31], promote the formation of empty, morphologically normal capsids [32].

Recently we conducted a high-throughput screening (HTS) of commercial libraries in a thermal shift assay (TSA) that measures binding to HBV Cp and identified compound 11 as a strong binder (Fig. 3) [33]. In the subsequent antiviral assay 11 inhibited HBV total DNA production (EC50 = 3.4 μM) with no cytotoxicity observed (CC50 > 100 μM). Mechanistically, compound 11 promoted the formation of large Cp aggregates and prevented Cp from entering nucleus [33]. These results confirmed 11 as a valid CAE hit. We report herein the optimization of CAE hit 11 through analogue synthesis, SAR as well as ADME and PK studies.

Section snippets

Results and discussion

In the present work, compound 11 was first evaluated in various in vitro assays for its physicochemical and ADME properties, including aqueous solubility and stability, plasma stability, plasma protein binding and microsomal stability. Overall, compound 11 exhibited favorable ADME properties (Fig. 3) predicting good oral bioavailability. This was confirmed through in vivo PK studies in mice from which the oral bioavailability of 11 was determined to be 31%. In addition, compound 11 has

Chemistry

General Procedures. All commercial chemicals were used as supplied unless otherwise indicated. Dry solvents were either purchased (toluene and dioxane, EtOH) or dispensed under argon from an anhydrous solvent system with two packed columns of neutral alumina or molecular sieves. Flash chromatography was performed on a Teledyne Combiflash RF-200 with RediSep columns (silica) and indicated mobile phase. All moisture sensitive reactions were performed under an inert atmosphere of ultra-pure argon

Reagents

Biologicals. HepG2 (ATCC) cells were maintained in complete media [Dulbecco's Modified Eagle Medium (DMEM), 10% fetal bovine serum (FBS)]. HepAD38 cells (ATCC) [48] were maintained in tet media [complete media plus 0.4 μg/mL tetracycline (tet) and 400 μg/mL G418 (Gibco)]. All cells were incubated at 37 °C with 5% CO2.

Chemicals. The Maybridge Hitfinder chemical library of compounds (version 6) was purchased from Maybridge.

Cp purification

A gBlock Gene Fragment coding for the 149 amino acid assembly domain of

Acknowledgements

This research was supported by the National Institutes of Health (R01AI121315 to SGS and ZW).

References (63)

  • M.C. Lo et al.

    Evaluation of fluorescence-based thermal shift assays for hit identification in drug discovery

    Anal. Biochem.

    (2004)
  • E. Baka et al.

    Study of equilibrium solubility measurement by saturation shake-flask method using hydrochlorothiazide as model compound

    J. Pharmaceut. Biomed. Anal.

    (2008)
  • N.J. Waters et al.

    Validation of a Rapid equilibrium dialysis approach for the measurement of plasma protein binding

    J. Pharmacol. Sci.

    (2008)
  • Global Hepatitis Report

    (2017)
  • S.L. Chan et al.

    Infection and cancer: the case of hepatitis B

    J. Clin. Oncol.

    (2016)
  • Y.J. Tian et al.

    Effects of interferon-alpha/beta on HBV replication determined by viral load

    PLoS Pathog.

    (2011)
  • L. Kang et al.

    Anti-hbv drugs: progress, unmet needs, and new hope

    Viruses

    (2015)
  • Y.C. Cheng

    L-Nucleoside Analogues as a novel class of HBV drugs

    Antivir. Ther.

    (2002)
  • P.A. Furman et al.

    The effect of absolute-configuration on the anti-hiv and anti-hbv activity of nucleoside analogs

    Antiviral Chem. Chemother.

    (1995)
  • H. Tang et al.

    The discovery and development of a potent antiviral drug, entecavir, for the treatment of chronic hepatitis B

    J. Clin. Transl. Hepatol.

    (2013)
  • C. Ying et al.

    Novel acyclic nucleoside phosphonate analogues as potent and selective anti-HBV agents

    Antivir. Res.

    (2004)
  • P. Lampertico et al.

    Review article: long-term safety of nucleoside and nucleotide analogues in HBV-monoinfected patients

    Aliment. Pharmacol. Ther.

    (2016)
  • S. Piano et al.

    The impact of nucleoside/nucleotide analogues on the natural history of patients with hbv-related cirrhosis: a prospective comparative study towards patients with hcv-related cirrhosis in the interferon and ribavirin era

    Dig. Liver Dis.

    (2014)
  • K. Ma et al.

    Nucleoside analogs prevent disease progression in HBV-related acute-on-chronic liver failure: validation of the TPPM model

    Hepatology

    (2013)
  • M. Nassal

    HBV cccDNA: viral persistence reservoir and key obstacle for a cure of chronic hepatitis B

    Gut

    (2015)
  • L. Tang et al.

    The current status and future directions of hepatitis B antiviral drug discovery

    Expert Opin. Drug Discov.

    (2017)
  • Y.M. Pei et al.

    Past, current, and future developments of therapeutic agents for treatment of chronic hepatitis B virus infection

    J. Med. Chem.

    (2017)
  • S. Boucle et al.

    Toward elimination of hepatitis B virus using novel drugs, approaches, and combined modalities

    Clin. Liver Dis.

    (2016)
  • X.L. Li et al.

    Design, synthesis, and evaluation of tetrahydropyrrolo[1,2-c]pyrimidines as capsid assembly inhibitors for HBV treatment

    ACS Med. Chem. Lett.

    (2017)
  • N. Mani et al.

    Antiviral characterization of a next generation chemical series of HBV capsid inhibitors in vitro and in vivo

    Hepatology

    (2017)
  • L. Belloni et al.

    HAPs hepatitis B virus (HBV) capsid inhibitors affect core protein interaction with the minichromosome and target cccDNA function

    J. Hepatol.

    (2013)
  • Cited by (0)

    1

    These authors contributed equally.

    View full text