Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Antiviral Res ; 147: 149-158, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29037976

ABSTRACT

Dengue is the most important mosquito-transmitted viral disease and a major global health concern. Over the last decade, dengue virus (DENV) drug discovery and development has intensified, however, this has not resulted in approved DENV-specific antiviral treatments yet. DENV and hepatitis C virus (HCV) belong to the same Flaviviridae family and, in contrast to DENV, antiviral treatments for HCV have been licensed. Therefore, applying the knowledge gained on anti-HCV drugs may foster the discovery and development of dengue antiviral drugs. Here, we screened a library of compounds with established anti-HCV activity in a DENV-2 sub-genomic replicon inhibition assay and selected compounds with single-digit micromolar activity. These compounds were advanced into a hit-to-lead medicinal chemistry program resulting in lead compound JNJ-1A, which inhibited the DENV-2 sub-genomic replicon at 0.7 µM, in the absence of cytotoxicity. In addition, JNJ-1A showed equipotent antiviral activity against DENV serotypes 1, 2, and 4. In vitro resistance selection experiments with JNJ-1A induced mutation T108I in non-structural protein 4B (NS4B), pointing towards a mechanism of action linked to this protein. Collectively, we described the discovery and characterization of a novel DENV inhibitor potentially targeting NS4B.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Drug Resistance, Viral/genetics , Viral Nonstructural Proteins/genetics , Virus Replication/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/toxicity , Cell Line, Tumor , Chlorocebus aethiops , Dengue , Dengue Virus/genetics , Dengue Virus/physiology , Drug Discovery , Drug Resistance, Viral/drug effects , Hepacivirus/genetics , Humans , Mutation , RNA, Viral/genetics , Replicon/drug effects , Sequence Analysis, RNA , Small Molecule Libraries , Vero Cells
2.
Mol Divers ; 21(2): 475-481, 2017 May.
Article in English | MEDLINE | ID: mdl-28293834

ABSTRACT

Direct acting antiviral agents to cure hepatitis C virus (HCV) infection has emerged as the gold standard therapy. Along with protease inhibitors, nucleoside polymerase inhibitors and non-nucleoside polymerase inhibitors, the inhibition of NS5a has proved to be an effective way to treat HCV patients. Here we report on novel HCV NS5a inhibitors which were synthesized and evaluated in the HCV replicon assay. A series of inhibitors were formed by a cycloaddition reaction in parallel to establish new leads and explore the effects of unsymmetrical cap substitution. This led to the identification of several triazoles with picomolar potency in vitro against hepatitis C virus.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepacivirus/physiology , Virus Replication/drug effects , Cell Line , Chemistry Techniques, Synthetic , Viral Nonstructural Proteins/antagonists & inhibitors
3.
PLoS One ; 10(5): e0126959, 2015.
Article in English | MEDLINE | ID: mdl-26010881

ABSTRACT

BACKGROUND: The study assessed the antiviral activity of TMC353121, a respiratory syncytial virus (RSV) fusion inhibitor, in a preclinical non-human primate challenge model with a viral shedding pattern similar to that seen in humans, following continuous infusion (CI). METHODS: African green monkeys were administered TMC353121 through CI, in 2 studies. Study 1 evaluated the prophylactic and therapeutic efficacy of TMC353121 at a target plasma level of 50 ng/mL (n=15; Group 1: prophylactic arm [Px50], 0.033 mg/mL TMC353121, flow rate 2.5 mL/kg/h from 24 hours pre-infection to 10 days; Group 2: therapeutic arm [Tx50], 0.033 mg/mL TMC353121 from 24 hours postinfection to 8 days; Group 3: control [Vh1] vehicle, 24 hours post-infection to 8 days). Study 2 evaluated the prophylactic efficacy of TMC353121 at target plasma levels of 5 and 500 ng/mL (n=12; Group 1: prophylactic 5 arm [Px5], 0.0033 mg/mL TMC353121, flow rate 2.5 mL/kg/h from 72 hours pre-infection to 14 days; Group 2: prophylactic 500 arm [Px500], 0.33 mg/mL TMC353121; Group 3: control [Vh2] vehicle, 14 days). Bronchoalveolar lavage fluid and plasma were collected every 2 days from day 1 postinfection for pharmacokinetics and safety analysis. FINDINGS: TMC353121 showed a dose-dependent antiviral activity, varying from 1 log10 reduction of peak viral load to complete inhibition of the RSV replication. Complete inhibition of RSV shedding was observed for a relatively low plasma exposure (0.39 µg/mL) and was associated with a dose-dependent reduction in INFγ, IL6 and MIP1α. TMC353121 administered as CI for 16 days was generally well-tolerated. CONCLUSION: TMC353121 exerted dose-dependent antiviral effect ranging from full inhibition to absence of antiviral activity, in a preclinical model highly permissive for RSV replication. No new safety findings emerged from the study.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Pyridines/pharmacology , Respiratory Syncytial Viruses/physiology , Virus Internalization/drug effects , Animals , Antiviral Agents/pharmacokinetics , Area Under Curve , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacokinetics , Bronchoalveolar Lavage Fluid/virology , Chlorocebus aethiops , Cytokines/metabolism , Disease Models, Animal , Humans , Lung/drug effects , Lung/pathology , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Respiratory Syncytial Viruses/drug effects , Time Factors , Virus Shedding/drug effects
4.
J Acquir Immune Defic Syndr ; 65(3): 283-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24121756

ABSTRACT

OBJECTIVES: TMC310911 is a novel HIV type-1 (HIV-1) protease inhibitor with broad in vitro antiviral activity. In this phase 2a, open-label randomized study, the antiviral activity, pharmacokinetics, and safety and tolerability of ritonavir-boosted TMC310911 was assessed. METHODS: In this study, treatment-naive HIV-1 patients (aged 18-60 years) received 1 of the 4 dosing regimens of TMC310911: 150 mg twice-daily (bid) (n = 8), 300 mg bid (n = 8), 75 mg bid (n = 9), or 300 mg once-daily (qd) (n = 8), for 14 days, all coadministered with 100 mg of ritonavir, as only antiretroviral therapy. RESULTS: The mean change from baseline in HIV-1 RNA (log10 copies per milliliter; primary efficacy endpoint) was -1.30 (75 mg bid), -1.14 (150 mg bid), -1.07 (300 mg bid), and -1.06 (300 mg qd) on day 8 and -1.53 (75 mg bid), -1.79 (150 mg bid), -1.69 (300 mg bid), and -1.55 (300 mg qd) on day 15. At steady state (day 14), the mean maximum plasma concentration and mean area under the plasma concentration-time curve from 0 to 12 hours tended to increase dose proportionally for bid doses; TMC310911 daily exposures for the 300 mg qd treatment and 150 mg bid treatment were comparable. The most common (≥ 10%) treatment-emergent adverse events were fatigue (27.3%) and nausea (12.1%); no deaths or serious treatment-emergent adverse events were reported in this study. CONCLUSIONS: Combination treatment with TMC310911 and ritonavir showed potent antiviral activity (>1.5 log10 copies/mL decrease in plasma HIV-1 RNA) at all evaluated doses, and treatment was generally safe and well tolerated.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/therapeutic use , Adult , Anti-HIV Agents/adverse effects , Blood/virology , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , HIV Protease Inhibitors/adverse effects , HIV-1/isolation & purification , Humans , Male , Middle Aged , Ritonavir/therapeutic use , Treatment Outcome , Viral Load , Young Adult
5.
J Acquir Immune Defic Syndr ; 65(3): 299-305, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24121757

ABSTRACT

OBJECTIVES: To evaluate safety, tolerability, and pharmacokinetics of TMC310911, a novel human immunodeficiency virus type-1 protease inhibitor. METHODS: Healthy participants aged 18-55 years with body mass index 18-30 kg/m were enrolled in 2 phase 1 studies. In the first-in-human, single-dose study, 18 participants received placebo or TMC310911 (75-2000 mg) in the double-blind phase and 8 participants received 300 or 600 mg of TMC310911 [administered alone or with 100 mg ritonavir twice daily (bid)] in the subsequent open-label phase. The multiple-dose double-blind study included 5 successive treatment sessions wherein healthy participants received placebo or TMC310911 [300 mg bid, 600 mg once daily or 150 mg bid (plus 100 mg ritonavir bid), 900 mg bid (alone) or 300 mg bid (plus ritonavir 50 mg bid)]; in all sessions, TMC310911 and ritonavir were administered for 6 and 9 days, respectively. RESULTS: In the single-dose study, no dose-limiting toxicity was observed up to 2000 mg of TMC310911. Systemic exposure to TMC310911 generally increased in a dose-proportional manner after the single- or multiple-dose administrations. Coadministration of ritonavir increased the systemic exposure to TMC310911. The mean Cmax and area under plasma concentration-time curve values (single-dose: 1200 mg TMC310911) were higher under fasted conditions than in fed condition. In both studies, most treatment-emergent adverse events were related to gastrointestinal system. CONCLUSIONS: TMC310911 exhibited a linear pharmacokinetic profile after the single- (up to 2000 mg) and multiple-dose (up to 900 mg) administrations; ritonavir improved the pharmacokinetic profile of TMC310911. TMC310911 was generally safe and tolerable when administered with or without ritonavir.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Double-Blind Method , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Female , HIV Protease Inhibitors/administration & dosage , Healthy Volunteers , Humans , Male , Middle Aged , Placebos/administration & dosage , Ritonavir/administration & dosage , Young Adult
6.
Gastroenterology ; 143(5): 1176-1178.e6, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22885330

ABSTRACT

In the TMC435-C101 study, 6 patients infected with hepatitis C virus genotype 1 were treated with the protease inhibitor TMC435 (200 mg once daily) as monotherapy for 5 days. Approximately 1.5 years later, 5 of these patients were re-treated with TMC435 (200 mg once daily) plus pegylated interferon alfa-2a and ribavirin (PegIFNα-2a and RBV) for 4 weeks, followed by PegIFNα-2a and RBV until week 48 (in the Optimal Protease inhibitor Enhancement of Response to therApy [OPERA-1] study). TMC435-resistant variants, which emerged in all 5 patients during the TMC435-C101 study, were no longer detected at the beginning of the OPERA-1 study based on virus population sequencing. During the OPERA-1 study, 3 patients had a sustained virologic response; deep sequencing indicated low-level persistence of resistant variants in the remaining 2 patients, which might have affected their response to re-treatment.


Subject(s)
Hepacivirus/genetics , Hepatitis C/drug therapy , Heterocyclic Compounds, 3-Ring/therapeutic use , Protease Inhibitors/therapeutic use , RNA, Viral/blood , Sulfonamides/therapeutic use , Antiviral Agents/therapeutic use , Drug Resistance, Viral , Drug Therapy, Combination , Genotype , Hepatitis C/blood , Hepatitis C/virology , Humans , Interferon-alpha/therapeutic use , Male , Mutation , Polyethylene Glycols/therapeutic use , RNA, Viral/drug effects , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Simeprevir , Viral Load
7.
Antimicrob Agents Chemother ; 56(9): 4676-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22710121

ABSTRACT

Hepatitis C virus (HCV) infection is a major global health burden and is associated with an increased risk of liver cirrhosis and hepatocellular carcinoma. There remains an unmet medical need for efficacious and safe direct antivirals with complementary modes of action for combination in treatment regimens to deliver a high cure rate with a short duration of treatment for HCV patients. Here we report the in vitro inhibitory activity, mode of action, binding kinetics, and resistance profile of TMC647055, a novel and potent nonnucleoside inhibitor of the HCV NS5B RNA-dependent RNA polymerase. In vitro combination studies with an HCV NS3/4A protease inhibitor demonstrated potent suppression of HCV RNA replication, confirming the potential for combination of these two classes in the treatment of chronic HCV infection. TMC647055 is a potent nonnucleoside NS5B polymerase inhibitor of HCV replication with a promising in vitro biochemical, kinetic, and virological profile that is currently undergoing clinical evaluation.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Cell Line , Cloning, Molecular , Drug Combinations , Drug Synergism , Escherichia coli/genetics , Genes, Reporter , Hepacivirus/enzymology , Hepacivirus/genetics , Hepacivirus/growth & development , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Plasmids , RNA-Dependent RNA Polymerase/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transfection , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
8.
Bioorg Med Chem Lett ; 22(13): 4431-6, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22542193

ABSTRACT

Novel conformationaly constrained 1,6- and 2,6-macrocyclic HCV NS5b polymerase inhibitors, in which either the nitrogen or the phenyl ring in the C2 position of the central indole core is tethered to an acylsulfamide acid bioisostere, have been designed and tested for their anti-HCV potency. This transformational route toward non-zwitterionic finger loop-directed inhibitors led to the discovery of derivatives with improved cell potency and pharmacokinetic profile.


Subject(s)
Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Hepacivirus/enzymology , Indoles/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Allosteric Regulation , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Cell Line, Tumor , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Indoles/chemical synthesis , Indoles/pharmacokinetics , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
9.
Bioorg Med Chem Lett ; 22(9): 3265-8, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22472694

ABSTRACT

4'-Azido-2'-deoxy-2'-methylcytidine (14) is a potent nucleoside inhibitor of the HCV NS5B RNA-dependent RNA polymerase, displaying an EC(50) value of 1.2 µM and showing moderate in vivo bioavailability in rat (F=14%). Here we describe the synthesis and biological evaluation of 4'-azido-2'-deoxy-2'-methylcytidine and prodrug derivatives thereof.


Subject(s)
Antiviral Agents/chemistry , Cytidine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Hepacivirus/drug effects , Prodrugs/pharmacology , Animals , Antiviral Agents/pharmacology , Cytidine/pharmacology , Deoxycytidine/pharmacology , Drug Discovery , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Rats , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects
10.
Antivir Ther ; 16(7): 1021-33, 2011.
Article in English | MEDLINE | ID: mdl-22024518

ABSTRACT

BACKGROUND: Antiviral activity of TMC435, an oral, once-daily, HCV NS3/4A protease inhibitor, was evaluated with pegylated interferon-α2a/ribavirin (P/R) in HCV genotype-1 patients. METHODS: Optimal Protease inhibitor Enhancement of Response to TherApy (OPERA-1; TMC435-C201; NCT00561353) is a Phase IIa, randomized, placebo-controlled study. Treatment-naive patients (n=74) received 25, 75 or 200 mg TMC435 once daily, or placebo for 7 days followed by 21 days of triple therapy with P/R, or triple therapy for 28 days. Treatment-experienced patients (n=37; 56.8% with cirrhosis) received 75, 150 or 200 mg TMC435 once daily, or placebo with P/R for 28 days. Patients continued P/R up to week 48. RESULTS: Treatment-naive patients who received initial monotherapy had a rapid decline in HCV RNA by day 3. At day 7, HCV RNA reductions were greatest for the 75 and 200 mg doses (0.02, -2.63, -3.43 and -4.13 log(10) IU/ml for placebo, and TMC435 25, 75 and 200 mg, respectively). At day 28, all patients who received triple therapy with TMC435 75 or 200 mg had HCV RNA<25 IU/ml versus 4/9 for placebo. In total, 18/28 treatment-experienced patients (9/9 prior relapsers, 9/19 non-responders) who received TMC435 had HCV RNA<25 IU/ml at day 28 versus 0/9 for placebo; similar results were observed for the 150 and 200 mg doses. Most adverse events were grade 1/2. No relevant changes in laboratory parameters occurred, except mild and reversible bilirubin elevations, mostly at the 200 mg dose. CONCLUSIONS: Once-daily TMC435 with P/R showed potent, dose-dependent antiviral activity over 28 days, and had a favourable tolerability profile.


Subject(s)
Antiviral Agents/administration & dosage , Hepacivirus/drug effects , Hepatitis C/drug therapy , Heterocyclic Compounds, 3-Ring/administration & dosage , Interferon-alpha/administration & dosage , Polyethylene Glycols/administration & dosage , Protease Inhibitors/administration & dosage , Ribavirin/administration & dosage , Sulfonamides/administration & dosage , Adolescent , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Bilirubin/analysis , Double-Blind Method , Drug Therapy, Combination , Female , Genotype , Hepatitis C/virology , Heterocyclic Compounds, 3-Ring/adverse effects , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Interferon-alpha/therapeutic use , Male , Middle Aged , Polyethylene Glycols/therapeutic use , Protease Inhibitors/adverse effects , Protease Inhibitors/therapeutic use , RNA, Viral/blood , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Ribavirin/pharmacology , Ribavirin/therapeutic use , Simeprevir , Sulfonamides/adverse effects , Sulfonamides/therapeutic use , Viral Load
11.
J Med Chem ; 53(22): 8150-60, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-21033671

ABSTRACT

The current therapy for hepatitis C virus (HCV) infection has limited efficacy, in particular against the genotype 1 virus, and a range of side effects. In this context of high unmet medical need, more efficacious drugs targeting HCV nonstructural proteins are of interest. Here we describe 2'-deoxy-2'-spirocyclopropylcytidine (5) as a new inhibitor of the HCV NS5B RNA-dependent RNA polymerase, displaying an EC(50) of 7.3 µM measured in the Huh7-Rep cell line and no associated cytotoxicity (CC(50) > 98.4 µM). Computational results indicated high similarity between 5 and related HCV inhibiting nucleosides. A convenient synthesis was devised, facilitating synthesis of multigram quantities of 5. As the exposure measured after oral administration of 5 was found to be limited, the 3'-mono- and 3',5'-diisobutyryl ester prodrugs 20 and 23, respectively, were evaluated. The oral dosing of 23 led to substantially increased exposure to 5 in both rats and dogs.


Subject(s)
Antiviral Agents/chemical synthesis , Cytidine/analogs & derivatives , Hepacivirus/drug effects , Prodrugs/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Cell Line , Cytidine/chemical synthesis , Cytidine/chemistry , Cytidine/pharmacology , Dogs , Esters , Humans , Male , Models, Molecular , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Rats , Rats, Sprague-Dawley , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Structure-Activity Relationship , Virus Replication
12.
J Virol ; 84(21): 11124-33, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739521

ABSTRACT

Resistance to hepatitis C virus (HCV) inhibitors targeting viral enzymes has been observed in in vitro replicon studies and during clinical trials. The factors determining the emergence of resistance and the changes in the viral quasispecies population under selective pressure are not fully understood. To assess the dynamics of variants emerging in vitro under various selective pressures with TMC380765, a potent macrocyclic HCV NS3/4A protease inhibitor, HCV genotype 1b replicon-containing cells were cultured in the presence of a low, high, or stepwise-increasing TMC380765 concentration(s). HCV replicon RNA from representative samples thus obtained was analyzed using (i) population, (ii) clonal, and (iii) 454 deep sequencing technologies. Depending on the concentration of TMC380765, distinct mutational patterns emerged. In particular, culturing with low concentrations resulted in the selection of low-level resistance mutations (F43S and A156G), whereas high concentrations resulted in the selection of high-level resistance mutations (A156V, D168V, and D168A). Clonal and 454 deep sequencing analysis of the replicon RNA allowed the identification of low-frequency preexisting mutations possibly contributing to the mutational pattern that emerged. Stepwise-increasing TMC380765 concentrations resulted in the emergence and disappearance of multiple replicon variants in response to the changing selection pressure. Moreover, two different codons for the wild-type amino acids were observed at certain NS3 positions within one population of replicons, which may contribute to the emerging mutational patterns. Deep sequencing technologies enabled the study of minority variants present in the HCV quasispecies population present at baseline and during antiviral drug pressure, giving new insights into the dynamics of resistance acquisition by HCV.


Subject(s)
Biological Evolution , Drug Resistance , Hepacivirus/genetics , Mutation, Missense , Protease Inhibitors/pharmacology , Selection, Genetic/drug effects , Base Sequence , Cell Line , DNA Mutational Analysis , Hepacivirus/growth & development , Humans , RNA, Viral/analysis , RNA, Viral/genetics , Replicon/genetics
13.
Bioorg Med Chem Lett ; 20(14): 4004-11, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20541405

ABSTRACT

Novel NS3/4A protease inhibitors comprising quinazoline derivatives as P2 substituent were synthesized. High potency inhibitors displaying advantageous PK properties have been obtained through the optimization of quinazoline P2 substituents in three series exhibiting macrocyclic P2 cyclopentane dicarboxylic acid and P2 proline urea motifs. For the quinazoline moiety it was found that 8-methyl substitution in the P2 cyclopentane dicarboxylic acid series improved on the metabolic stability in human liver microsomes. By comparison, the proline urea series displayed advantageous Caco-2 permeability over the cyclopentane series. Pharmacokinetic properties in vivo were assessed in rat on selected compounds, where excellent exposure and liver-to-plasma ratios were demonstrated for a member of the 14-membered quinazoline substituted P2 proline urea series.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Hepacivirus/enzymology , Protease Inhibitors/chemical synthesis , Quinazolines/chemical synthesis , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Area Under Curve , Caco-2 Cells , Humans , Intracellular Signaling Peptides and Proteins , Microsomes, Liver/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Structure-Activity Relationship
14.
Antimicrob Agents Chemother ; 54(5): 1878-87, 2010 May.
Article in English | MEDLINE | ID: mdl-20176898

ABSTRACT

TMC435 is a small-molecule inhibitor of the NS3/4A serine protease of hepatitis C virus (HCV) currently in phase 2 development. The in vitro resistance profile of TMC435 was characterized by selection experiments with HCV genotype 1 replicon cells and the genotype 2a JFH-1 system. In 80% (86/109) of the sequences from genotype 1 replicon cells analyzed, a mutation at NS3 residue D168 was observed, with changes to V or A being the most frequent. Mutations at NS3 positions 43, 80, 155, and 156, alone or in combination, were also identified. A transient replicon assay confirmed the relevance of these positions for TMC435 inhibitory activity. The change in the 50% effective concentrations (EC(50)s) observed for replicons with mutations at position 168 ranged from <10-fold for those with the D168G or D168N mutation to approximately 2,000-fold for those with the D168V or D168I mutation, compared to the EC(50) for the wild type. Of the positions identified, mutations at residue Q80 had the least impact on the activity of TMC435 (<10-fold change in EC(50)s), while greater effects were observed for some replicons with mutations at positions 43, 155, and 156. TMC435 remained active against replicons with the specific mutations observed after in vitro or in vivo exposure to telaprevir or boceprevir, including most replicons with changes at positions 36, 54, and 170 (<3-fold change in EC(50)s). Replicons carrying mutations affecting the activity of TMC435 remained fully susceptible to alpha interferon and NS5A and NS5B inhibitors. Finally, combinations of TMC435 with alpha interferon and NS5B polymerase inhibitors prevented the formation of drug-resistant replicon colonies.


Subject(s)
Hepacivirus/drug effects , Hepatitis C/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Protease Inhibitors/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/pharmacology , Cell Line , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , Drug Synergism , Genotype , Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C/virology , Humans , In Vitro Techniques , Interferon-alpha/pharmacology , Mutagenesis , Simeprevir , Viral Nonstructural Proteins/genetics , Virus Replication/drug effects
16.
J Virol ; 84(6): 2923-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20071590

ABSTRACT

The RNA-dependent RNA polymerase (NS5B) of hepatitis C virus (HCV) is an unusually attractive target for drug discovery since it contains five distinct drugable sites. The success of novel antiviral therapies will require nonnucleoside inhibitors to be active in at least patients infected with HCV of subtypes 1a and 1b. Therefore, the genotypic assessment of these agents against clinical isolates derived from genotype 1-infected patients is an important prerequisite for the selection of suitable candidates for clinical development. Here we report the 1a/1b subtype profiling of polymerase inhibitors that bind at each of the four known nonnucleoside binding sites. We show that inhibition of all of the clinical isolates tested is maintained, except for inhibitors that bind at the palm-1 binding site. Subtype coverage varies across chemotypes within this class of inhibitors, and inhibition of genotype 1a improves when hydrophobic contact with the polymerase is increased. We investigated if the polymorphism of the palm-1 binding site is the sole cause of the reduced susceptibility of subtype 1a to inhibition by 1,5-benzodiazepines by using reverse genetics, X-ray crystallography, and surface plasmon resonance studies. We showed Y415F to be a key determinant in conferring resistance on subtype 1a, with this effect being mediated through an inhibitor- and enzyme-bound water molecule. Binding studies revealed that the mechanism of subtype 1a resistance is faster dissociation of the inhibitor from the enzyme.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/enzymology , Hepatitis C/drug therapy , Isoenzymes/antagonists & inhibitors , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Binding Sites , Crystallography, X-Ray , Drug Discovery , Hepacivirus/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Replicon/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
17.
Gastroenterology ; 138(3): 913-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19852962

ABSTRACT

BACKGROUND & AIMS: The search for targeted anti-hepatitis C virus (HCV) drugs is driven by the adverse effect profile and limited efficacy of the current standard of care (pegylated interferon-alpha/ribavirin). In a first-in-human trial, we tested the safety, tolerability, and pharmacokinetics of the macrocyclic HCV NS3/4A protease inhibitor TMC435 in healthy volunteers, followed by HCV genotype 1-infected patients to assess antiviral activity. METHODS: The TMC435350-C101 study was a phase I, randomized, double-blind, placebo-controlled trial in 49 healthy volunteers, followed by an open-label, nonplacebo-controlled panel in 6 genotype 1 hepatitis C patients. Healthy volunteers received oral, single, ascending doses (up to 600 mg) or 5-day multiple ascending doses (200 mg twice daily or 100, 200, or 400 mg once daily). Patients received 200 mg once daily for 5 days. Pharmacokinetics and safety were evaluated for all panels, and plasma HCV-RNA levels were determined in patients. RESULTS: There were no serious adverse events, no grade 3 reactions, and no treatment-related discontinuations; pharmacokinetics supported a once daily dosing regimen. Plasma HCV-RNA levels dropped rapidly in all patients, with a median maximal reduction of 3.9-log(10) IU/mL and a median of 6 days to maximal reduction. The initial steep reduction of HCV-RNA (median 3.5-log(10) IU/mL at day 3) was followed by a more gradual decline that was maintained over the dosing period. No viral breakthroughs (>1-log(10) IU/mL HCV-RNA increase from nadir) were observed during treatment nor in the 3 days posttreatment; HCV-RNA returned to pretreatment levels by week 4. CONCLUSIONS: Once daily TMC435 given orally was generally safe and well tolerated and demonstrated potent antiviral activity.


Subject(s)
Antiviral Agents/administration & dosage , Hepacivirus/drug effects , Hepatitis C/drug therapy , Heterocyclic Compounds, 3-Ring/administration & dosage , Protease Inhibitors/administration & dosage , RNA, Viral/blood , Sulfonamides/administration & dosage , Viral Load/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Adult , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , DNA, Viral/analysis , Double-Blind Method , Drug Administration Schedule , Female , Genotype , Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C/diagnosis , Heterocyclic Compounds, 3-Ring/adverse effects , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Male , Middle Aged , Protease Inhibitors/adverse effects , Protease Inhibitors/pharmacokinetics , Simeprevir , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Time Factors , Treatment Outcome , Viral Nonstructural Proteins/genetics , Young Adult
18.
Bioorg Med Chem Lett ; 19(9): 2492-6, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19342234

ABSTRACT

Optimization through parallel synthesis of a novel series of hepatitis C virus (HCV) NS5B polymerase inhibitors led to the identification of (R)-11-(4-benzyloxy-2-fluorophenyl)-6-hydroxy-3,3-dimethyl-10-(6-methylpyridine-2-carbonyl)-2,3,4,5,10,11-hexahydro-dibenzo[b,e][1,4]diazepin-1-one 11zc and (R)-11-(4-benzyloxy-2-fluorophenyl)-6-hydroxy-3,3-dimethyl-10-(2,5-dimethyloxazol-4-carbonyl)-2,3,4,5,10,11-hexahydro-dibenzo[b,e][1,4]diazepin-1-one 11zk as potent (replicon EC(50)=400nM and 270nM, respectively) and selective (CC(50)>20muM) inhibitors of HCV replication. These data warrant further lead-optimization efforts.


Subject(s)
Antiviral Agents/chemical synthesis , Benzodiazepines/chemistry , Chemistry, Pharmaceutical/methods , Hepacivirus/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Acrylates/chemistry , Antiviral Agents/pharmacology , Crystallography, X-Ray , Drug Design , Hepacivirus/enzymology , Humans , Inhibitory Concentration 50 , Models, Chemical , Molecular Structure , Structure-Activity Relationship
19.
Antimicrob Agents Chemother ; 53(4): 1377-85, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19171797

ABSTRACT

The hepatitis C virus (HCV) NS3/4A serine protease has been explored as a target for the inhibition of viral replication in preclinical models and in HCV-infected patients. TMC435350 is a highly specific and potent inhibitor of NS3/4A protease selected from a series of novel macrocyclic inhibitors. In biochemical assays using NS3/4A proteases of genotypes 1a and 1b, inhibition constants of 0.5 and 0.4 nM, respectively, were determined. TMC435350 inhibited HCV replication in a cellular assay (subgenomic 1b replicon) with a half-maximal effective concentration (EC(50)) of 8 nM and a selectivity index of 5,875. The compound was synergistic with alpha interferon and an NS5B inhibitor in the replicon model and additive with ribavirin. In rats, TMC435350 was extensively distributed to the liver and intestinal tract (tissue/plasma area under the concentration-time curve ratios of >35), and the absolute bioavailability was 44% after a single oral administration. Compound concentrations detected in both plasma and liver at 8 h postdosing were above the EC(99) value measured in the replicon. In conclusion, given the selective and potent in vitro anti-HCV activity, the potential for combination with other anti-HCV agents, and the favorable pharmacokinetic profile, TMC435350 has been selected for clinical development.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Sulfonamides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Drug Therapy, Combination , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Humans , Interferon-alpha/administration & dosage , Male , Protein Binding , Rats , Rats, Sprague-Dawley , Simeprevir , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Tissue Distribution , Virus Replication/drug effects
20.
Antimicrob Agents Chemother ; 52(12): 4420-31, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18852280

ABSTRACT

The exogenous control of hepatitis C virus (HCV) replication can be mediated through the inhibition of the RNA-dependent RNA polymerase (RdRp) activity of NS5B. Small-molecule inhibitors of NS5B include nucleoside and nonnucleoside analogs. Here, we report the discovery of a novel class of HCV polymerase nonnucleoside inhibitors, 1,5-benzodiazepines (1,5-BZDs), identified by high-throughput screening of a library of small molecules. A fluorescence-quenching assay and X-ray crystallography revealed that 1,5-BZD 4a bound stereospecifically to NS5B next to the catalytic site. When introduced into replicons, mutations known to confer resistance against chemotypes that bind at this site were detrimental to inhibition by 1,5-BZD 7a. Using a panel of enzyme isolates that covered genotypes 1 to 6, we showed that compound 4a inhibited genotype 1 only. In mechanistic studies, 4a was found to inhibit the RdRp activity of NS5B noncompetitively with GTP and to inhibit the formation of the first phosphodiester bond during the polymerization cycle. The specificity for the HCV target was evaluated by profiling the 1,5-BZDs against other viral and human polymerases, as well as BZD receptors.


Subject(s)
Benzodiazepines/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/metabolism , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Genotype , Hepacivirus/enzymology , Hepacivirus/genetics , Hepacivirus/physiology , Humans , Models, Molecular , Molecular Sequence Data , Mutation , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...