Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
ACS Med Chem Lett ; 5(6): 679-84, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24944743

ABSTRACT

Nucleoside analogues have long been recognized as prospects for the discovery of direct acting antivirals (DAAs) to treat hepatitis C virus because they have generally exhibited cross-genotype activity and a high barrier to resistance. C-Nucleosides have the potential for improved metabolism and pharmacokinetic properties over their N-nucleoside counterparts due to the presence of a strong carbon-carbon glycosidic bond and a non-natural heterocyclic base. Three 2'CMe-C-adenosine analogues and two 2'CMe-guanosine analogues were synthesized and evaluated for their anti-HCV efficacy. The nucleotide triphosphates of four of these analogues were found to inhibit the NS5B polymerase, and adenosine analogue 1 was discovered to have excellent pharmacokinetic properties demonstrating the potential of this drug class.

2.
Antimicrob Agents Chemother ; 57(10): 4928-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877706

ABSTRACT

Faldaprevir (BI 201335) is a selective NS3/4A protease inhibitor under development for the treatment of chronic hepatitis C virus (HCV) infection. NS3/4A genotyping and NS3 protease phenotyping analyses were performed to monitor the emergence of resistance in patients with HCV genotype 1 infection receiving faldaprevir alone or combined with pegylated interferon alfa 2a and ribavirin (PegIFN-RBV) during a phase 1b study. Among all baseline variants, a maximum 7-fold reduction in in vitro sensitivity to faldaprevir was observed for a rare NS3 (V/I)170T polymorphism. During faldaprevir monotherapy in treatment-naive patients, virologic breakthrough was common (77%, 20/26) and was associated with the emergence of resistance mutations predominantly carrying NS3 substitutions R155K in GT1a and D168V in GT1b. D168V conferred a greater reduction in faldaprevir sensitivity (1,800-fold) than R155K (330-fold); however, D168V was generally less fit than R155K in the absence of selective drug pressure. Treatment-experienced patients treated with faldaprevir-PegIFN-RBV triple therapy showed higher viral load reductions, lower rates of breakthrough (8%, 5/62), and less frequent emergence of resistance-associated variants compared with faldaprevir monotherapy. (This study has been registered at ClinicalTrials.gov under registration no. NCT00793793.).


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/drug effects , Oligopeptides/therapeutic use , Thiazoles/therapeutic use , Aminoisobutyric Acids , Cell Line , Genotype , Genotyping Techniques/methods , Humans , Leucine/analogs & derivatives , Proline/analogs & derivatives , Protease Inhibitors/therapeutic use , Quinolines , Viral Nonstructural Proteins/antagonists & inhibitors
3.
J Biol Chem ; 288(8): 5673-81, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23271737

ABSTRACT

Although optimizing the resistance profile of an inhibitor can be challenging, it is potentially important for improving the long term effectiveness of antiviral therapy. This work describes our rational approach toward the identification of a macrocyclic acylsulfonamide that is a potent inhibitor of the NS3-NS4A proteases of all hepatitis C virus genotypes and of a panel of genotype 1-resistant variants. The enhanced potency of this compound versus variants D168V and R155K facilitated x-ray determination of the inhibitor-variant complexes. In turn, these structural studies revealed a complex molecular basis of resistance and rationalized how such compounds are able to circumvent these mechanisms.


Subject(s)
Carrier Proteins/chemistry , Drug Resistance, Viral , Hepatitis C/drug therapy , Viral Nonstructural Proteins/chemistry , Antiviral Agents/pharmacology , Cell Line , Cloning, Molecular , Crystallography, X-Ray/methods , Genotype , Humans , Intracellular Signaling Peptides and Proteins , Models, Chemical , Models, Molecular , Molecular Conformation , Protease Inhibitors/pharmacology , Sulfonamides/chemistry , X-Rays
4.
Antimicrob Agents Chemother ; 56(1): 569-72, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22024816

ABSTRACT

The in vitro resistance profile of BI 201335 was evaluated through selection and characterization of variants in genotype 1a (GT 1a) and genotype 1b (GT 1b) replicons. NS3 R155K and D168V were the most frequently observed resistant variants. Phenotypic characterization of the mutants revealed shifts in sensitivity specific to BI 201335 that did not alter susceptibility to alpha interferon. In contrast to macrocyclic and covalent protease inhibitors, changes at V36, T54, F43, and Q80 did not confer resistance to BI 201335.


Subject(s)
Hepacivirus/genetics , Interferon-alpha/pharmacology , Oligopeptides/pharmacology , Thiazoles/pharmacology , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Aminoisobutyric Acids , Antiviral Agents/pharmacology , Crystallography, X-Ray , Drug Resistance, Viral , Genotype , Hepacivirus/drug effects , Hepacivirus/enzymology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Inhibitory Concentration 50 , Kinetics , Leucine/analogs & derivatives , Mutagenesis, Site-Directed , Mutation Rate , Phenotype , Proline/analogs & derivatives , Protease Inhibitors/pharmacology , Quinolines , Replicon , Viral Nonstructural Proteins/metabolism
5.
Antimicrob Agents Chemother ; 54(11): 4611-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823284

ABSTRACT

BI 201335 is a hepatitis C virus (HCV) NS3-NS4A (NS3 coexpressed with NS4A) protease inhibitor that has been shown to have potent clinical antiviral activity. It is a highly optimized noncovalent competitive inhibitor of full-length NS3-NS4A proteases of HCV genotypes 1a and 1b with K(i) values of 2.6 and 2.0 nM, respectively. K(i) values of 2 to 230 nM were measured against the NS3-NS4A proteases of HCV genotypes 2 to 6, whereas it was a very weak inhibitor of cathepsin B and showed no measurable inhibition of human leukocyte elastase. BI 201335 was also shown to be a potent inhibitor of HCV RNA replication in vitro with 50% effective concentrations (EC(50)s) of 6.5 and 3.1 nM obtained in genotype 1a and 1b replicon assays. Combinations of BI 201335 with either interferon or ribavirin had additive effects in replicon assays. BI 201335 had good permeability in Caco-2 cell assays and high metabolic stability after incubation with human, rat, monkey, and dog liver microsomes. Its good absorption, distribution, metabolism, and excretion (ADME) profile in vitro, as well as in rat, monkey, and dog, predicted good pharmacokinetics (PK) in humans. Furthermore, drug levels were significantly higher in rat liver than in plasma, suggesting that distribution to the target organ may be especially favorable. BI 201335 is a highly potent and selective NS3-NS4A protease inhibitor with good in vitro and animal ADME properties, consistent with its good human PK profile, and shows great promise as a treatment for HCV infection.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Hepacivirus/enzymology , Oligopeptides/pharmacology , Oligopeptides/pharmacokinetics , Thiazoles/pharmacology , Thiazoles/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Aminoisobutyric Acids , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins , Leucine/analogs & derivatives , Male , Proline/analogs & derivatives , Proline/pharmacology , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Quinolines , Rats , Viral Nonstructural Proteins/drug effects , Virus Replication/drug effects
6.
Bioorg Med Chem Lett ; 20(3): 857-61, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20074949

ABSTRACT

SAR studies at the N(1)-position of allosteric indole-based HCV NS5B inhibitors has led to the discovery of acetamide derivatives with good cellular potency in subgenomic replicons (EC(50) <200 nM). This class of inhibitors displayed improved physicochemical properties and favorable ADME-PK profiles over previously described analogs in this class.


Subject(s)
Acetamides/chemistry , Antiviral Agents/chemical synthesis , Carboxylic Acids/chemistry , Drug Discovery , Hepacivirus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Acetamides/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Antiviral Agents/pharmacology , Caco-2 Cells , Carboxylic Acids/pharmacology , Cell Line , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Drug Discovery/methods , Hepacivirus/drug effects , Humans , Microsomes, Liver/enzymology , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism
7.
Bioorg Med Chem Lett ; 16(19): 4987-93, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16908138

ABSTRACT

Benzimidazole-based allosteric inhibitors of the hepatitis C virus (HCV) NS5B polymerase were diversified to a variety of topologically related scaffolds. Replacement of the polar benzimidazole core by lipophilic indoles led to inhibitors with improved potency in the cell-based subgenomic HCV replicon system. Transposing the indole scaffold into a previously described series of benzimidazole-tryptophan amides generated the most potent inhibitors of HCV RNA replication in cell culture reported to date in this series (EC(50) approximately 50 nM).


Subject(s)
Benzimidazoles/pharmacology , Hepacivirus/drug effects , Indoles/pharmacology , Replicon/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Allosteric Regulation , Cell Line , Humans , Inhibitory Concentration 50 , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Structure-Activity Relationship
8.
J Virol ; 79(20): 13105-15, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16189012

ABSTRACT

Respiratory syncytial virus (RSV) is a major cause of respiratory illness in infants, immunocompromised patients, and the elderly. New antiviral agents would be important tools in the treatment of acute RSV disease. RSV encodes its own RNA-dependent RNA polymerase that is responsible for the synthesis of both genomic RNA and subgenomic mRNAs. The viral polymerase also cotranscriptionally caps and polyadenylates the RSV mRNAs at their 5' and 3' ends, respectively. We have previously reported the discovery of the first nonnucleoside transcriptase inhibitor of RSV polymerase through high-throughput screening. Here we report the design of inhibitors that have improved potency both in vitro and in antiviral assays and that also exhibit activity in a mouse model of RSV infection. We have isolated virus with reduced susceptibility to this class of inhibitors. The mutations conferring resistance mapped to a novel motif within the RSV L gene, which encodes the catalytic subunit of RSV polymerase. This motif is distinct from the catalytic region of the L protein and bears some similarity to the nucleotide binding domain within nucleoside diphosphate kinases. These findings lead to the hypothesis that this class of inhibitors may block synthesis of RSV mRNAs by inhibiting guanylylation of viral transcripts. We show that short transcripts produced in the presence of inhibitor in vitro do not contain a 5' cap but, instead, are triphosphorylated, confirming this hypothesis. These inhibitors constitute useful tools for elucidating the molecular mechanism of RSV capping and represent valid leads for the development of novel anti-RSV therapeutics.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA-Dependent RNA Polymerase/metabolism , Respiratory Syncytial Viruses/drug effects , Respiratory Syncytial Viruses/enzymology , Ribonucleoproteins/pharmacology , Administration, Intranasal , Amino Acid Sequence , Animals , Catalytic Domain/genetics , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA Caps/biosynthesis , RNA Caps/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/genetics , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Ribonucleoproteins/administration & dosage , Ribonucleoproteins/chemistry , Sequence Alignment , Virus Replication/drug effects
9.
Can J Physiol Pharmacol ; 83(2): 198-213, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15791294

ABSTRACT

Intranasal infection of BALB/c mice with respiratory syncytial virus (RSV)-A2 (0.5 x 10(8) - 2.0 x 10(8) plaque-forming units, PFU) produced disease characterized by weight loss (2-3 g) and mortality (60%-100%) with the mean day of death ranging from 6-7 d after infection. The extent of RSV disease was inoculum titer-dependent and required a replication competent virus. Lung titers of virus peaked at 0.5-1 x 10(6) PFU/g wet weight. Bronchoalveolar lavage fluid (BALF) levels of IL-1beta, TNF-alpha, INF-gamma IL-12, IL-6, MIP-1alpha, RANTES, and protein were elevated, whereas IL-2, IL-4, IL-5, IL-13, and IL-10 were unchanged. Histological assessment of lungs revealed marked inflammatory pathology characterized by bronchiolitis, vasculitis, and interstitial pneumonia. Whole-body plethysmography revealed significant disease-associated deficits of respiratory function. Therapy with ribavirin administered either by the intranasal, subcutaneous, or oral route significantly reduced disease in a dose-dependent manner. Delaying the initiation of therapy resulted in a loss of activity for ribavirin. Synagis administered either intramuscularly as a single dose in prophylaxis or intranasally in prophylaxis, followed by therapy, also significantly reduced disease in a dose-dependent manner. Infection of mice with a high titer inoculum of RSV-A2 resulted in severe and fatal pulmonary disease that was responsive to treatment. This model may be useful to characterize the in vivo activity of experimental therapies for RSV infection.


Subject(s)
Antiviral Agents/therapeutic use , Disease Models, Animal , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Viruses/pathogenicity , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/virology , Cytokines/metabolism , Lung/pathology , Lung/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , Palivizumab , Respiratory Function Tests , Respiratory Syncytial Virus Infections/mortality , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/physiology , Ribavirin/therapeutic use , Viral Proteins/metabolism , Virus Replication
10.
Nucleic Acids Res ; 32(16): 4758-67, 2004.
Article in English | MEDLINE | ID: mdl-15356293

ABSTRACT

RNA-dependent RNA polymerase from respiratory syncytial virus (RSV) is a multi-subunit ribonucleoprotein (RNP) complex that, in addition to synthesizing the full 15 222 nt viral genomic RNA, is able to synthesize all 10 viral mRNAs. We have prepared crude RNP from RSV-infected HEp-2 cells, based on a method previously used for Newcastle disease virus, and established a novel polyadenylation-dependent capture [poly(A) capture] assay to screen for potential inhibitors of RSV transcriptase activity. In this homogeneous assay, radiolabeled full-length polyadenylated mRNAs produced by the viral RNP are detected through capture on immobilized biotinylated oligo(dT) in a 96-well streptavidin-coated FlashPlate. Possible inhibitors identified with this assay could interfere at any step required for the production of complete RSV mRNAs, including transcription, polyadenylation and, potentially, co-transcriptional guanylylation. A specific inhibitor of RSV transcriptase with antiviral activity was identified through screening of this assay.


Subject(s)
RNA-Directed DNA Polymerase/metabolism , Respiratory Syncytial Viruses/enzymology , Reverse Transcriptase Inhibitors/analysis , Cell Line , Genetic Techniques , Humans , Polyadenylation , RNA, Messenger/metabolism , RNA-Directed DNA Polymerase/isolation & purification , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Transcription, Genetic
11.
J Virol ; 78(14): 7352-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15220408

ABSTRACT

Hepatitis C virus (HCV) displays a high degree of genetic variability. Six genotypes and more than 50 subtypes have been identified to date. In this report, kinetic profiles were determined for NS3 proteases of genotypes 1a, 1b, 2ac, 2b, and 3a, revealing no major differences in activity. In vitro sensitivity studies with BILN 2061 showed a decrease in affinity for proteases of genotypes 2 and 3 (K(i), 80 to 90 nM) compared to genotype 1 enzymes (K(i), 1.5 nM). To understand the reduced sensitivity of genotypes 2 and 3 to BILN 2061, active-site residues in the proximity of the inhibitor binding site were replaced in the genotype-1b enzyme with the corresponding genotype-2b or -3a residues. The replacement of five residues at positions 78, 79, 80, 122, and 132 accounted for most of the reduced sensitivity of genotype 2b, while replacement of residue 168 alone could account for the reduced sensitivity of genotype 3a. BILN 2061 remains a potent inhibitor of these non-genotype-1 NS3-NS4A proteins, with K(i) values below 100 nM. This in vitro potency, in conjunction with the good pharmacokinetic data reported for humans, suggests that there is potential for BILN 2061 as an antiviral agent for individuals infected with non-genotype-1 HCV.


Subject(s)
Antiviral Agents/pharmacology , Carbamates/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Macrocyclic Compounds , Quinolines , Serine Proteinase Inhibitors/pharmacology , Thiazoles/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Amino Acid Sequence , Binding Sites , Escherichia coli/enzymology , Escherichia coli/genetics , Genotype , Hepacivirus/classification , Humans , Kinetics , Microbial Sensitivity Tests/methods , Models, Molecular , Molecular Sequence Data , Mutation , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
12.
Nature ; 426(6963): 186-9, 2003 Nov 13.
Article in English | MEDLINE | ID: mdl-14578911

ABSTRACT

Hepatitis C virus (HCV) infection is a serious cause of chronic liver disease worldwide with more than 170 million infected individuals at risk of developing significant morbidity and mortality. Current interferon-based therapies are suboptimal especially in patients infected with HCV genotype 1, and they are poorly tolerated, highlighting the unmet medical need for new therapeutics. The HCV-encoded NS3 protease is essential for viral replication and has long been considered an attractive target for therapeutic intervention in HCV-infected patients. Here we identify a class of specific and potent NS3 protease inhibitors and report the evaluation of BILN 2061, a small molecule inhibitor biologically available through oral ingestion and the first of its class in human trials. Administration of BILN 2061 to patients infected with HCV genotype 1 for 2 days resulted in an impressive reduction of HCV RNA plasma levels, and established proof-of-concept in humans for an HCV NS3 protease inhibitor. Our results further illustrate the potential of the viral-enzyme-targeted drug discovery approach for the development of new HCV therapeutics.


Subject(s)
Antiviral Agents/therapeutic use , Carbamates/pharmacology , Hepacivirus/drug effects , Hepacivirus/physiology , Hepatitis C/drug therapy , Macrocyclic Compounds , Quinolines , Serine Proteinase Inhibitors/therapeutic use , Thiazoles/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Carbamates/administration & dosage , Carbamates/chemistry , Carbamates/pharmacokinetics , Double-Blind Method , Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C/virology , Humans , Male , Polyproteins/metabolism , Protein Processing, Post-Translational/drug effects , Serine Proteinase Inhibitors/administration & dosage , Serine Proteinase Inhibitors/pharmacokinetics , Serine Proteinase Inhibitors/pharmacology , Thiazoles/administration & dosage , Thiazoles/chemistry , Thiazoles/pharmacokinetics , Viral Load , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism
13.
J Biol Chem ; 278(22): 20374-80, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12646587

ABSTRACT

The hepatitis C virus (HCV) NS3 protease is essential for polyprotein maturation and viral propagation, and it has been proposed as a suitable target for antiviral drug discovery. An N-terminal hexapeptide cleavage product of a dodecapeptide substrate identified as a weak competitive inhibitor of the NS3 protease activity was optimized to a potent and highly specific inhibitor of the enzyme. The effect of this potent NS3 protease inhibitor was evaluated on replication of subgenomic HCV RNA and compared with interferon-alpha (IFN-alpha), which is currently used in the treatment of HCV-infected patients. Treatment of replicon-containing cells with the NS3 protease inhibitor or IFN-alpha showed a dose-dependent decrease in subgenomic HCV RNA that reached undetectable levels following a 14-day treatment. Kinetic studies in the presence of either NS3 protease inhibitor or IFN-alpha also revealed similar profiles in HCV RNA decay with half-lives of 11 and 14 h, respectively. The finding that an antiviral specifically targeting the NS3 protease activity inhibits HCV RNA replication further validates the NS3 enzyme as a prime target for drug discovery and supports the development of NS3 protease inhibitors as a novel therapeutic approach for HCV infection.


Subject(s)
Hepacivirus/drug effects , RNA, Viral/drug effects , Serine Proteinase Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Hepacivirus/enzymology , Hepacivirus/genetics , Hepatitis C/drug therapy , Humans , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , RNA, Viral/biosynthesis , Serine Proteinase Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...