Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Hepatol ; 60(5): 920-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24434503

ABSTRACT

BACKGROUND & AIMS: Samatasvir is a pan-genotypic inhibitor of the hepatitis C (HCV) non-structural protein 5A (NS5A). This study evaluated the antiviral activity, pharmacokinetics and safety of samatasvir monotherapy in treatment-naïve subjects infected with HCV genotype 1-4. METHODS: Thirty-four genotype 1 and thirty genotype 2, 3 or 4 subjects were randomized to receive for 3days placebo or samatasvir 25-100mg per day. Plasma samples for HCV RNA, pharmacokinetics and sequencing were collected up to day 10. RESULTS: Samatasvir achieved potent antiviral activity across genotypes: mean maximum reductions from baseline were 3.2-3.6 (genotype 1a), 3.0-4.3 (genotype 1b), 3.2-3.4 (genotype 3), and 3.6-3.9 (genotype 4) log10/ml respectively; no viral rebound was observed during the 3-day treatment period. For genotype 2 HCV, samatasvir was active in subjects with NS5A L31 polymorphism at baseline (individual range 2.5-4.1 log10/ml), but showed minimal activity in those with baseline M31 polymorphism. Samatasvir exhibited a long plasma half-life of approximately 20h which supports once daily dosing. Samatasvir was well tolerated in all subjects with no safety-related discontinuations or serious adverse events. The most common adverse events included constipation, nausea and headache and occurred at similar frequency in active and placebo subjects. All events were mild or moderate in intensity. There were no patterns or dose dependence of adverse events, vital signs, laboratory parameters or electrocardiograms. CONCLUSIONS: Samatasvir 25-100mg monotherapy for 3days was well tolerated and induced a rapid and profound reduction in plasma HCV RNA in subjects infected with HCV genotype 1-4. Samatasvir is being evaluated in combination with other direct-acting antiviral agents in subjects with HCV infection.


Subject(s)
Antiviral Agents/administration & dosage , Benzimidazoles/administration & dosage , Carbamates/administration & dosage , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Viral Nonstructural Proteins/antagonists & inhibitors , Adult , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Carbamates/adverse effects , Carbamates/pharmacokinetics , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Genotype , Half-Life , Hepacivirus/drug effects , Humans , Male , Middle Aged , Polymorphism, Genetic , RNA, Viral/blood , RNA, Viral/genetics , Viral Nonstructural Proteins/genetics
2.
Bioorg Med Chem Lett ; 24(12): 2699-702, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24815510

ABSTRACT

The synthesis of 2'-O,4'-C-methylene-bridged bicyclic guanine ribonucleosides bearing 2'-C-methyl or 5'-C-methyl modifications is described. Key to the successful installation of the methyl functionality in both cases was the use of a one-pot oxidation-Grignard procedure to avoid formation of the respective unreactive hydrates prior to alkylation. The 2'-C-methyl- and 5'-C-methyl-modified bicyclic guanosines were evaluated, along with the known uracil-, cytosine-, adenine-, guanine-LNA and guanine-ENA nucleosides, as potential antiviral agents and found to be inactive in the hepatitis C virus (HCV) cell-based replicon assay. Examination of the corresponding nucleoside triphosphates, however, against the purified HCV NS5B polymerase indicated that LNA-G and 2'-C-methyl-LNA-G are potent inhibitors of both 1b wild type and S282T mutant enzymes in vitro. Activity was further demonstrated for the LNA-G-triphosphate against HCV NS5B polymerase genotypes 1a, 2a, 3a and 4a. A phosphorylation by-pass prodrug strategy may be required to promote anti-HCV activity in the replicon assay.


Subject(s)
Hepacivirus/enzymology , Nucleic Acid Synthesis Inhibitors/chemical synthesis , Nucleic Acid Synthesis Inhibitors/pharmacology , Ribonucleosides/chemical synthesis , Ribonucleosides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Enzyme Activation/drug effects , Hepacivirus/drug effects , Molecular Structure , Ribonucleosides/chemistry
3.
Antimicrob Agents Chemother ; 56(12): 6372-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23070151

ABSTRACT

IDX184 is a liver-targeted prodrug of 2'-methylguanosine (2'-MeG) monophosphate. This study investigated the safety, tolerability, antiviral activity, and pharmacokinetics of IDX184 as a single agent in treatment-naïve patients with genotype-1 chronic hepatitis C virus (HCV) infection. Forty-one patients with baseline HCV RNA ≥ 5 log(10) IU/ml, alanine aminotransferase (ALT) ≤ 2.5× the upper limit of normal, and compensated liver disease were dosed. Sequential cohorts of 10 patients, randomized 8:2 (active:placebo), received 25, 50, 75, and 100 mg of IDX184 once daily for 3 days, with a 14-day follow-up. There were no safety-related treatment discontinuations or serious adverse events. The adverse events and laboratory abnormalities observed for IDX184- and placebo-treated patients were similar. At the end of the 3-day treatment period, changes from baseline in HCV RNA levels (means ± standard deviations) were -0.5 ± 0.6, -0.7 ± 0.2, -0.6 ± 0.3, and -0.7 ± 0.5 log(10) for the 25-, 50-, 75-, and 100-mg doses, respectively, while viral load remained unchanged for the pooled placebo patients (-0.05 ± 0.3 log(10)). Patients with genotype-1a and patients with genotype-1b responded similarly. Serum ALT levels decreased, especially at daily doses ≥ 75 mg. During the posttreatment period, plasma viremia and serum aminotransferase levels returned to near pretreatment levels. No resistance mutations associated with IDX184 were detected. Plasma exposure of IDX184 and its nucleoside metabolite 2'-MeG was dose related and low. Changes in plasma viral load correlated with plasma exposure of 2'-MeG. In conclusion, the results from this proof-of-concept study show that small doses of the liver-targeted prodrug IDX184 were able to deliver significant antiviral activity and support further clinical evaluation of the drug candidate.


Subject(s)
Antiviral Agents/therapeutic use , Guanosine Monophosphate/analogs & derivatives , Hepatitis C, Chronic/drug therapy , Liver/drug effects , Nucleic Acid Synthesis Inhibitors , Adolescent , Adult , Aged , Alanine Transaminase/blood , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Area Under Curve , Demography , Double-Blind Method , Drug Delivery Systems , Female , Guanosine Monophosphate/adverse effects , Guanosine Monophosphate/pharmacokinetics , Guanosine Monophosphate/therapeutic use , Half-Life , Hepatitis C, Chronic/virology , Humans , Liver/metabolism , Male , Middle Aged , Prodrugs , RNA, Viral/blood , RNA, Viral/genetics , Viral Load , Young Adult
4.
J Virol Methods ; 225: 76-86, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26391876

ABSTRACT

The hepatitis C virus (HCV) NS3/4A protease is a key target of efforts to develop direct-acting antiviral inhibitors for treatment of chronic HCV infection. In vitro analyses of the effects of NS3/4A mutations and polymorphisms on protease inhibitor (PI) susceptibility are essential to nonclinical and clinical compound characterization, but can be hampered by time and technical limitations of current in vitro methods using replicon or purified protein systems. We have developed a fast and simple method utilizing full-length NS3/4A protease inducibly expressed in Escherichia coli cells. Minimally processed E. coli whole cell lysate was used for analyzing NS3/4A protease activity and inhibition by antiviral compounds. Assay conditions were optimized to develop a reproducible assay that can be used for efficient analysis of NS3 protease mutants with poor replication capacity in the replicon system. IC50 fold-changes for NS3 mutants relative to their wild-types generated by this NS3 assay are comparable to those observed in the replicon system, with an R(2) of 0.82 for the values obtained by the two methods. In addition, we demonstrate that this assay can be successfully used for population and clonal phenotyping of patient samples and characterization of PIs against the NS3/4A protease from HCV genotypes 1-6.


Subject(s)
Hepacivirus/enzymology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Antiviral Agents/metabolism , Escherichia coli/genetics , Hepatitis C/virology , Humans , Inhibitory Concentration 50 , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protease Inhibitors/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproducibility of Results , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
5.
Antivir Ther ; 18(6): 755-64, 2013.
Article in English | MEDLINE | ID: mdl-23439365

ABSTRACT

BACKGROUND: IDX184 is a liver-targeted nucleotide prodrug that selectively inhibits HCV NS5B polymerase. METHODS: This randomized, double-blind, placebo-controlled, ascending-dose study investigated the antiviral activity, safety and pharmacokinetics of IDX184 plus pegylated interferon-α2a and ribavirin (P/R) in treatment-naive patients with genotype-1 HCV. A total of 81 patients with baseline HCV RNA≥5 log10 IU/ml, alanine aminotransferase ≤3× upper limit of normal and compensated liver disease were dosed. Sequential cohorts of 20 patients, randomized 16:4 (active:placebo), received IDX184 for 14 days at rising daily doses of 50, 100, 150 or 200 mg in combination with P/R for 14 days. RESULTS: At the end of triple dosing, HCV RNA changes from baseline (mean ±sd log10) and proportion of patients achieving undetectable viral load (<15 IU/ml) based on the efficacy-evaluable population were -2.7 ±1.3 (13%), -4.0 ±1.7 (50%), -4.2 ±1.9 (50%), -4.1 ±1.2 (40%), -4.3 ±1.5 (29%) and -3.7 ±1.2 (25%) for the 50 mg once daily, 50 mg twice daily, 100 mg once daily, 150 mg once daily, 100 mg twice daily and 200 mg once daily IDX184 doses, respectively. P/R alone resulted in a reduction of -1.5 ±1.3 log10 with only 6% of patients with undetectable viral load. Patients with genotypes-1a or -1b responded similarly. No viral breakthrough or resistance associated with IDX184 was observed. Anti-HCV activity of IDX184 correlated with plasma exposure of its nucleoside metabolite 2'-methylguanosine. Most adverse events were mild or moderate in severity and were consistent with those associated with P/R. The most common adverse events were fatigue and headache. CONCLUSIONS: IDX184 in combination with P/R for 14 days was well tolerated and demonstrated greater antiviral activity with more patients achieving undetectable viral load than P/R.


Subject(s)
Antiviral Agents/therapeutic use , Guanosine Monophosphate/analogs & derivatives , Hepatitis C, Chronic/drug therapy , Interferon-alpha/therapeutic use , Polyethylene Glycols/therapeutic use , Ribavirin/therapeutic use , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Drug Interactions , Drug Therapy, Combination , Female , Genotype , Guanosine Monophosphate/administration & dosage , Guanosine Monophosphate/adverse effects , Guanosine Monophosphate/pharmacokinetics , Guanosine Monophosphate/therapeutic use , Hepatitis C/genetics , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/virology , Humans , Interferon-alpha/administration & dosage , Interferon-alpha/adverse effects , Interferon-alpha/pharmacokinetics , Interferons , Interleukins/genetics , Male , Middle Aged , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics , Polymorphism, Genetic , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/therapeutic use , Ribavirin/administration & dosage , Ribavirin/adverse effects , Ribavirin/pharmacokinetics , Treatment Outcome , Viral Load
6.
Antivir Ther ; 17(4): 633-42, 2012.
Article in English | MEDLINE | ID: mdl-22427481

ABSTRACT

BACKGROUND: The addition of direct-acting antivirals to pegylated interferon-α plus ribavirin for the treatment of chronic HCV infection can result in an increased sustained viral response rate and may permit reduction in treatment duration. IDX320 is a potent non-covalent macrocyclic inhibitor of the HCV NS3/4A protease. METHODS: This was a randomized double-blind placebo-controlled single- and multiple-dose study to assess the safety, tolerability, antiviral activity and pharmacokinetics of IDX320 in healthy volunteers (HV) and patients with chronic HCV genotype 1 infection. HV (n=48) received single or multiple ascending doses of IDX320. Two HCV-infected patients received a single dose of 200 mg IDX320. Dosages for other HCV-infected patients were as follows: placebo, 50, 100, 200 or 400 mg of IDX320 orally once daily for 3 days (n=30) or placebo/200 mg of IDX320 twice-daily for 3 days (n=8). RESULTS: In total, 48 HV and 40 HCV-infected patients were enrolled and all completed the study. There were no serious adverse events. The majority of adverse events were of mild or moderate intensity. Pharmacokinetics supported a once-daily dosing regimen. A rapid decline in plasma HCV RNA was observed in all patients. In the multiple-dose study, mean HCV RNA reductions were 2.6, 3.1, 3.1, 3.3 and 3.8 log(10) IU/ml after 3 days in the IDX320 50, 100, 200, 400 mg once-daily and 200 mg twice-daily treatment groups, respectively. This compared to a mean HCV RNA reduction of 0.04 log(10) in the placebo group. CONCLUSIONS: Once-daily IDX320 dosing demonstrated potent dose-dependent antiviral activity in treatment-naive HCV genotype-1-infected patients.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/enzymology , Hepatitis C, Chronic/drug therapy , Macrocyclic Compounds/therapeutic use , Protease Inhibitors/therapeutic use , RNA, Viral/blood , Adolescent , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Viral/drug effects , Genotype , Half-Life , Hepacivirus/drug effects , Hepacivirus/genetics , Humans , Macrocyclic Compounds/administration & dosage , Macrocyclic Compounds/blood , Macrocyclic Compounds/pharmacokinetics , Male , Middle Aged , Protease Inhibitors/administration & dosage , Protease Inhibitors/blood , Protease Inhibitors/pharmacokinetics , Young Adult
7.
J Virol ; 80(8): 3833-43, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16571800

ABSTRACT

Levels of the p53 tumor suppressor protein are increased in human cytomegalovirus (HCMV)-infected cells and may be important for HCMV pathogenesis. In normal cells p53 levels are kept low due to an autoregulatory feedback loop where p53 activates the transcription of mdm2 and mdm2 binds and ubiquitinates p53, targeting p53 for proteasomal degradation. Here we report that, in contrast to uninfected cells, mdm2 was undetectable upon treatment of infected fibroblasts with the proteasome inhibitor MG132. Cellular depletion of mdm2 was reproducible in p53-null cells transfected with the HCMV IE2-86 protein, but not with IE172, independently of the endogenous mdm2 promoter. IE2-86 also prevented the emergence of presumably ubiquitinated species of p53. The regions of IE2-86 important for mdm2 depletion were those containing the sequences corresponding to the putative zinc finger and C-terminal acidic motifs. mdm2 and IE2-86 coimmunoprecipitated in transfected and infected cell lysates and in a cell-free system. IE2-86 blocked mdm2's p53-independent transactivation of the cyclin A promoter in transient-transfection experiments. Pulse-chase experiments revealed that IE2-86 but not IE1-72 or several loss-of-function IE2-86 mutants increased the half-life of p53 and reduced the half-life of mdm2. Short interfering RNA-mediated depletion of IE2-86 restored the ability of HCMV-infected cells to accumulate mdm2 in response to proteasome inhibition. Taken together, the data suggest that specific interactions between IE2-86 and mdm2 cause proteasome-independent degradation of mdm2 and that this may be important for the accumulation of p53 in HCMV-infected cells.


Subject(s)
Immediate-Early Proteins/physiology , Proto-Oncogene Proteins c-mdm2/metabolism , Trans-Activators/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Cricetinae , Cyclin A/genetics , Half-Life , Humans , Leupeptins/pharmacology , Molecular Sequence Data , Promoter Regions, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Ubiquitin/metabolism
8.
J Biol Chem ; 278(25): 22828-37, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12676951

ABSTRACT

We have undertaken a study of the yeast cullin family members Cul3 and Cul8, as little is known about their biochemical and physiological functions. We demonstrate that these cullins are associated in vivo with ubiquitin ligase activity. We show that Cul3 and Cul8 are functionally distinct from Cdc53 and do not interact with ySkp1, suggesting that they target substrates by Skp1- and possibly F-box protein-independent mechanisms. Whereas null mutants of CUL3 appear normal, yeast cells lacking CUL8 have a slower growth rate and are delayed in their progress through anaphase. The anaphase delay phenotype can be complemented by ectopic expression of Cul8 but not by any other yeast or human cullins, nor by a cul8 mutant deficient in binding to RING finger protein Roc1. Deletion of the RAD9 gene suppressed the anaphase delay phenotype of cul8delta, suggesting that loss of Cul8 function may compromise genomic integrity. These results indicate that in addition to the anaphase promoting complex, mitotic progression may involve another E3 ubiquitin ligase mediated by Cul8 protein.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cullin Proteins , Ligases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Amino Acid Sequence , Anaphase , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Genetic Complementation Test , Genotype , Ligases/chemistry , Ligases/genetics , Molecular Sequence Data , Mutagenesis , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/radiation effects , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL