Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 48(9): 3498-507, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15328117

ABSTRACT

Entecavir (ETV) exhibits potent antiviral activity in patients chronically infected with wild-type or lamivudine (3TC)-resistant (3TC(r)) hepatitis B virus (HBV). Among the patients treated in phase II ETV clinical trials, two patients for whom previous therapies had failed exhibited virologic breakthrough while on ETV. Isolates from these patients (arbitrarily designated patients A and B) were analyzed genotypically for emergent substitutions in HBV reverse transcriptase (RT) and phenotypically for reduced susceptibility in cultures and in HBV polymerase assays. After 54 weeks of 3TC therapy, patient A (AI463901-A) received 0.5 mg of ETV for 52 weeks followed by a combination of ETV and 100 mg of 3TC for 89 weeks. Viral rebound occurred at 133 weeks after ETV was started. The 3TC(r) RT substitutions rtV173L, rtL180M, and rtM204V were present at study entry, and the additional substitutions rtI169T and rtM250V emerged during ETV-3TC combination treatment. Reduced ETV susceptibility in vitro required the rtM250V substitution in addition to the 3TC(r) substitutions. For liver transplant patient B (AI463015-B), previous famciclovir, ganciclovir, foscarnet, and 3TC therapies had failed, and RT changes rtS78S/T, rtV173L, rtL180M, rtT184S, and rtM204V were present at study entry. Viral rebound occurred after 76 weeks of therapy with ETV at 1.0 mg, with the emergence of rtT184G, rtI169T, and rtS202I substitutions within the preexisting 3TC(r) background. Reduced susceptibility in vitro was highest when both the rtT184G and the rtS202I changes were combined with the 3TC(r) substitutions. In summary, infrequent ETV resistance can emerge during prolonged therapy, with selection of additional RT substitutions within a 3TC(r) HBV background, leading to reduced ETV susceptibility and treatment failure.


Subject(s)
Antiviral Agents/pharmacology , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Lamivudine/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adult , Amino Acid Substitution/genetics , Antiviral Agents/therapeutic use , Capsid Proteins/biosynthesis , Capsid Proteins/genetics , Cell Line, Tumor , Cells, Cultured , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Viral , Genotype , Guanine/therapeutic use , Hepatitis B/drug therapy , Hepatitis B/virology , Hepatitis B virus/enzymology , Humans , Lamivudine/therapeutic use , Male , Middle Aged , Mutation , Phenotype , RNA-Directed DNA Polymerase/genetics , Reverse Transcriptase Inhibitors/therapeutic use , Treatment Failure , Virus Replication/drug effects , Virus Replication/genetics
2.
Antimicrob Agents Chemother ; 46(8): 2525-32, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12121928

ABSTRACT

Entecavir (ETV) is a potent and selective inhibitor of hepatitis B virus (HBV) replication in vitro and in vivo that is currently in clinical trials for the treatment of chronic HBV infections. A major limitation of the current HBV antiviral therapy, lamivudine (3TC), is the emergence of drug-resistant HBV in a majority of treated patients due to specific mutations in the nucleotide binding site of HBV DNA polymerase (HBV Pol). To determine the effects of 3TC resistance mutations on inhibition by ETV triphosphate (ETV-TP), a series of in vitro studies were performed. The inhibition of wild-type and 3TC-resistant HBV Pol by ETV-TP was measured using recombinant HBV nucleocapsids, and compared to that of 3TC-TP. These enzyme inhibition studies demonstrated that ETV-TP is a highly potent inhibitor of wild-type HBV Pol and is 100- to 300-fold more potent than 3TC-TP against 3TC-resistant HBV Pol. Cell culture assays were used to gauge the potential for antiviral cross-resistance of 3TC-resistant mutants to ETV. Results demonstrated that ETV inhibited the replication of 3TC-resistant HBV, but 20- to 30-fold higher concentrations were required. To gain further perspective regarding the potential therapeutic use of ETV, its phosphorylation was examined in hepatoma cells treated with extracellular concentrations representative of drug levels in plasma in ETV-treated patients. At these concentrations, intracellular ETV-TP accumulated to levels expected to inhibit the enzyme activity of both wild-type and 3TC-resistant HBV Pol. These findings are predictive of potent antiviral activity of ETV against both wild-type and 3TC-resistant HBV.


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed DNA Polymerase/biosynthesis , Guanine/analogs & derivatives , Guanine/pharmacology , Hepatitis B virus/drug effects , Lamivudine/pharmacology , Virus Replication/drug effects , Antiviral Agents/metabolism , Blotting, Southern , Capsid Proteins/metabolism , Cells, Cultured , DNA-Directed DNA Polymerase/genetics , Drug Resistance, Microbial , Genes, Viral/genetics , Genome, Viral , Guanine/metabolism , Humans , Kinetics , Phosphorylation , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
3.
J Infect Dis ; 184(10): 1236-45, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11679911

ABSTRACT

Entecavir (ETV) is a guanosine nucleoside analogue with potent antiviral efficacy in woodchucks chronically infected with woodchuck hepatitis virus. To explore the consequences of prolonged virus suppression, woodchucks received ETV orally for 8 weeks and then weekly for 12 months. Of the 6 animals withdrawn from therapy and monitored for an additional 28 months, 3 had a sustained antiviral response and had no evidence of hepatocellular carcinoma (HCC). Of the 6 animals that continued on a weekly ETV regimen for an additional 22 months, 4 exhibited serum viral DNA levels near the lower limit of detection for >2 years and had no evidence of HCC. Viral antigens and covalently closed circular DNA levels in liver samples were significantly reduced in all animals. ETV was well tolerated, and there was no evidence of resistant variants. On the basis of historical data, long-term ETV treatment appeared to significantly prolong the life of treated animals and delay the emergence of HCC.


Subject(s)
Antiviral Agents/therapeutic use , Guanine/analogs & derivatives , Guanine/therapeutic use , Hepatitis B Virus, Woodchuck , Hepatitis B, Chronic/drug therapy , Marmota , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , DNA, Circular/analysis , DNA, Viral/blood , Disease Models, Animal , Hepatitis B Surface Antigens/blood , Hepatitis B Virus, Woodchuck/genetics , Hepatitis B Virus, Woodchuck/immunology , Hepatitis B Virus, Woodchuck/isolation & purification , Hepatitis B, Chronic/pathology , Humans , Liver/immunology , Liver/virology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Time Factors , Virus Replication/drug effects
4.
Antimicrob Agents Chemother ; 44(9): 2319-26, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10952574

ABSTRACT

BMS-232632 is an azapeptide human immunodeficiency virus (HIV) type 1 (HIV-1) protease inhibitor that displays potent anti-HIV-1 activity (50% effective concentration [EC(50)], 2.6 to 5.3 nM; EC(90), 9 to 15 nM). In vitro passage of HIV-1 RF in the presence of inhibitors showed that BMS-232632 selected for resistant variants more slowly than nelfinavir or ritonavir did. Genotypic and phenotypic analysis of three different HIV strains resistant to BMS-232632 indicated that an N88S substitution in the viral protease appeared first during the selection process in two of the three strains. An I84V change appeared to be an important substitution in the third strain used. Mutations were also observed at the protease cleavage sites following drug selection. The evolution to resistance seemed distinct for each of the three strains used, suggesting multiple pathways to resistance and the importance of the viral genetic background. A cross-resistance study involving five other protease inhibitors indicated that BMS-232632-resistant virus remained sensitive to saquinavir, while it showed various levels (0. 1- to 71-fold decrease in sensitivity)-of cross-resistance to nelfinavir, indinavir, ritonavir, and amprenavir. In reciprocal experiments, the BMS-232632 susceptibility of HIV-1 variants selected in the presence of each of the other HIV-1 protease inhibitors showed that the nelfinavir-, saquinavir-, and amprenavir-resistant strains of HIV-1 remained sensitive to BMS-232632, while indinavir- and ritonavir-resistant viruses displayed six- to ninefold changes in BMS-232632 sensitivity. Taken together, our data suggest that BMS-232632 may be a valuable protease inhibitor for use in combination therapy.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease/metabolism , HIV-1/drug effects , Oligopeptides/pharmacology , Pyridines/pharmacology , Amino Acid Sequence , Atazanavir Sulfate , Drug Resistance, Microbial/physiology , Drug Resistance, Multiple/physiology , HIV Protease/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Mutation , Sequence Homology, Amino Acid , Substrate Specificity
5.
Antimicrob Agents Chemother ; 44(8): 2093-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10898681

ABSTRACT

BMS-232632 is an azapeptide human immunodeficiency virus type 1 (HIV-1) protease (Prt) inhibitor that exhibits potent anti-HIV activity with a 50% effective concentration (EC(50)) of 2.6 to 5.3 nM and an EC(90) of 9 to 15 nM in cell culture. Proof-of-principle studies indicate that BMS-232632 blocks the cleavage of viral precursor proteins in HIV-infected cells, proving that it functions as an HIV Prt inhibitor. Comparative studies showed that BMS-232632 is generally more potent than the five currently approved HIV-1 Prt inhibitors. Furthermore, BMS-232632 is highly selective for HIV-1 Prt and exhibits cytotoxicity only at concentrations 6,500- to 23, 000-fold higher than that required for anti-HIV activity. To assess the potential of this inhibitor when used in combination with other antiretrovirals, BMS-232632 was evaluated for anti-HIV activity in two-drug combination studies. Combinations of BMS-232632 with either stavudine, didanosine, lamivudine, zidovudine, nelfinavir, indinavir, ritonavir, saquinavir, or amprenavir in HIV-infected peripheral blood mononuclear cells yielded additive to moderately synergistic antiviral effects. Importantly, combinations of drug pairs did not result in antagonistic anti-HIV activity or enhanced cytotoxic effects at the highest concentrations used for antiviral evaluation. Our results suggest that BMS-232632 may be an effective HIV-1 inhibitor that may be utilized in a variety of different drug combinations.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Oligopeptides/pharmacology , Pyridines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Atazanavir Sulfate , Blood Proteins , Cells, Cultured , Drug Combinations , Drug Interactions , Gene Products, gag/metabolism , Humans , In Vitro Techniques , Microbial Sensitivity Tests , Protein Precursors/metabolism
6.
J Virol ; 74(2): 851-63, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623748

ABSTRACT

Hepatitis C virus (HCV) NS5B protein possesses an RNA-dependent RNA polymerase (RdRp) activity, a major function responsible for replication of the viral RNA genome. To further characterize the RdRp activity, NS5B proteins were expressed from recombinant baculoviruses, purified to near homogeneity, and examined for their ability to synthesize RNA in vitro. As a result, a highly active NS5B RdRp (1b-42), which contains an 18-amino acid C-terminal truncation resulting from a newly created stop codon, was identified among a number of independent isolates. The RdRp activity of the truncated NS5B is comparable to the activity of the full-length protein and is 20 times higher in the presence of Mn(2+) than in the presence of Mg(2+). When a 384-nucleotide RNA was used as the template, two major RNA products were synthesized by 1b-42. One is a complementary RNA identical in size to the input RNA template (monomer), while the other is a hairpin dimer RNA synthesized by a "copy-back" mechanism. Substantial evidence derived from several experiments demonstrated that the RNA monomer was synthesized through de novo initiation by NS5B rather than by a terminal transferase activity. Synthesis of the RNA monomer requires all four ribonucleotides. The RNA monomer product was verified to be the result of de novo RNA synthesis, as two expected RNA products were generated from monomer RNA by RNase H digestion. In addition, modification of the RNA template by the addition of the chain terminator cordycepin at the 3' end did not affect synthesis of the RNA monomer but eliminated synthesis of the self-priming hairpin dimer RNA. Moreover, synthesis of RNA on poly(C) and poly(U) homopolymer templates by 1b-42 NS5B did not require the oligonucleotide primer at high concentrations (>/=50 microM) of GTP and ATP, further supporting a de novo initiation mechanism. These findings suggest that HCV NS5B is able to initiate RNA synthesis de novo.


Subject(s)
Hepacivirus/enzymology , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cations, Divalent , Cations, Monovalent , Cell Line , Hepacivirus/genetics , Humans , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Templates, Genetic , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/isolation & purification
7.
Antiviral Res ; 48(3): 197-203, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11164506

ABSTRACT

Lobucavir (BMS-180194), a cyclobutyl-guanosine nucleoside analogue, effectively reduced WHV-viremia in chronically infected carrier woodchucks (Marmota monax) by daily per os treatment. WHV-viremia in the animals was measured by the serum content of hybridizable WHV-genomic DNA. Lobucavir, given at daily doses of 10 and 20 mg/kg body weight, reduced WHV-viremia by a 10- to 200-fold range during therapy. Lobucavir, given at 5 mg/kg, suppressed WHV-viremia by a 10- to 30-fold range, whereas a 0.5 mg/kg dose had no significant effect. WHV-viremia was also measured by hepadnaviral endogenous polymerase activity (EPA) in sera of animals treated for 6 weeks at 5 and 0.5 mg/kg. Changes in EPA in sera of lobucavir treated animals were comparable to changes in WHV DNA levels. Viremia in treated carriers recrudesced to pretreatment levels by 2 weeks of therapy cessation. These results indicated that the minimally effective antiviral daily per os dose of lobucavir in WHV-carrier woodchucks was approximately 5 mg/kg.


Subject(s)
DNA, Viral/blood , Guanine/analogs & derivatives , Guanine/therapeutic use , Hepatitis B Virus, Woodchuck/drug effects , Hepatitis B, Chronic/virology , Reverse Transcriptase Inhibitors/therapeutic use , Animals , Disease Models, Animal , Guanine/pharmacology , Hepatitis B, Chronic/drug therapy , Humans , Marmota , Reverse Transcriptase Inhibitors/pharmacology , Viremia/virology
8.
J Virol ; 73(3): 1785-94, 1999 Mar.
Article in English | MEDLINE | ID: mdl-9971755

ABSTRACT

The hemagglutinin (HA) protein undergoes a low-pH-induced conformational change in the acidic milieu of the endosome, resulting in fusion of viral and cellular membranes. A class of compounds that specifically interact with the HA protein of H1 and H2 subtype viruses and inhibit this conformational change was recently described (G. X. Luo et al., Virology 226:66-76, 1996, and J. Virol. 71:4062-4070, 1997). In this study, purified HA trimers (bromelain-cleaved HA [BHA]) are used to examine the properties and binding characteristics of these inhibitors. Compounds were able to inhibit the low-pH-induced change of isolated trimers, as detected by resistance to digestion with trypsin. Protection from digestion was extremely stable, as BHA-inhibitor complexes could be incubated for 24 h in low pH with almost no change in BHA structure. One inhibitor was prepared as a radiolabeled photoaffinity analog and used to probe for specific drug interactions with the HA protein. Analysis of BHA after photoaffinity analog binding and UV cross-linking revealed that the HA2 subunit of the HA was specifically radiolabeled. Cross-linking of the photoaffinity analog to BHA under neutral (native) pH conditions identified a stretch of amino acids within the alpha-helix of HA2 that interact with the inhibitor. Interestingly, cross-linking of the analog under acidic conditions identified a different region within the HA2 N terminus which interacts with the photoaffinity compound. These attachment sites help to delineate a potential binding pocket and suggest a model whereby the BHA is able to undergo a partial, reversible structural change in the presence of inhibitor compound.


Subject(s)
Antiviral Agents/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Photoaffinity Labels/metabolism , Amino Acid Sequence , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hydrogen-Ion Concentration , Peptide Mapping , Protein Conformation , Quinolizines/pharmacology
9.
Antimicrob Agents Chemother ; 43(1): 190-3, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9869593

ABSTRACT

BMS-200475 was recently shown to have potent antiviral activity against hepatitis B virus (50% effective concentration = 3.7 nM; 50% cytotoxic concentration = 30 microM). In metabolic studies in both HepG2 and hepatitis B virus-transfected 2.2.15 human hepatoma cell lines, the metabolism was similar, the primary products being the di- and triphosphates. The accumulation of triphosphate was rapid and detectable down to a 5 nM concentration of added drug. When cells were labeled at 25 microM, the intracellular triphosphate concentration attained 30 pmol/10(6) cells ( approximately 30 microM). The intracellular half-life of the triphosphate was about 15 h. Compared with five other nucleoside analogs of medical interest (lamivudine, penciclovir, ganciclovir, acyclovir, and lobucavir), BMS-200475 was most efficiently phosphorylated to the triphosphate in HepG2 cells.


Subject(s)
Antiviral Agents/metabolism , Deoxyguanosine/analogs & derivatives , Hepatitis B virus/metabolism , Antiviral Agents/pharmacology , Deoxyguanosine/metabolism , Deoxyguanosine/pharmacology , Half-Life , Hepatitis B virus/drug effects , Humans , Phosphorylation , Spectrophotometry, Ultraviolet , Transfection , Tumor Cells, Cultured
10.
Antivir Ther ; 4(1): 21-8, 1999.
Article in English | MEDLINE | ID: mdl-10682125

ABSTRACT

The current report summarizes the available published and unpublished data from several investigators on resistance in clinical isolates following prolonged stavudine therapy. Results suggest that stavudine resistance is both modest in degree and infrequent in appearance. Phenotypic evaluation of 61 patients on stavudine therapy showed only modest changes in drug sensitivity following up to 29 months of treatment. The post-treatment isolates from 15 patients exhibited an increase in EC50 value > fourfold (level above variability of assay) when compared with the corresponding pretreatment isolates. However, the vast majority (11) of these pretreatment isolates either had unexpectedly low EC50 levels and/or had post-treatment isolates that lacked any amino acid changes within their reverse transcriptase (RT) gene to account for the observed change in sensitivity. Of the four remaining isolates, two appeared to have a multi-resistant phenotype to several nucleoside analogues and two had no detectable RT amino acid changes to account for the observed change in stavudine sensitivity. To date, clinical HIV-1 isolates displaying stavudine-specific resistance have yet to be reported. Furthermore, full or partial RT sequence analysis of 194 post-treatment isolates failed to identify any consistent amino acid changes. The strain-specific V75T mutation reported to confer stavudine resistance to the HXB2 HIV-1 strain in vitro, was found in only six isolates and did not correlate with stavudine resistance. This low incidence of stavudine resistance is in striking contrast to that observed with other nucleoside analogues and further supports the use of stavudine in first-line combination therapy for HIV patients.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/therapeutic use , Stavudine/therapeutic use , Drug Resistance , HIV-1/drug effects , Humans , Phenotype , RNA-Directed DNA Polymerase/genetics
11.
Antimicrob Agents Chemother ; 42(12): 3200-8, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9835515

ABSTRACT

The guanosine analogs BMS-200475 and lobucavir have previously been shown to effectively suppress propagation of the human hepatitis B virus (HBV) and woodchuck hepatitis virus (WHV) in 2.2.15 liver cells and in the woodchuck animal model system, respectively. This repression was presumed to occur via inhibition of the viral polymerase (Pol) by the triphosphate (TP) forms of BMS-200475 and lobucavir which are both produced in mammalian cells. To determine the exact mode of action, BMS-200475-TP and lobucavir-TP, along with several other guanosine analog-TPs and lamivudine-TP were tested against the HBV, WHV, and duck hepatitis B virus (DHBV) polymerases in vitro. Estimates of the 50% inhibitory concentrations revealed that BMS-200475-TP and lobucavir-TP inhibited HBV, WHV, and DHBV Pol comparably and were superior to the other nucleoside-TPs tested. More importantly, both analogs blocked the three distinct phases of hepadnaviral replication: priming, reverse transcription, and DNA-dependent DNA synthesis. These data suggest that the modest potency of lobucavir in 2.2.15 cells may be the result of poor phosphorylation in vivo. Kinetic studies revealed that BMS-200475-TP and lobucavir-TP competitively inhibit HBV Pol and WHV Pol with respect to the natural dGTP substrate and that both drugs appear to bind to Pol with very high affinities. Endogenous sequencing reactions conducted in replicative HBV nucleocapsids suggested that BMS-200475-TP and lobucavir-TP are nonobligate chain terminators that stall Pol at sites that are distinct yet characteristically two to three residues downstream from dG incorporation sites.


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/antagonists & inhibitors , Deoxyguanosine/analogs & derivatives , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Hepadnaviridae/enzymology , Nucleic Acid Synthesis Inhibitors , Deoxyguanosine/pharmacology , Guanine/pharmacology , Hepatitis B Virus, Duck/drug effects , Hepatitis B Virus, Duck/enzymology , Hepatitis B Virus, Woodchuck/drug effects , Hepatitis B Virus, Woodchuck/enzymology , Hepatitis B virus/drug effects , Hepatitis B virus/enzymology , Humans , Kinetics , Phosphates , RNA-Directed DNA Polymerase/metabolism , Structure-Activity Relationship
12.
Antimicrob Agents Chemother ; 42(12): 3209-17, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9835516

ABSTRACT

Daily oral treatment with the cyclopentyl 2'-deoxyguanosine nucleoside BMS-200475 at doses ranging from 0.02 to 0.5 mg/kg of body weight for 1 to 3 months effectively reduced the level of woodchuck hepatitis virus (WHV) viremia in chronically infected woodchucks as measured by reductions in serum WHV DNA levels and endogenous hepadnaviral polymerase activity. Within 4 weeks of daily therapy with 0.5 or 0.1 mg of BMS-200475 per kg, endogenous viral polymerase levels in serum were reduced about 1,000-fold compared to pretreatment levels. Serum WHV DNA levels determined by a dot blot hybridization technique were comparably decreased in these treated animals. In the 3-month study, the sera of animals that had undetectable levels of WHV DNA by the dot blot technique were further analyzed by a highly sensitive semiquantitative PCR assay. The results indicate that BMS-200475 therapy reduced mean WHV titers by 10(7)- to 10(8)-fold, down to levels as low as 10(2) to 10(3) virions/ml of serum. Southern blot hybridization analysis of liver biopsy samples taken from animals during and after BMS-200475 treatment showed remarkable reductions in the levels of WHV DNA replicative intermediates and in the levels of covalently closed circular viral DNA. WHV viremia in BMS-200475-treated WHV carriers eventually returned to pretreatment levels after therapy was stopped. These results indicate that BMS-200475 should be evaluated in clinical trials for the therapy of chronic human hepatitis B virus infections.


Subject(s)
Antiviral Agents/therapeutic use , Deoxyguanosine/analogs & derivatives , Hepatitis B Virus, Woodchuck , Hepatitis B/drug therapy , Animals , Antiviral Agents/administration & dosage , DNA, Viral/blood , DNA, Viral/metabolism , Deoxyguanosine/administration & dosage , Deoxyguanosine/therapeutic use , Hepatitis B/virology , Hepatitis B Virus, Woodchuck/metabolism , Liver/metabolism , Liver/virology , Marmota , Polymerase Chain Reaction , Time Factors
14.
Antimicrob Agents Chemother ; 41(7): 1444-8, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9210663

ABSTRACT

BMS-200475 is a novel carbocyclic 2'-deoxyguanosine analog found to possess potent and selective anti-hepatitis B virus (anti-HBV) activity. BMS-200475 is distinguished from guanosine by replacement of the natural furanose oxygen on the sugar moiety with an exo carbon-carbon double bond. In the HepG2 stably transfected cell line 2.2.15, BMS-200475 had a 50% effective concentration (EC50) of 3.75 nM against HBV, as determined by analysis of secreted HBV DNA. Structurally related compounds with adenine, iodouracil, or thymine base substitutions were significantly less potent or were inactive. Direct comparison of the antiviral activities of BMS-200475 with those of a variety of other nucleoside analogs, including lamivudine (EC50 = 116.26 nM), demonstrated the clearly superior in vitro potency of BMS-200475 in 2.2.15 cells. Intracellular HBV replicative intermediates were uniformly reduced when cells were treated with BMS-200475, but rebounded after treatment was terminated. The concentration of BMS-200475 causing 50% cytotoxicity in 2.2.15 cell cultures was 30 microM, approximately 8,000-fold greater than the concentration required to inhibit HBV replication in the same cell line. Treatment with BMS-200475 resulted in no apparent inhibitory effects on mitochondrial DNA content.


Subject(s)
Antiviral Agents/pharmacology , Deoxyguanosine/analogs & derivatives , Hepatitis B virus/drug effects , Cell Survival/drug effects , DNA, Mitochondrial/drug effects , Deoxyguanosine/pharmacology , Drug Evaluation, Preclinical , Humans , Nucleosides/pharmacology , Tumor Cells, Cultured , Virus Replication/drug effects
15.
Virus Res ; 50(1): 65-75, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9255936

ABSTRACT

The RNA-dependent RNA polymerase of influenza virus transcribes messenger RNA through a unique cap scavenging mechanism. Viral enzyme binds to the cap structure of host mRNA, cleaves the molecule 9-15 bases downstream of the cap, and uses the short capped oligonucleotide as a primer for mRNA synthesis. Previously, we have shown that the viral polymerase can efficiently bind capped RNAs shorter than 9 nucleotides in length, but the viral enzyme can not utilize these RNAs as primers. For this reason, these short capped oligonucleotides are potent inhibitors of influenza virus transcription. In these studies, it is now shown that short capped oligomers inhibit capped-RNA dependent transcription at the initial step of cap binding. In contrast, low concentrations of these short capped RNAs can actually stimulate viral transcription primed with high concentrations of the dinucleotide ApG. Another capped RNA derivative containing phosphorothioate oligonucleotides was also investigated as a potential polymerase inhibitor. This longer capped RNA was able to bind to the polymerase, but could not be cleaved to primer length by the enzyme associated endonuclease. Thus, the capped phosphorothioate RNA inhibited cap-primed transcription at the step of cap binding. However, in contrast to the short capped oligonucleotide, it also inhibited ApG primed viral transcription.


Subject(s)
Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism , RNA, Viral/pharmacology , Thionucleotides/pharmacology , Transcription, Genetic/drug effects , Oligoribonucleotides/pharmacology , Orthomyxoviridae/chemistry , Protein Binding/drug effects , Protein Binding/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/pharmacology , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Substrate Specificity/genetics , Thionucleotides/metabolism
16.
Antimicrob Agents Chemother ; 41(12): 2680-5, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9420038

ABSTRACT

Lobucavir (LBV) is a deoxyguanine nucleoside analog with broad-spectrum antiviral activity. LBV was previously shown to inhibit herpes simplex virus (HSV) DNA polymerase after phosphorylation by the HSV thymidine kinase. Here we determined the mechanism of action of LBV against human cytomegalovirus (HCMV). LBV inhibited HCMV DNA synthesis to a degree comparable to that of ganciclovir (GCV), a drug known to target the viral DNA polymerase. The expression of late proteins and RNA, dependent on viral DNA synthesis, was also inhibited by LBV. Immediate-early and early HCMV gene expression was unaffected, suggesting that LBV acts temporally coincident with HCMV DNA synthesis and not through cytotoxicity. In vitro, the triphosphate of LBV was a potent inhibitor of HCMV DNA polymerase with a Ki of 5 nM. LBV was phosphorylated to its triphosphate form intracellularly in both infected and uninfected cells, with phosphorylated metabolite levels two- to threefold higher in infected cells. GCV-resistant HCMV isolates, with deficient GCV phosphorylation due to mutations in the UL97 protein kinase, remained sensitive to LBV. Overall, these results suggest that LBV-triphosphate halts HCMV DNA replication by inhibiting the viral DNA polymerase and that LBV phosphorylation can occur in the absence of viral factors including the UL97 protein kinase. Furthermore, LBV may be effective in the treatment of GCV-resistant HCMV.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/metabolism , Cytomegalovirus/drug effects , Cytomegalovirus/enzymology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Nucleic Acid Synthesis Inhibitors , Viral Proteins/antagonists & inhibitors , Cells, Cultured , Cytomegalovirus/metabolism , DNA Replication/drug effects , DNA, Viral/biosynthesis , DNA, Viral/drug effects , Drug Resistance, Microbial , Fibroblasts/metabolism , Fibroblasts/virology , Ganciclovir/pharmacology , Guanine/metabolism , Guanine/pharmacology , Humans , Phosphorylation , Viral Proteins/genetics , Viral Proteins/metabolism
17.
J Virol ; 70(7): 4317-28, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8676454

ABSTRACT

The herpes simplex virus type 1 (HSV-1) protease (Pra) and related proteins are involved in the assembly of viral capsids and virion maturation. Pra is a serine protease, and the active-site residue has been mapped to amino acid (aa) 129 (Ser). This 635-aa protease, encoded by the UL26 gene, is autoproteolytically processed at two sites, the release (R) site between amino acid residues 247 and 248 and the maturation (M) site between residues 610 and 611. When the protease cleaves itself at both sites, it releases Nb, the catalytic domain (N0), and the C-terminal 25 aa. ICP35, a substrate of the HSV-1 protease, is the product of the UL26.5 gene. As it is translated from a Met codon within the UL26 gene, ICP35 cd are identical to the C-terminal 329-aa sequence of the protease and are trans cleaved at an identical C-terminal site to generate ICP35 e,f and a 25-aa peptide. Only fully processed Pra (N0 and Nb) and ICP35 (ICP35 e,f) are present in B capsids, which are believed to be precursors of mature virions. Using an R-site mutant A247S virus, we have recently shown that this mutant protease retains enzymatic activity but fails to support viral growth, suggesting that the release of N0 is required for viral replication. Here we report that another mutant protease, with an amino acid substitution (Ser to Cys) at the active site, can complement the A247S mutant but not a protease deletion mutant. Cell lines expressing the active-site mutant protease were isolated and shown to complement the A247S mutant at the levels of capsid assembly, DNA packaging, and viral growth. Therefore, the complementation between the R-site mutant and the active-site mutant reconstituted wild-type Pra function. One feature of this intragenic complementation is that following sedimentation of infected-cell lysates on sucrose gradients, both N-terminally unprocessed and processed proteases were isolated from the fractions where normal B capsids sediment, suggesting that proteolytic processing occurs inside capsids. Our results demonstrate that the HSV-1 protease has distinct functional domains and some of these functions can complement in trans.


Subject(s)
Capsid/metabolism , Herpesvirus 1, Human/enzymology , Serine Endopeptidases/metabolism , Viral Proteins , Animals , Base Sequence , Binding Sites , Cell Line , Chlorocebus aethiops , DNA Primers , Genetic Complementation Test , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Molecular Sequence Data , Mutation , Serine Endopeptidases/genetics , Vero Cells , Virus Replication
18.
Antimicrob Agents Chemother ; 40(6): 1346-51, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8725999

ABSTRACT

Current treatments for human immunodeficiency virus (HIV) include both reverse transcriptase and protease inhibitors. Results from in vitro and clinical studies suggest that combination therapy can be more effective than single drugs in reducing viral burden. To evaluate compounds for combination therapy, stavudine (d4T), didanosine (ddI), or BMS-186,318, an HIV protease inhibitor, were combined with other clinically relevant compounds and tested in a T-cell line (CEM-SS) that was infected with HIV-RF or in peripheral blood mononuclear cells infected with a clinical HIV isolate. The combined drug effects were analyzed by the methods described by Chou and Talalay (Adv. Enzyme Regul. 22:27-55, 1984) as well as by Prichard et al. (Antimicrob. Agents Chemother. 37:540-545, 1993). The results showed that combining two nucleoside analogs (d4T-ddI, d4T-zidovudine [AZT], and d4T-zalcitabine [ddC]), two HIV protease inhibitors (BMS-186,318-saquinavir, BMS-186,318-SC-52151, and BMS-186,318-MK-639) or a reverse transcriptase and a protease inhibitor (BMS-186,318-d4T, BMS-186,318-ddI, BMS-186,318-AZT, d4T-saquinavir, d4T-MK-639, and ddI-MK-639) yielded additive to synergistic antiviral effects. In general, analysis of data by either method gave consistent results. In addition, combined antiviral treatments involving nucleoside analogs gave slightly different outcomes in the two cell types, presumably because of a difference in phosphorylation patterns. Importantly, no strong antagonism was observed with the drug combinations studied. These data should provide useful information for the design of clinical trials of combined chemotherapy.


Subject(s)
HIV/drug effects , Protease Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication/drug effects , Cell Line , Drug Combinations , Drug Interactions , Evaluation Studies as Topic , Humans , Virus Replication/physiology
19.
J Med Chem ; 39(10): 1991-2007, 1996 May 10.
Article in English | MEDLINE | ID: mdl-8642558

ABSTRACT

A series of novel aminodiol inhibitors of HIV protease based on the lead compound 1 with structural modifications at P1' were synthesized in order to reduce the cytotoxicity of 1. We have observed a high degree of correlation between the lipophilicity and cytotoxicity of this series of inhibitors. It was found that appropriate substitution at the para position of the P1' phenyl group of 1 resulted in the identification of equipotent (both against the enzyme and in cell culture) compounds (10l, 10m, 10n, and 15c) which possess significantly decreased cytotoxicity.


Subject(s)
Amines/chemical synthesis , HIV Protease Inhibitors/chemical synthesis , Amines/chemistry , Amines/pharmacology , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Humans , Structure-Activity Relationship
20.
Proc Natl Acad Sci U S A ; 93(4): 1648-53, 1996 Feb 20.
Article in English | MEDLINE | ID: mdl-8643685

ABSTRACT

The observed in vitro and in vivo benefit of combination treatment with anti-human immunodeficiency virus (HIV) agents prompted us to examine the potential of resistance development when two protease inhibitors are used concurrently. Recombinant HIV-1 (NL4-3) proteases containing combined resistance mutations associated with BMS-186318 and A-77003 (or saquinavir) were either inactive or had impaired enzyme activity. Subsequent construction of HIV-1 (NL4-3) proviral clones containing the same mutations yielded viruses that were severely impaired in growth or nonviable, confirming that combination therapy may be advantageous. However, passage of BMS-186318-resistant HIV-1 (RF) in the presence of either saquinavir or SC52151, which represented sequential drug treatment, produced viable viruses resistant to both BMS-186318 and the second compound. The predominant breakthrough virus contained the G48V/A71T/V82A protease mutations. The clone-purified RF (G48V/A71T/V82A) virus, unlike the corresponding defective NL4-3 triple mutant, grew well and displayed cross-resistance to four distinct protease inhibitors. Chimeric virus and in vitro mutagenesis studies indicated that the RF-specific protease sequence, specifically the Ile at residue 10, enabled the NL4-3 strain with the triple mutant to grow. Our results clearly indicate that viral genetic background will play a key role in determining whether cross-resistance variants will arise.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , HIV-1/drug effects , Amino Acid Sequence , Carbamates/pharmacology , Clone Cells , DNA Mutational Analysis , DNA, Recombinant/genetics , DNA, Viral/genetics , Drug Administration Schedule , Drug Resistance, Microbial/genetics , Drug Therapy, Combination , Ethanolamines/pharmacology , HIV Protease Inhibitors/administration & dosage , HIV-1/enzymology , HIV-1/genetics , HeLa Cells , Humans , Indinavir , Isoquinolines/pharmacology , Methylurea Compounds/pharmacology , Molecular Sequence Data , Point Mutation , Proviruses/enzymology , Proviruses/genetics , Pyridines/pharmacology , Quinolines/pharmacology , Reassortant Viruses/drug effects , Reassortant Viruses/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Saquinavir , T-Lymphocytes , Urea/analogs & derivatives , Urea/pharmacology , Valine/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...