Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Open Forum Infect Dis ; 10(3): ofad101, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36968959

ABSTRACT

Background: We assessed the efficacy and safety of dolutegravir/lamivudine (DTG/3TC) in a US test-and-treat setting at a secondary 48-week time point of the multicenter, single-arm, phase IIIb STAT study. Methods: Participants were eligible adults newly diagnosed with human immunodeficiency virus (HIV)-1 and had started once-daily DTG/3TC within 14 days of diagnosis, before laboratory results were available. Antiretroviral therapy (ART) was modified if baseline testing indicated DTG or 3TC resistance, hepatitis B virus (HBV) coinfection, or creatinine clearance <30 mL/min per 1.73 m2, and these participants remained in the study. A proportion with HIV-1 ribonucleic acid (RNA) <50 copies/mL at Week 48 was calculated among all participants (intention-to-treat-exposed [ITT-E] missing = failure analysis) and those with available data (observed analysis). Results: At Week 48, 82% of all participants regardless of ART (107 of 131; ITT-E missing = failure) and 97% with available data (107 of 110; observed analysis) achieved HIV-1 RNA <50 copies/mL. High proportions of virologic response were seen overall, including in participants with high viral load (≥500 000 copies/mL; 89%) or low CD4+ cell count (<200 cells/mm3; 78%) at baseline. Ten participants had treatment modification (baseline HBV coinfection, n = 5; participant/proxy decision, n = 2; baseline M184V resistance mutation, adverse event [AE; rash], and pregnancy, n = 1 each) before Week 48. Two participants met confirmed virologic failure criteria. No treatment-emergent resistance was observed. Ten participants reported drug-related AEs (all grade 1-2); no serious drug-related AEs occurred. Conclusions: Results demonstrated high proportions of participants with sustained virologic suppression, no treatment-emergent resistance, and good safety over 48 weeks, supporting first-line use of DTG/3TC in a test-and-treat setting.

2.
AIDS ; 35(12): 1957-1965, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34115650

ABSTRACT

OBJECTIVES: Dolutegravir/lamivudine (DTG/3TC) is indicated for treatment-naive and experienced people with HIV; however, questions remain about its utility in a test-and-treat setting because of potential transmitted resistance and baseline hepatitis B virus (HBV) co-infection. We present feasibility and efficacy of DTG/3TC in newly diagnosed individuals in a test-and-treat setting. DESIGN: The single-arm STAT study evaluated DTG/3TC in a US test-and-treat setting. METHODS: Eligible adults initiated DTG/3TC 14 days or less after HIV-1 diagnosis without availability of baseline laboratory results. If baseline testing indicated DTG or 3TC resistance, HBV co-infection, or creatinine clearance less than 30 ml/min per 1.73 m2, participants remained on study with treatment modification. Efficacy endpoints included proportions of participants with HIV-1 RNA less than 50 copies/ml at Week 24, regardless of antiretroviral regimen, among all participants (intention-to-treat exposed) and those with available HIV-1 RNA data (observed). RESULTS: Of 131 participants enrolled, 8% were female and 50% were non-white. Through Week 24, treatment was modified in eight participants [five with HBV co-infection, one with baseline M184V, one for adverse event (rash), one participant decision]. At Week 24, 78% (102/131) of all participants and 92% (102/111) of those with available data achieved HIV-1 RNA less than 50 copies/ml. Incidence of drug-related adverse events was low (7%); no drug-related serious adverse events occurred. CONCLUSION: These data demonstrate the feasibility, efficacy, and safety of using DTG/3TC as a first-line regimen in a test-and-treat setting, with therapy adjustments for baseline resistance or HBV co-infection occurring safely via routine clinical care as needed [ClinicalTrials.gov, NCT03945981; see Supplemental Digital Content 1, video abstract (Video abstract summarizing the STAT study design and results), http://links.lww.com/QAD/C189].


Subject(s)
Anti-HIV Agents , HIV Infections , HIV-1 , Adult , Anti-HIV Agents/adverse effects , Female , HIV Infections/complications , HIV Infections/drug therapy , Heterocyclic Compounds, 3-Ring/adverse effects , Humans , Lamivudine/adverse effects , Male , Oxazines/therapeutic use , Piperazines/therapeutic use , Pyridones
3.
Antimicrob Agents Chemother ; 59(5): 2596-606, 2015 May.
Article in English | MEDLINE | ID: mdl-25691633

ABSTRACT

The recently approved HIV-1 integrase strand transfer inhibitor (INSTI) dolutegravir (DTG) (S/GSK1349572) has overall advantageous activity when tested in vitro against HIV-1 with raltegravir (RAL) and elvitegravir (EVG) resistance signature mutations. We conducted an in vitro resistance selection study using wild-type HIV-1 and mutants with the E92Q, Y143C, Y143R, Q148H, Q148K, Q148R, and N155H substitutions to assess the DTG in vitro barrier to resistance. No viral replication was observed at concentrations of ≥ 32 nM DTG, whereas viral replication was observed at 160 nM RAL or EVG in the mutants. In the Q148H, Q148K, or Q148R mutants, G140S/Q148H, E138K/Q148K, E138K/Q148R, and G140S/Q148R secondary mutations were identified with each INSTI and showed high resistance to RAL or EVG but limited resistance to DTG. E138K and G140S, as secondary substitutions to Q148H, Q148K, or Q148R, were associated with partial recovery in viral infectivity and/or INSTI resistance. In the E92Q, Y143C, Y143R, and N155H mutants, no secondary substitutions were associated with DTG. These in vitro results suggest that DTG has a high barrier to the development of resistance in the presence of RAL or EVG signature mutations other than Q148. One explanation for this high barrier to resistance is that no additional secondary substitution of E92Q, Y143C, Y143R, or N155H simultaneously increased the fold change in 50% effective concentration (EC50) to DTG and infectivity. Although increased DTG resistance via the Q148 pathway and secondary substitutions occurs at low concentrations, a higher starting concentration may reduce or eliminate the development of DTG resistance in this pathway in vitro.


Subject(s)
Drug Resistance, Viral/genetics , Quinolones/pharmacology , Raltegravir Potassium/pharmacology , HIV Integrase/metabolism , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , HIV-1/metabolism , Heterocyclic Compounds, 3-Ring/pharmacology , Mutation/genetics , Oxazines , Piperazines , Pyridones
4.
Antimicrob Agents Chemother ; 59(1): 397-406, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25367908

ABSTRACT

GSK1265744 is a new HIV integrase strand transfer inhibitor (INSTI) engineered to deliver efficient antiviral activity with a once-daily, low-milligram dose that does not require a pharmacokinetic booster. The in vitro antiviral profile and mechanism of action of GSK1265744 were established through integrase enzyme assays, resistance passage experiments, and cellular assays with site-directed molecular (SDM) HIV clones resistant to other classes of anti-HIV-1 agents and earlier INSTIs. GSK1265744 inhibited HIV replication with low or subnanomolar efficacy and with a selectivity index of at least 22,000 under the same culture conditions. The protein-adjusted half-maximal inhibitory concentration (PA-EC50) extrapolated to 100% human serum was 102 nM. When the virus was passaged in the presence of GSK1265744, highly resistant mutants with more than a 10-fold change (FC) in EC50 relative to that of the wild-type were not observed for up to 112 days of culture. GSK1265744 demonstrated activity against SDM clones containing the raltegravir (RAL)-resistant Y143R, Q148K, N155H, and G140S/Q148H signature variants (FC less than 6.1), while these mutants had a high FC in the EC50 for RAL (11 to >130). Either additive or synergistic effects were observed when GSK1265744 was tested in combination with representative anti-HIV agents, and no antagonistic effects were seen. These findings demonstrate that, similar to dolutegravir, GSK1265744 is differentiated as a new INSTI, having a markedly distinct resistance profile compared with earlier INSTIs, RAL, and elvitegravir (EVG). The collective data set supports further clinical development of GSK1265744.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV-1/drug effects , Pyridones/therapeutic use , Cell Line , Drug Resistance, Viral/genetics , HIV Infections/virology , HIV Integrase/drug effects , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Microbial Sensitivity Tests , Oxazines , Piperazines , Quinolones/therapeutic use , Raltegravir Potassium/therapeutic use , Virus Replication/drug effects
5.
PLoS One ; 8(10): e77448, 2013.
Article in English | MEDLINE | ID: mdl-24146996

ABSTRACT

Signature HIV-1 integrase mutations associated with clinical raltegravir resistance involve 1 of 3 primary genetic pathways, Y143C/R, Q148H/K/R and N155H, the latter 2 of which confer cross-resistance to elvitegravir. In accord with clinical findings, in vitro drug resistance profiling studies with wild-type and site-directed integrase mutant viruses have shown significant fold increases in raltegravir and elvitegravir resistance for the specified viral mutants relative to wild-type HIV-1. Dolutegravir, in contrast, has demonstrated clinical efficacy in subjects failing raltegravir therapy due to integrase mutations at Y143, Q148 or N155, which is consistent with its distinct in vitro resistance profile as dolutegravir's antiviral activity against these viral mutants is equivalent to its activity against wild-type HIV-1. Kinetic studies of inhibitor dissociation from wild-type and mutant integrase-viral DNA complexes have shown that dolutegravir also has a distinct off-rate profile with dissociative half-lives substantially longer than those of raltegravir and elvitegravir, suggesting that dolutegravir's prolonged binding may be an important contributing factor to its distinct resistance profile. To provide a structural rationale for these observations, we constructed several molecular models of wild-type and clinically relevant mutant HIV-1 integrase enzymes in complex with viral DNA and dolutegravir, raltegravir or elvitegravir. Here, we discuss our structural models and the posited effects that the integrase mutations and the structural and electronic properties of the integrase inhibitors may have on the catalytic pocket and inhibitor binding and, consequently, on antiviral potency in vitro and in the clinic.


Subject(s)
HIV Integrase Inhibitors/metabolism , HIV Integrase/genetics , HIV-1/genetics , HIV-1/metabolism , Heterocyclic Compounds, 3-Ring/metabolism , Proviruses/genetics , Drug Resistance, Viral/genetics , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV Long Terminal Repeat/genetics , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Molecular Structure , Nucleic Acid Conformation , Oxazines , Piperazines , Protein Binding , Protein Conformation , Pyridones
6.
J Acquir Immune Defic Syndr ; 61(3): 297-301, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22878423

ABSTRACT

BACKGROUND: Dolutegravir (DTG, S/GSK1349572) is an integrase inhibitor with low nanomolar potency. Susceptibility to dolutegravir and raltegravir was determined for raltegravir-resistant clinical isolates. METHODS: Genotypic and phenotypic susceptibility to integrase inhibitors was examined using 39 clinical isolate samples obtained from 18 adults who had exhibited incomplete viral suppression on a raltegravir-based regimen. RESULTS: Of 39 samples evaluated, 30 had genotypic and phenotypic resistance to raltegravir. All samples lacking raltegravir resistance retained complete susceptibility to dolutegravir. Of the 30 samples with genotypic evidence of raltegravir resistance, the median level of phenotypic resistance to raltegravir was high (median fold change in inhibitory concentration at 50%, >81; range, 3.7 to >87), while the level of resistance to dolutegravir was close to that of wild-type variants (median fold change, 1.5; range, 0.9-19.0). Longitudinal samples from 5 subjects collected during long-term failure of raltegravir revealed time-dependent general decreases in phenotypic susceptibility to raltegravir, with minimal changes in phenotypic susceptibility to dolutegravir. The median fold change to dolutegravir for isolates containing changes at G140S + Q148H, G140S + Q148R, T97A + Y143R, and N155H (thus including raltegravir signature resistance codons) were 3.75, 13.3, 1.05, and 1.37, respectively. CONCLUSIONS: Dolutegravir retained in vitro activity against clinical isolates obtained from subjects who failed raltegravir-based therapy at near wild-type levels for variants containing the Y143 and N155 resistance mutations. Isolates with Q148 plus additional integrase mutations possessed a broader range of and more reduced susceptibility to dolutegravir.


Subject(s)
HIV Infections/drug therapy , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Pyrrolidinones/therapeutic use , Adult , Drug Resistance, Viral , Genotype , HIV Infections/virology , HIV Integrase Inhibitors/therapeutic use , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Humans , Mutation , Oxazines , Phenotype , Piperazines , Pyridones , Raltegravir Potassium
7.
J Virol ; 86(5): 2696-705, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205735

ABSTRACT

Integrase (IN) strand transfer inhibitors (INSTIs) have been developed to inhibit the ability of HIV-1 integrase to irreversibly link the reverse-transcribed viral DNA to the host genome. INSTIs have proven their high efficiency in inhibiting viral replication in vitro and in patients. However, first-generation INSTIs have only a modest genetic barrier to resistance, allowing the virus to escape these powerful drugs through several resistance pathways. Second-generation INSTIs, such as dolutegravir (DTG, S/GSK1349572), have been reported to have a higher resistance barrier, and no novel drug resistance mutation has yet been described for this drug. Therefore, we performed in vitro selection experiments with DTG using viruses of subtypes B, C, and A/G and showed that the most common mutation to emerge was R263K. Further analysis by site-directed mutagenesis showed that R263K does confer low-level resistance to DTG and decreased integration in cell culture without altering reverse transcription. Biochemical cell-free assays performed with purified IN enzyme containing R263K confirmed the absence of major resistance against DTG and showed a slight decrease in 3' processing and strand transfer activities compared to the wild type. Structural modeling suggested and in vitro IN-DNA binding assays show that the R263K mutation affects IN-DNA interactions.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/genetics , HIV-1/drug effects , HIV-1/enzymology , Heterocyclic Compounds, 3-Ring/pharmacology , Mutation, Missense , Cell Line , Drug Resistance, Viral , HIV Infections/virology , HIV Integrase/metabolism , HIV-1/genetics , Humans , Models, Molecular , Oxazines , Piperazines , Pyridones
8.
Antimicrob Agents Chemother ; 55(10): 4552-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807982

ABSTRACT

The integrase inhibitor (INI) dolutegravir (DTG; S/GSK1349572) has significant activity against HIV-1 isolates with raltegravir (RAL)- and elvitegravir (ELV)-associated resistance mutations. As an initial step in characterizing the different resistance profiles of DTG, RAL, and ELV, we determined the dissociation rates of these INIs with integrase (IN)-DNA complexes containing a broad panel of IN proteins, including IN substitutions corresponding to signature RAL and ELV resistance mutations. DTG dissociates slowly from a wild-type IN-DNA complex at 37°C with an off-rate of 2.7 × 10(-6) s(-1) and a dissociative half-life (t(1/2)) of 71 h, significantly longer than the half-lives for RAL (8.8 h) and ELV (2.7 h). Prolonged binding (t(1/2), at least 5 h) was observed for DTG with IN-DNA complexes containing E92, Y143, Q148, and N155 substitutions. The addition of a second substitution to either Q148 or N155 typically resulted in an increase in the off-rate compared to that with the single substitution. For all of the IN substitutions tested, the off-rate of DTG from IN-DNA complexes was significantly slower (from 5 to 40 times slower) than the off-rate of RAL or ELV. These data are consistent with the potential for DTG to have a higher genetic barrier to resistance, provide evidence that the INI off-rate may be an important component of the mechanism of INI resistance, and suggest that the slow dissociation of DTG may contribute to its distinctive resistance profile.


Subject(s)
DNA, Viral/metabolism , HIV Integrase Inhibitors/metabolism , HIV Integrase/metabolism , HIV-1/drug effects , Heterocyclic Compounds, 3-Ring/metabolism , Pyrrolidinones/metabolism , Quinolones/metabolism , Amino Acid Substitution , DNA, Complementary , Drug Resistance, Viral , Genotype , HIV Integrase/genetics , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , HIV-1/genetics , Heterocyclic Compounds, 3-Ring/pharmacology , Mutation , Oxazines , Piperazines , Pyridones , Pyrrolidinones/pharmacology , Quinolones/pharmacology , Raltegravir Potassium
9.
Antimicrob Agents Chemother ; 55(2): 813-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115794

ABSTRACT

S/GSK1349572 is a next-generation HIV integrase (IN) inhibitor designed to deliver potent antiviral activity with a low-milligram once-daily dose requiring no pharmacokinetic (PK) booster. In addition, S/GSK1349572 demonstrates activity against clinically relevant IN mutant viruses and has potential for a high genetic barrier to resistance. S/GSK1349572 is a two-metal-binding HIV integrase strand transfer inhibitor whose mechanism of action was established through in vitro integrase enzyme assays, resistance passage experiments, activity against viral strains resistant to other classes of anti-HIV agents, and mechanistic cellular assays. In a variety of cellular antiviral assays, S/GSK1349572 inhibited HIV replication with low-nanomolar or subnanomolar potency and with a selectivity index of 9,400. The protein-adjusted half-maximal effective concentration (PA-EC(50)) extrapolated to 100% human serum was 38 nM. When virus was passaged in the presence of S/GSK1349572, highly resistant mutants were not selected, but mutations that effected a low fold change (FC) in the EC(50) (up to 4.1 fold) were identified in the vicinity of the integrase active site. S/GSK1349572 demonstrated activity against site-directed molecular clones containing the raltegravir-resistant signature mutations Y143R, Q148K, N155H, and G140S/Q148H (FCs, 1.4, 1.1, 1.2, and 2.6, respectively), while these mutants led to a high FC in the EC(50) of raltegravir (11- to >130-fold). Either additive or synergistic effects were observed when S/GSK1349572 was tested in combination with representative approved antiretroviral agents; no antagonistic effects were seen. These findings demonstrate that S/GSK1349572 would be classified as a next-generation drug in the integrase inhibitor class, with a resistance profile markedly different from that of first-generation integrase inhibitors.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Integrase/drug effects , HIV-1/drug effects , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Cell Line , Cell Line, Transformed , Drug Resistance, Viral , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase Inhibitors/chemistry , HIV-1/enzymology , HIV-1/physiology , Humans , Microbial Sensitivity Tests/methods , Mutation , Naphthyridines/chemical synthesis , Naphthyridines/chemistry , Naphthyridines/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/virology , Virus Integration/drug effects , Virus Replication/drug effects
10.
Antiviral Res ; 81(2): 141-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19027039

ABSTRACT

Passage of HIV-1 in the presence of integrase inhibitors (INIs) generates resistant viruses that have mutations in the integrase region. Integrase-resistant mutations Q148K and Q148R were identified as primary mutations with the passage of HIV-1 IIIB in the presence of INIs S-1360 or S/GSK-364735, respectively. Secondary amino acid substitutions E138K or G140S were observed when passage with INI was continued. The role of these mutations was investigated with molecular clones. Relative to Q148K alone, Q148K/E138K had 2- and >6-fold increases in resistance to S-1360 and S/GSK-364735, respectively, and the double mutant had slightly better infectivity and replication kinetics. In contrast, Q148K/G140S and Q148R/E138K had nearly equivalent or slightly reduced fold resistance to the INI compared with their respective Q148 primary mutants, and had increases in infectivity and replication kinetics. Recovery of these surrogates of viral fitness coincided with the recovery of integration efficiency of viral DNA into the host cell chromosome for these double mutants. These data show that recovery of viral integration efficiency can be an important factor for the emergence and maintenance of INI-resistant mutations.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Integrase/metabolism , HIV-1/drug effects , HIV-1/physiology , Mutation, Missense , Virus Replication/drug effects , Furans/pharmacology , HIV Integrase/genetics , HIV-1/genetics , Humans , Jurkat Cells , Triazoles/pharmacology
11.
J Infect Dis ; 199(1): 84-8, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19032103

ABSTRACT

Thymidine-sparing triple-nucleoside regimens have exhibited poor virologic response despite apparent phenotypic susceptibility to 2 of 3 regimen components at early time points. Phenotypic resistance masking by wild-type virus may explain this discrepancy.Consistent with this notion were (1) the presence of low level nucleoside reverse-transcriptase inhibitor-resistant human immunodeficiency virus in subjects receiving failing first-line regimens consisting of tenofovir (TDF), abacavir (ABC), and lamivudine (3TC); (2) lower fold resistance associated with mixtures versus mutants in a clinical-isolate database; and (3) dose dependent changes in susceptibility to ABC, 3TC, TDF, and didanosine on titration of K65R and/or M184V with wild-type virus. These findings underscore the limitations of stand-alone phenotypic susceptibility measures and emphasize the importance of complementary and/or more sensitive techniques.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV-1/genetics , Adenine/analogs & derivatives , Adenine/therapeutic use , Didanosine/therapeutic use , Dideoxynucleosides/therapeutic use , Drug Therapy, Combination , Genetic Predisposition to Disease , Genotype , Humans , Lamivudine/therapeutic use , Organophosphonates/therapeutic use , Phenotype , Plasmids , Tenofovir , Viral Load
12.
Antimicrob Agents Chemother ; 52(3): 901-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160521

ABSTRACT

The naphthyridinone GSK364735 potently inhibited recombinant human immunodeficiency virus type 1 (HIV-1) integrase in a strand transfer assay (mean 50% inhibitory concentration +/- standard deviation, 8 +/- 2 nM). As expected based on the structure of the drug, it bound competitively with another two-metal binding inhibitor (Kd [binding constant], 6 +/- 4 nM). In a number of different cellular assays, GSK364735 inhibited HIV replication with potency at nanomolar concentrations (e.g., in peripheral blood mononuclear cells and MT-4 cells, 50% effective concentrations were 1.2 +/- 0.4 and 5 +/- 1 nM, respectively), with selectivity indexes of antiviral activity versus in-assay cytotoxicity of at least 2,200. When human serum was added, the antiviral potency decreased (e.g., a 35-fold decrease in the presence of 100% human serum was calculated by extrapolation from the results of the MT-4 cell assay). In cellular assays, GSK364735 blocked viral DNA integration, with a concomitant increase in two-long-terminal-repeat circles. As expected, this integrase inhibitor was equally active against wild-type viruses and mutant viruses resistant to approved drugs targeting either reverse transcriptase or protease. In contrast, some but not all viruses resistant to other integrase inhibitors were resistant to GSK364735. When virus was passaged in the presence of the inhibitor, we identified resistance mutations within the integrase active site that were the same as or similar to mutations arising in response to other two-metal binding inhibitors. Finally, either additive or synergistic effects were observed when GSK364735 was tested in combination with approved antiretrovirals (i.e., no antagonistic effects were seen). Thus, based on all the data, GSK364735 exerted potent antiviral activity through the inhibition of viral DNA integration by interacting at the two-metal binding site within the catalytic center of HIV integrase.


Subject(s)
HIV Integrase Inhibitors/pharmacology , HIV Integrase/drug effects , HIV-1/drug effects , Naphthyridines/pharmacology , Anti-HIV Agents/pharmacology , Cell Line , Cells, Cultured , Drug Resistance, Viral , Drug Synergism , HIV Integrase/genetics , HIV Protease Inhibitors/pharmacology , HIV-1/enzymology , HIV-1/physiology , Humans , Leukocytes, Mononuclear/virology , Microbial Sensitivity Tests/methods , Mutation , Reverse Transcriptase Inhibitors/pharmacology , Virus Integration/drug effects , Virus Replication/drug effects
13.
J Acquir Immune Defic Syndr ; 43(5): 567-70, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17075395

ABSTRACT

The impact of drug resistance-associated mutations on subsequent antiretroviral therapy is an important consideration in managing treatment-experienced, HIV-1-infected patients. Lamivudine (3TC) and emtricitabine (FTC) are structurally related nucleoside reverse transcriptase inhibitors (NRTIs) approved for use in HIV-1-infected individuals. To evaluate whether susceptibility differences exist between lamivudine and emtricitabine, the phenotypic impact of common NRTI resistance-associated mutations was compared in HIV-1 from patient samples with paired FTC and 3TC susceptibility results. FTC phenotypic susceptibility was more greatly impacted than 3TC susceptibility in the presence of thymidine analogue mutations (TAMs), as the mean fold-change values were higher for FTC than for 3TC in groups of samples containing TAMs (P < 0.001 for 6 of 7 groups). For samples with K65R, L74I/V, or Q151M mutations, the phenotypic impact was similar, as the mean fold-change was not significantly different between drugs. Although the long-term clinical significance of these differences is unclear, they may suggest differential efficacy in some patients with prior NRTI experience, especially those with HIV harboring TAMs.


Subject(s)
Anti-HIV Agents/pharmacology , Deoxycytidine/analogs & derivatives , Drug Resistance, Viral/genetics , HIV-1/drug effects , HIV-1/genetics , Lamivudine/pharmacology , Deoxycytidine/pharmacology , Emtricitabine , Mutation , Thymidine
14.
Antimicrob Agents Chemother ; 48(5): 1647-51, 2004 May.
Article in English | MEDLINE | ID: mdl-15105116

ABSTRACT

New human cytomegalovirus (HCMV) therapies with novel mechanisms of action are needed to treat drug-resistant HCMV that arises during therapy with currently approved agents. 2-Bromo-5,6-dichloro-1-beta-D-ribofuranosyl-1H-benzimidazole (BDCRB) is an effective anti-HCMV agent with a novel mechanism of action, but problems with in vivo stability preclude clinical development. A D-ribopyranosyl derivative of BDCRB, GW275175X, displays similar antiviral activity without the in vivo stability problems. We present an initial description of the activity of GW275175X against HCMV, other herpesviruses, and selected nonherpesviruses. In addition, we show that it acts as a DNA maturation inhibitor like the parent compound, BDCRB, rather than via the mechanisms of action of 1263W94 or any anti-HCMV drugs approved for marketing. GW275175X is a promising candidate for clinical development as an anti-HCMV agent.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Cytomegalovirus/drug effects , Ribonucleosides/pharmacology , Cell Line , DNA/genetics , Electrophoresis , Herpesviridae/drug effects , Humans , In Situ Hybridization , Indicators and Reagents , Viral Plaque Assay
15.
J Virol ; 78(5): 2637-41, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14963172

ABSTRACT

Peptide antagonists of the human papillomavirus type 11 (HPV-11) E2-DNA association were identified using a filamentous bacteriophage random peptide library. Synthetic peptides antagonized the E2-DNA interaction, effectively blocked E2-mediated transcriptional activation of a reporter gene in cell culture, and inhibited E1-E2-mediated HPV-11 DNA replication in vitro. These peptides may prove to be useful tools for characterizing E2 function and for exploring the effectiveness of E2-inhibitor-based treatments for HPV-associated diseases.


Subject(s)
DNA Replication/drug effects , DNA, Viral/metabolism , Gene Expression Regulation, Viral/drug effects , Peptides/pharmacology , Transcriptional Activation/drug effects , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell-Free System , Chlorocebus aethiops , DNA, Viral/antagonists & inhibitors , Electron Spin Resonance Spectroscopy , Genes, Reporter/genetics , Inhibitory Concentration 50 , Molecular Sequence Data , Papillomaviridae/drug effects , Papillomaviridae/genetics , Papillomaviridae/physiology , Peptide Library , Peptides/chemical synthesis , Peptides/chemistry , Protein Binding/drug effects , Thermodynamics , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Trans-Activators/metabolism , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism
16.
Antimicrob Agents Chemother ; 47(7): 2186-92, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12821466

ABSTRACT

Herpes simplex virus types 1 and 2 (HSV-1 and HSV-2), varicella-zoster virus (VZV), cytomegalovirus (CMV), Epstein-Barr virus (EBV), human herpesvirus 6 (HHV-6), and human herpesvirus 8 (HHV-8) are responsible for a number of clinical manifestations in both normal and immunocompromised individuals. The parent benzimidazole ribonucleosides evaluated in this series, 2-bromo-5,6-dichloro-1-(beta-D-ribofuranosyl)benzimidazole (BDCRB) and maribavir (1263W94), are potent and selective inhibitors of human CMV replication. These nucleosides act by two different mechanisms. BDCRB blocks the processing and maturation of viral DNA, whereas 1263W94 inhibits the viral enzyme pUL97 and interferes with DNA synthesis. In the present study, we have evaluated the in vitro antiviral activity of BDCRB, an analog, GW275175X (175X), and 1263W94 against the replication of HSV-1, HSV-2, VZV, CMV, EBV, HHV-6, and HHV-8. By using various methodologies, significant activity was observed against human CMV and EBV but not against HSV-1, HSV-2, VZV, HHV-6, or HHV-8. Plaque reduction assays performed on a variety of laboratory and clinical isolates of human CMV indicated that all strains, including those resistant to ganciclovir (GCV) and foscarnet, were sensitive to all three benzimidazole ribonucleosides, with mean 50% effective concentration values of about 1 to 5 microM compared to that of GCV at 6 microM. The toxicity of these compounds in tissue culture cells appeared to be similar to that observed with GCV. These results demonstrate that the benzimidazole ribonucleosides are active against human CMV and EBV and suggest that they may be useful for the treatment of infections caused by these herpesviruses.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 2, Human/drug effects , Ribonucleosides/pharmacology , Antiviral Agents/chemistry , Benzimidazoles/chemistry , Cytomegalovirus/drug effects , Cytomegalovirus/growth & development , Herpesvirus 1, Human/growth & development , Herpesvirus 2, Human/growth & development , Herpesvirus 3, Human/drug effects , Herpesvirus 3, Human/growth & development , Herpesvirus 6, Human/drug effects , Herpesvirus 6, Human/growth & development , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/growth & development , In Vitro Techniques , Ribonucleosides/chemistry , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...