Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 18: 815-830, 2019 Dec 06.
Article in English | MEDLINE | ID: mdl-31734561

ABSTRACT

U1 interference (U1i) RNAs can be designed to correct splicing defects and target pathogenic RNA, such as HIV-1 RNA. In this study, we show that U1i RNAs that enhance HIV-1 RNA splicing are more effective at inhibiting HIV-1 production compared to top U1i RNAs that inhibit polyadenylation of HIV-1 RNA. A U1i RNA was also identified targeting a site upstream of the first splice acceptor site in the Gag coding region that was effective at inhibiting HIV-1 production. U1-T6, which enhanced HIV-1 RNA splicing, was superior to an antiviral short hairpin RNA (shRNA) currently in clinical trials. To increase specificity, the recognition domain of U1-T6 was elongated by 3-6 nt. The elongated molecules inhibited HIV-1 production from different HIV-1 strains, including one with a mismatch in the target site. These results suggest that lengthening the recognition domain can enhance the specificity of U1i RNAs for their intended target sites while at the same time allowing them to tolerate single mismatch mutations. Overall, our results demonstrate that U1-T6 with an elongated recognition domain inhibits HIV-1 production and has both the efficacy and specificity to be a promising candidate for HIV-1 gene therapy.

2.
Sci Rep ; 7(1): 6345, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740124

ABSTRACT

We evaluated Sofosbuvir (SOF), the anti-hepatitis C virus prodrug of ß-d-2'-deoxy-2'-α-fluoro-2'-ß-C-methyluridine-5'-monophosphate, for potential inhibitory activity against DENV replication. Both cell-based and biochemical assays, based on use of purified DENV full-length NS5 enzyme, were studied. Cytopathic effect protection and virus yield reduction assays confirmed that SOF possessed anti-DENV activity in cell culture with a 50% effective concentration (EC50) of 4.9 µM and 1.4 µM respectively. Real-time RT-PCR verified that SOF inhibits generation of viral RNA with an EC50 of 9.9 µM. Purified DENV NS5 incorporated the active triphosphate form (SOF-TP) into nascent RNA, causing chain-termination. Relative to the natural UTP, the incorporation efficiency of SOF-TP was low (discrimination value = 327.5). In a primer extension assay, SOF-TP was active against DENV NS5 wild-type polymerase activity with an IC50 of 14.7 ± 2.5 µM. The S600T substitution in the B Motif of DENV polymerase conferred 4.3-fold resistance to SOF-TP; this was due to decreased incorporation efficiency rather than enhanced excision of the incorporated SOF nucleotide. SOF has antiviral activity against DENV replication. The high discrimination value in favor of UTP in enzyme assays may not necessarily preclude antiviral activity in cells. SOF may be worthy of evaluation against severe DENV infections in humans.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/physiology , Sofosbuvir/pharmacology , Virus Replication/drug effects , Cell Line , Dengue/drug therapy , Dengue/virology , Dengue Virus/drug effects , Dengue Virus/enzymology , Drug Evaluation , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Uridine Triphosphate/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
3.
J Antimicrob Chemother ; 72(3): 727-734, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28069884

ABSTRACT

Background: The viral RNA-dependent RNA polymerase (RdRp) enzymes of the Flaviviridae family are essential for viral replication and are logically important targets for development of antiviral therapeutic agents. Zika virus (ZIKV) is a rapidly re-emerging human pathogen for which no vaccine or antiviral agent is currently available. Methods: To facilitate development of ZIKV RdRp inhibitors, we have established an RdRp assay using purified recombinant ZIKV NS5 polymerase. Results: We have shown that both the hepatitis C virus (HCV) nucleoside inhibitor sofosbuvir triphosphate and a pyridoxine-derived non-nucleoside small-molecule inhibitor, DMB213, can act against ZIKV RdRp activity at IC 50 s of 7.3 and 5.2 µM, respectively, in RNA synthesis reactions catalysed by recombinant ZIKV NS5 polymerase. Cell-based assays confirmed the anti-ZIKV activity of sofosbuvir and DMB213 with 50% effective concentrations (EC 50 s) of 8.3 and 4.6 µM, respectively. Control studies showed that DMB213 did not inhibit recombinant HIV-1 reverse transcriptase and showed only very weak inhibition of HIV-1 integrase strand-transfer activity. The S604T substitution in motif B of the ZIKV RdRp, which corresponds to the S282T substitution in motif B of HCV RdRp, which confers resistance to nucleotide inhibitors, also conferred resistance to sofosbuvir triphosphate, but not to DMB213. Enzyme assays showed that DMB213 appears to be competitive with natural nucleoside triphosphate (NTP) substrates. Conclusions: Recombinant ZIKV RdRp assays can be useful tools for the screening of both nucleos(t)ide compounds and non-nucleotide metal ion-chelating agents that interfere with ZIKV replication.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Zika Virus/enzymology , Drug Discovery/methods , HIV Integrase/metabolism , HIV Reverse Transcriptase/antagonists & inhibitors , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Recombinant Proteins/metabolism , Sofosbuvir/pharmacology , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus/physiology
4.
Antimicrob Agents Chemother ; 60(1): 600-8, 2016 01.
Article in English | MEDLINE | ID: mdl-26574011

ABSTRACT

The viral RNA-dependent RNA polymerase (RdRp) activity of the dengue virus (DENV) NS5 protein is an attractive target for drug design. Here, we report the identification of a novel class of inhibitor (i.e., an active-site metal ion chelator) that acts against DENV RdRp activity. DENV RdRp utilizes a two-metal-ion mechanism of catalysis; therefore, we constructed a small library of compounds, through mechanism-based drug design, aimed at chelating divalent metal ions in the catalytic site of DENV RdRp. We now describe a pyridoxine-derived small-molecule inhibitor that targets DENV RdRp and show that 5-benzenesulfonylmethyl-3-hydroxy-4-hydroxymethyl-pyridine-2-carboxylic acid hydroxyamide (termed DMB220) inhibited the RdRp activity of DENV serotypes 1 to 4 at low micromolar 50% inhibitory concentrations (IC50s of 5 to 6.7 µM) in an enzymatic assay. The antiviral activity of DMB220 against DENV infection was also verified in a cell-based assay and showed a 50% effective concentration (EC50) of <3 µM. Enzyme assays proved that DMB220 was competitive with nucleotide incorporation. DMB220 did not inhibit the enzymatic activity of recombinant HIV-1 reverse transcriptase and showed only weak inhibition of HIV-1 integrase strand transfer activity, indicating high specificity for DENV RdRp. S600T substitution in the DENV RdRp, which was previously shown to confer resistance to nucleoside analogue inhibitors (NI), conferred 3-fold hypersusceptibility to DMB220, and enzymatic analyses showed that this hypersusceptibility may arise from the decreased binding/incorporation efficiency of the natural NTP substrate without significantly impacting inhibitor binding. Thus, metal ion chelation at the active site of DENV RdRp represents a viable anti-DENV strategy, and DMB220 is the first of a new class of DENV inhibitor.


Subject(s)
Antiviral Agents/pharmacology , Chelating Agents/pharmacology , Dengue Virus/drug effects , Hydroxamic Acids/pharmacology , Picolines/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Sulfones/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Aedes , Amino Acid Substitution , Animals , Antiviral Agents/chemical synthesis , Binding Sites , Catalytic Domain , Cell Line , Chelating Agents/chemical synthesis , Cricetinae , Dengue Virus/enzymology , Dengue Virus/genetics , Dose-Response Relationship, Drug , Drug Design , Epithelial Cells/drug effects , Epithelial Cells/virology , Gene Expression , Histidine/genetics , Histidine/metabolism , Humans , Hydroxamic Acids/chemical synthesis , Kinetics , Molecular Docking Simulation , Oligopeptides/genetics , Oligopeptides/metabolism , Picolines/chemical synthesis , Protein Binding , Protein Structure, Secondary , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Sulfones/chemical synthesis , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
5.
Antivir Chem Chemother ; 24(1): 28-38, 2015 Feb.
Article in English | MEDLINE | ID: mdl-26149264

ABSTRACT

BACKGROUND: Due to resistance to all classes of anti-HIV drugs and drug toxicity, there is a need for the discovery and development of new anti-HIV drugs. METHODS: HIV-1 inhibitors were identified and biologically characterized for mechanism of action. RESULTS: We identified a dibenzocyclooctadiene lignan, termed HDS2 that possessed anti-HIV activity against a wide variety of viral strains with EC50 values in the 1-3 µM range. HDS2 was shown to act as an NNRTI by qPCR and in vitro enzyme assays. CONCLUSIONS: This compound provides a new scaffold for further optimization of activity through structure-guided design.


Subject(s)
Cyclooctanes/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , HIV-1/enzymology , Lignans/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Cell Line , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , HIV-2/drug effects , HIV-2/enzymology , Humans , Species Specificity , Structure-Activity Relationship
6.
Antimicrob Agents Chemother ; 59(6): 3189-96, 2015.
Article in English | MEDLINE | ID: mdl-25779585

ABSTRACT

Compound A is a novel nucleotide-competing HIV-1 reverse transcriptase (RT) inhibitor (NcRTI) that selects for a unique W153L substitution that confers hypersusceptibility to tenofovir, while the K65R substitution in RT confers resistance against tenofovir and enhances susceptibility to NcRTIs. Although the K65R substitution is more common in subtype C viruses, the impact of subtype variability on NcRTI susceptibility has not been studied. In the present study, we performed experiments with compound A by using purified recombinant RT enzymes and viruses of subtypes B and C and circulating recombinant form CRF_A/G. We confirmed the hypersusceptibility of K65R substitution-containing RTs to compound A for subtype C, CRF_A/G, and subtype B. Steady-state kinetic analysis showed that K65R RTs enhanced the susceptibility to compound A by increasing binding of the inhibitor to the nucleotide binding site of RT in a subtype-independent manner, without significantly discriminating against the natural nucleotide substrate. These data highlight the potential utility of NcRTIs, such as compound A, for treatment of infections with K65R substitution-containing viruses, regardless of HIV-1 subtype.


Subject(s)
HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Amino Acid Substitution , Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/genetics , Humans , Kinetics
7.
J Antimicrob Chemother ; 70(3): 750-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25433008

ABSTRACT

OBJECTIVES: Cenicriviroc is a potent antagonist of the chemokine coreceptors 5 and 2 (CCR5/CCR2) and blocks HIV-1 entry. The CCR5 inhibitor maraviroc has been shown in tissue culture to be able to repel cell-free virions from the cell surface into extracellular space. We hypothesized that cenicriviroc might exhibit a similar effect, and tested this using clinical samples from the Phase IIb study 652-2-202, by measuring rates of intracellular DNA decline. We also monitored viral RNA levels in culture fluids. METHODS: We infected PM-1 cells with CCR5-tropic HIV-1 BaL in the presence or absence of inhibitory concentrations of cenicriviroc (20 nM) or maraviroc (50 nM) or controls. Viral load levels and p24 were measured by ELISA, quantitative PCR and quantitative real-time reverse transcription PCR at 4 h post-infection. Frozen PBMC DNA samples from 30 patients with virological success in the Phase IIb study were studied, as were early and late reverse transcript levels. Docking studies compared binding between cenicriviroc/CCR5 and maraviroc/CCR5. RESULTS: Unlike maraviroc, cenicriviroc did not cause an increase in the amount of virus present in culture fluids at 4 h compared with baseline. The use of cenicriviroc did, however, result in lower levels of intracellular viral DNA after 4 h. Structural modelling indicates that cenicriviroc binds more deeply than maraviroc to the hydrophobic pocket of CCR5, providing an explanation for the absence of viral rebound with cenicriviroc. CONCLUSIONS: In contrast to maraviroc, cenicriviroc does not repel virus back into extracellular space. Differences in results may be due to superior binding of cenicriviroc to CCR5 compared with maraviroc.


Subject(s)
Anti-HIV Agents/pharmacology , DNA, Viral/analysis , HIV Infections/virology , HIV-1/isolation & purification , Imidazoles/pharmacology , Viral Load , Anti-HIV Agents/therapeutic use , Cell Line , Clinical Trials, Phase II as Topic , Culture Media , Enzyme-Linked Immunosorbent Assay , Extracellular Space/virology , HIV Infections/drug therapy , Humans , Imidazoles/therapeutic use , Leukocytes, Mononuclear/virology , Real-Time Polymerase Chain Reaction , Sulfoxides , Virus Cultivation
8.
J Int AIDS Soc ; 17(4 Suppl 3): 19531, 2014.
Article in English | MEDLINE | ID: mdl-25394040

ABSTRACT

INTRODUCTION: Cenicriviroc (CVC), a once-daily, dual CCR5/CCR2 co-receptor antagonist, has completed Phase 2b development. CVC demonstrated favourable safety and similar efficacy compared with efavirenz (EFV) in Study 202 (NCT01338883); an ex vivo sub-analysis evaluated treatment effects on HIV entry, measured by intracellular HIV DNA declines, in subjects with virologic success at Week 24. In addition, in vitro assays determined and compared the extent of any cell-free virion redistribution that CVC or maraviroc (MVC) may cause. METHODS: Ex vivo: intracellular DNA (frozen PBMCs) from 30 subjects with virologic success at Week 24 (10, 13 and 7 subjects on CVC 100 mg, CVC 200 mg and EFV, respectively). Early (strong-stop) and late (full-length) reverse transcript levels were measured by qPCR. In vitro: PM-1 cells were infected with CCR5-tropic HIV-1 BaL in the presence or absence of inhibitory concentrations of CVC (20 nM), MVC (50 nM) or controls. P24 and viral load levels were measured by ELISA and qRT-PCR after 4 hours. RESULTS: Ex vivo analysis showed full-length HIV DNA declines were similar across all groups (CVC 100 mg, CVC 200 mg and EFV) at Week 24. Strong-stop HIV DNA declines (a marker of HIV entry) at Week 24 were pronounced for both CVC arms (CVC 100 mg, 51% decline; CVC 200 mg, 37% decline) compared to no decline for the EFV arm. In vitro experiments revealed that CVC-treated cells had lower levels of supernatant P24 at 4 hours versus baseline (0 hrs: 506 ng/mL; 4 hrs: 192 ng/mL), but P24 levels remained constant for MVC-treated cells after 4 hours (0 hrs: 506 ng/mL; 4 hrs: 520 ng/mL). Viral load levels for CVC-treated cells remained stable after 4 hours (0 hrs: 1.19×10(10) copies/mL; 4 hrs: 1.26×10(10) copies/mL). MVC-treated cells exhibited a slight increase in viral load after 4 hours (0 hrs: 1.19×10(10) copies/mL; 4 hrs: 1.67×10(10) copies/mL). CONCLUSIONS: Ex vivo analysis confirmed that CVC treatment blocks HIV entry (strong-stop HIV DNA declines), while in vitro analysis showed that CVC-treated cells do not repel virus back into the extracellular space, as seen with MVC. Experiments are underway to determine whether or not interactions between CVC and HIV at the binding site may explain these unanticipated findings.

9.
Antimicrob Agents Chemother ; 58(8): 4515-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24867966

ABSTRACT

A W153L substitution in HIV-1 reverse transcriptase (RT) was recently identified by selection with a novel nucleotide-competing RT inhibitor (NcRTI) termed compound A that is a member of the benzo[4,5]furo[3,2,d]pyrimidin-2-one NcRTI family of drugs. To investigate the impact of W153L, alone or in combination with the clinically relevant RT resistance substitutions K65R (change of Lys to Arg at position 65), M184I, K101E, K103N, E138K, and Y181C, on HIV-1 phenotypic susceptibility, viral replication, and RT enzymatic function, we generated recombinant RT enzymes and viruses containing each of these substitutions or various combinations of them. We found that W153L-containing viruses were impaired in viral replicative capacity and were hypersusceptible to tenofovir (TFV) while retaining susceptibility to most nonnucleoside RT inhibitors. The nucleoside 3TC retained potency against W153L-containing viruses but not when the M184I substitution was also present. W153L was also able to reverse the effects of the K65R substitution on resistance to TFV, and K65R conferred hypersusceptibility to compound A. Biochemical assays demonstrated that W153L alone or in combination with K65R, M184I, K101E, K103N, E138K, and Y181C impaired enzyme processivity and polymerization efficiency but did not diminish RNase H activity, providing mechanistic insights into the low replicative fitness associated with these substitutions. We show that the mechanism of the TFV hypersusceptibility conferred by W153L is mainly due to increased efficiency of TFV-diphosphate incorporation. These results demonstrate that compound A and/or derivatives thereof have the potential to be important antiretroviral agents that may be combined with tenofovir to achieve synergistic results.


Subject(s)
Amino Acid Substitution , Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Alkynes , Benzoxazines/pharmacology , Cyclopropanes , HEK293 Cells , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Lamivudine/pharmacology , Microbial Sensitivity Tests , Mutagenesis, Site-Directed , Mutation , Nevirapine/pharmacology , Organophosphonates/pharmacology , Pyrimidines/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tenofovir , Virus Replication
10.
J Acquir Immune Defic Syndr ; 67(1): 7-14, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24872133

ABSTRACT

BACKGROUND: The development of envelope-specific neutralizing antibodies that can interfere with viral entry into target cells is important for the development of an HIV-1 vaccine. Another means of blocking viral entry is through the use of entry inhibitors such as the CCR5 inhibitor maraviroc (MVC), which can also repel cell-free virus particles from the cell surface. For this reason, we hypothesized that exposure to entry inhibitors might alter viral infectiousness and sensitivity to antibody-mediated neutralization. METHODS: The CCR5-tropic HIV-1 variants BaL, AD8, and CC 1/85 were used to infect PM-1 cells in the presence of 2 entry inhibitors, enfuvirtide and MVC. After 4 hours, culture fluids were ultrafiltered and the infectiousness and susceptibility to broadly neutralizing antibodies (2F5, 4E10, 2G12, b12, VRC01, PG9) of viruses exposed to these entry inhibitors were assessed using TZM-bl cells. RESULTS: Viruses exposed to the entry inhibitor MVC exhibited lower infectiousness than controls. Enfuvirtide exposure increased AD8 sensitivity to 2F5, 4E10, VRC01, and b12 and increased BaL sensitivity to 4E10 while lowering BaL sensitivity to b12 and VRC01. MVC-exposed BaL became less susceptible to the gp120-specific antibodies b12, 2G12, and VRC01. CONCLUSIONS: Exposure to entry inhibitors altered HIV-1 infectiousness and sensitivity to gp120-specific neutralizing antibodies. This alteration of entry inhibitor-exposed virus has implications for the development of future entry inhibitors and for vaccine development.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cyclohexanes/pharmacology , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Fusion Inhibitors/pharmacology , HIV Infections/drug therapy , HIV-1/physiology , Triazoles/pharmacology , Cell Line , Drug Synergism , HIV Infections/virology , Humans , Inhibitory Concentration 50 , Maraviroc , Neutralization Tests , Regression Analysis , Virus Replication/drug effects
11.
J Virol ; 88(3): 1536-47, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24227862

ABSTRACT

Clinical resistance to rilpivirine (RPV), a novel nonnucleoside reverse transcriptase (RT) inhibitor (NNRTI), is associated an E-to-K mutation at position 138 (E138K) in RT together with an M184I/V mutation that confers resistance against emtricitabine (FTC), a nucleoside RT inhibitor (NRTI) that is given together with RPV in therapy. These two mutations can compensate for each other in regard to fitness deficits conferred by each mutation alone, raising the question of why E138K did not arise spontaneously in the clinic following lamivudine (3TC) use, which also selects for the M184I/V mutations. In this context, we have investigated the role of a N348I connection domain mutation that is prevalent in treatment-experienced patients. N348I confers resistance to both the NRTI zidovudine (ZDV) and the NNRTI nevirapine (NVP) and was also found to be associated with M184V and to compensate for deficits associated with the latter mutation. Now, we show that both N348I alone and N348I/M184V can prevent or delay the emergence of E138K under pressure with RPV or a related NNRTI, termed etravirine (ETR). N348I also enhanced levels of resistance conferred by E138K against RPV and ETR by 2.2- and 2.3-fold, respectively. The presence of the N348I or M184V/N348I mutation decreased the replication capacity of E138K virus, and biochemical assays confirmed that N348I, in a background of E138K, impaired RT catalytic efficiency and RNase H activity. These findings help to explain the low viral replication capacity of viruses containing the E138K/N348I mutations and how N348I delayed or prevented the emergence of E138K in patients with M184V-containing viruses.


Subject(s)
Drug Resistance, Viral , HIV Infections/virology , HIV Reverse Transcriptase/chemistry , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , Mutation, Missense , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication , Amino Acid Motifs , HIV Reverse Transcriptase/metabolism , HIV-1/drug effects , HIV-1/genetics , HIV-1/physiology , Humans , Nitriles/pharmacology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Rilpivirine , Virus Replication/drug effects
12.
Antimicrob Agents Chemother ; 57(11): 5649-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002090

ABSTRACT

Resistance to the recently approved nonnucleoside reverse transcriptase inhibitor (NNRTI) rilpivirine (RPV) commonly involves substitutions at positions E138K and K101E in HIV-1 reverse transcriptase (RT), together with an M184I substitution that is associated with resistance to coutilized emtricitabine (FTC). Previous biochemical and virological studies have shown that compensatory interactions between substitutions E138K and M184I can restore enzyme processivity and the viral replication capacity. Structural modeling studies have also shown that disruption of the salt bridge between K101 and E138 can affect RPV binding. The current study was designed to investigate the impact of K101E, alone or in combination with E138K and/or M184I, on drug susceptibility, viral replication capacity, and enzyme function. We show here that K101E can be selected in cell culture by the NNRTIs etravirine (ETR), efavirenz (EFV), and dapivirine (DPV) as well as by RPV. Recombinant RT enzymes and viruses containing K101E, but not E138K, were highly resistant to nevirapine (NVP) and delavirdine (DLV) as well as ETR and RPV, but not EFV. The addition of K101E to E138K slightly enhanced ETR and RPV resistance compared to that obtained with E138K alone but restored susceptibility to NVP and DLV. The K101E substitution can compensate for deficits in viral replication capacity and enzyme processivity associated with M184I, while M184I can compensate for the diminished efficiency of DNA polymerization associated with K101E. The coexistence of K101E and E138K does not impair either viral replication or enzyme fitness. We conclude that K101E can play a significant role in resistance to RPV.


Subject(s)
Amino Acid Substitution , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Nitriles/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Alkynes , Benzoxazines/chemistry , Benzoxazines/pharmacology , Cyclopropanes , Delavirdine/chemistry , Delavirdine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Drug Resistance, Viral/drug effects , Emtricitabine , HEK293 Cells , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/virology , Microbial Sensitivity Tests , Mutagenesis, Site-Directed , Nevirapine/chemistry , Nevirapine/pharmacology , Nitriles/chemistry , Pyridazines/chemistry , Pyridazines/pharmacology , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/chemistry , Rilpivirine , Virus Replication/drug effects
13.
Antimicrob Agents Chemother ; 57(7): 3100-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23612196

ABSTRACT

Impacts of mutations at position E138 (A/G/K/Q/R/V) alone or in combination with M184I in HIV-1 reverse transcriptase (RT) were investigated. We also determined why E138K is the most prevalent nonnucleoside reverse transcriptase inhibitor mutation in patients failing rilpivirine (RPV) therapy. Recombinant RT enzymes and viruses containing each of the above-mentioned mutations were generated, and drug susceptibility was assayed. Each of the E138A/G/K/Q/R mutations, alone or in combination with M184I, resulted in decreased susceptibility to RPV and etravirine (ETR). The maximum decrease in susceptibility to RPV was observed for E138/R/Q/G by both recombinant RT assay and cell-based assays. E138Q/R-containing enzymes and viruses also showed the most marked decrease in susceptibility to ETR by both assays. The addition of M184I to the E138 mutations did not significantly change the levels of diminution in drug susceptibility. These findings indicate that E138R caused the highest level of loss of susceptibility to both RPV and ETR, and, accordingly, E138R should be recognized as an ETR resistance-associated mutation. The E138K/Q/R mutations can compensate for M184I in regard to both enzymatic fitness and viral replication capacity. The favored emergence of E138K over other mutations at position E138, together with M184I, is not due to an advantage in either the level of drug resistance or viral replication capacity but may reflect the fact that E138R and E138Q require two distinct mutations to occur, one of which is a disfavorable G-to-C mutation, whereas E138K requires only a single favorable G-to-A hypermutation. Of course, other factors may also affect the concept of barrier to resistance.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Nitriles/pharmacology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Cells, Cultured , Drug Resistance, Viral/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Mutation , Rilpivirine , Virus Replication/drug effects
14.
Antimicrob Agents Chemother ; 56(8): 4154-60, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615275

ABSTRACT

HIV entry inhibitors, such as maraviroc (MVC), prevent cell-free viruses from entering the cells. In clinical trials, patients who were treated with MVC often displayed viral loads that were above the limit of conventional viral load detection compared to efavirenz-based regimens. We hypothesize that viruses blocked by entry inhibitors may be redistributed to plasma, where they artificially increase viral load measurements compared to those with the use of antiretroviral drugs (ARVs) that act intracellularly. We infected PM-1 cells with CCR5-tropic HIV-1 BaL or CXCR4-tropic HIV-1 NL4-3 in the presence of inhibitory concentrations of efavirenz, raltegravir, enfuvirtide, maraviroc, and AMD3100, the latter three being entry inhibitors. Supernatant viral load, reverse transcriptase enzyme activity, and intracellular nucleic acid levels were measured at times up to 24 h postinfection. Infectivity of redistributed dual-tropic HIV-1 was assessed using TZM-bl cells. Extracellular viral load analysis revealed that entry inhibitor-treated cells had higher levels of virus in the supernatant than the cells treated with other ARVs at 8 h postinfection. By 24 h, the supernatant viral load was still higher for entry inhibitors than other ARVs. We observed a correlation between viral load and the step of entry inhibition. Dual-tropic virus infectivity was undiminished utilizing the CCR5 coreceptor following redistribution by CXCR4 entry inhibition. This in vitro model indicates that entry inhibitors exhibit a redistribution effect unseen with intracellular ARV drugs. Based on these results, the effectiveness of some entry inhibitors may be underestimated if plasma viral load is used as a sole indicator of clinical success.


Subject(s)
Cyclohexanes/pharmacology , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Triazoles/pharmacology , Viral Load/drug effects , Virus Internalization/drug effects , Alkynes , Anti-HIV Agents/pharmacology , Benzoxazines/pharmacology , Benzylamines , Cell Line , Cyclams , Cyclopropanes , DNA, Viral/analysis , Drug Resistance, Viral , Enfuvirtide , HIV Envelope Protein gp41/pharmacology , HIV Reverse Transcriptase/analysis , Heterocyclic Compounds/pharmacology , Humans , Maraviroc , Peptide Fragments/pharmacology , Pyrrolidinones/pharmacology , RNA, Viral/analysis , Raltegravir Potassium , Receptors, CCR5/metabolism , Receptors, CXCR4/metabolism
15.
Antimicrob Agents Chemother ; 56(8): 4257-67, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22615295

ABSTRACT

BMS-599793 is a small molecule entry inhibitor that binds to human immunodeficiency virus type 1 (HIV-1) gp120, resulting in the inhibition of CD4-dependent entry into cells. Since BMS-599793 is currently considered a candidate microbicide drug, we evaluated its efficacy against a number of primary patient HIV isolates from different subtypes and circulating recombinant forms (CRFs) and showed that activity varied between ∼3 ρM and 7 µM at 50% effective concentrations (EC(50)s). Interestingly, CRF01_AE HIV-1 isolates consistently demonstrated natural resistance against this compound. Genotypic analysis of >1,600 sequences (Los Alamos HIV sequence database) indicated that a single amino acid polymorphism in Env, H375, may account for the observed BMS-599793 resistance in CRF01_AE HIV-1. Results of site-directed mutagenesis experiments confirmed this hypothesis, and in silico drug docking simulations identified a drug resistance mechanism at the molecular level. In addition, CRF01_AE viruses were shown to be resistant to multiple broadly neutralizing monoclonal antibodies. Thus, our results not only provide insight into how Env polymorphisms may contribute to entry inhibitor resistance but also may help to elucidate how HIV can evade some broadly neutralizing antibodies. Furthermore, the high frequency of H375 in CRF01_AE HIV-1, and its apparent nonoccurrence in other subtypes, could serve as a means for rapid identification of CRF01_AE infections.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV Envelope Protein gp120/genetics , HIV-1/drug effects , Piperidines/pharmacology , Pyrazines/pharmacology , Virus Internalization/drug effects , Amino Acid Sequence , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Antibodies, Neutralizing/immunology , Cell Line, Tumor , Genes, env , Genotype , HIV Antibodies/immunology , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/genetics , HIV-1/immunology , HIV-1/isolation & purification , HIV-2/drug effects , Humans , Models, Molecular , Mutagenesis, Site-Directed , Piperidines/chemistry , Piperidines/metabolism , Polymorphism, Genetic , Protein Structure, Quaternary , Pyrazines/chemistry , Pyrazines/metabolism , Sequence Alignment
16.
Antimicrob Agents Chemother ; 56(2): 751-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22123692

ABSTRACT

Antiretroviral-based microbicides may offer a means to reduce the sexual transmission of HIV-1. Suboptimal use of a microbicide may, however, lead to the development of drug resistance in users that are already, or become, infected with HIV-1. In such cases, the efficacy of treatments may be compromised since the same (or similar) antiretrovirals used in treatments are being developed as microbicides. To help predict which drug resistance mutations may develop in the context of suboptimal use, HIV-1 primary isolates of different subtypes and different baseline resistance profiles were used to infect primary cells in vitro in the presence of increasing suboptimal concentrations of the two candidate microbicide antiretrovirals dapivirine (DAP) and tenofovir (TFV) alone or in combination. Infections were ongoing for 25 weeks, after which reverse transcriptase genotypes were determined and scrutinized for the presence of any clinically recognized reverse transcriptase drug resistance mutations. Results indicated that suboptimal concentrations of DAP alone facilitated the emergence of common nonnucleoside reverse transcriptase inhibitor resistance mutations, while suboptimal concentrations of DAP plus TFV gave rise to fewer mutations. Suboptimal concentrations of TFV alone did not frequently result in the development of resistance mutations. Sensitivity evaluations for stavudine (d4T), nevirapine (NVP), and lamivudine (3TC) revealed that the selection of resistance as a consequence of suboptimal concentrations of DAP may compromise the potential for NVP to be used in treatment, a finding of potential relevance in developing countries.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral/genetics , HIV-1/drug effects , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , HIV-1/genetics , HIV-1/isolation & purification , Humans , Microbial Sensitivity Tests , Mutation , Organophosphonates/pharmacology , Tenofovir
17.
AIDS ; 25(13): 1585-94, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21633286

ABSTRACT

OBJECTIVE: To evaluate the candidate antiretroviral microbicide compounds, dapivirine (DAP) and tenofovir (TFV), alone and in combination against the transmission of wild-type and nonnucleoside reverse transcriptase inhibitor (NNRTI)-resistant HIV-1 from different subtypes. DESIGN AND METHODS: We determined single-drug efficacy of the RTIs, DAP and TFV, against subtype B and non-B wild-type and NNRTI-resistant HIV-1 in vitro. To assess breadth of activity, compounds were tested alone and in combination against wild-type and NNRTI-resistant subtype C primary HIV-1 isolates and complimentary clonal HIV-1 from subtypes B, C and CRF02_AG to control for viral variation. Early infection was quantified by counting light units emitted from TZM-bl cells less than 48-h postinfection. Combination ratios were based on drug inhibitory concentrations (IC(50)s) and combined effects were determined by calculating combination indices. RESULTS: Both candidate microbicide antiretrovirals demonstrated potent anti-NNRTI-resistant HIV-1 activity in vitro, albeit the combination protected better than the single-drug treatments. Of particular interest, the DAP with TFV combination exhibited synergy (50% combination index, CI(50) = 0.567) against subtype C NNRTI-resistant HIV-1, whereas additivity (CI(50) = 0.987) was observed against the wild-type counterpart from the same patient. The effect was not compounded by the presence of subdominant viral fractions, as experiments using complimentary clonal subtype C wild-type (CI(50) = 0.968) and NNRTI-resistant (CI(50) = 0.672) HIV-1, in lieu of the patient quasispecies, gave similar results. CONCLUSION: This study supports the notion that antiretroviral drug combinations may retain antiviral activity against some drug-resistant HIV-1 despite subtype classification and quasispecies diversity.


Subject(s)
Adenine/analogs & derivatives , Drug Resistance, Viral/drug effects , HIV Infections/virology , HIV-1/drug effects , Organophosphonates/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adenine/pharmacology , Drug Therapy, Combination , HIV Infections/genetics , HIV-1/genetics , Humans , Tenofovir
18.
Virus Res ; 155(1): 189-94, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20937333

ABSTRACT

We demonstrate that a photo-labeled derivative of the non-nucleoside reverse transcriptase inhibitor (NNRTI) dapivirine termed DAPY, when used together with exposure to ultraviolet light, was able to completely and irreversibly inactivate both HIV-1 RT activity as well as infectiousness in each of a T cell line and peripheral blood mononuclear cells. Control experiments using various concentrations of DAPY revealed that a combination of exposure to ultraviolet light together with use of the specific, high affinity photo-labeled compound was necessary for complete inactivation to occur. This method of HIV RT inactivation may have applicability toward preservation of an intact viral structure and warrants further investigation in regard to the potential of this approach to elicit a durable, broad protective immune response.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/drug effects , Photosensitizing Agents/pharmacology , Pyrimidines/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Cells, Cultured , Humans , Leukocytes, Mononuclear/virology , Microbial Viability/drug effects , Microbial Viability/radiation effects , Photosensitizing Agents/chemistry , Pyrimidines/chemistry , Reverse Transcriptase Inhibitors/chemistry , T-Lymphocytes/virology , Ultraviolet Rays
19.
Vaccine ; 27(44): 6137-42, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19715783

ABSTRACT

We describe a new method for the development of a preventive inactivated-HIV vaccine, based on photo-inactivation of HIV reverse transcriptase (RT), which preserves both the conformational and functional integrity of viral surface proteins. The RT of HIV-1 was selectively targeted for inactivation using a photo-labeled compound with specific affinity for HIV-1 RT. The photo-labeled virions were then exposed to UV light causing the photo-labeled compound to form a covalent bond cross-linking the photo-active compound to RT. Replication capacity of the treated virions was significantly reduced when compared to controls suggesting that exposure of treated virions to UV light had caused a stable interaction of RT and the photo-labeling compound.


Subject(s)
AIDS Vaccines/chemistry , HIV Reverse Transcriptase/radiation effects , HIV-1/radiation effects , Reverse Transcriptase Inhibitors/chemistry , Virus Inactivation , Cell Line, Tumor , Cell Survival , Cross-Linking Reagents , HIV Core Protein p24/analysis , HIV Infections , Humans , Inhibitory Concentration 50 , Photoaffinity Labels , Substrate Specificity , Ultraviolet Rays
20.
AIDS Res Hum Retroviruses ; 20(10): 1057-62, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15585096

ABSTRACT

The antiretroviral protease inhibitors indinavir (IDV) and ritonavir (RTV) are used in highly active antiretroviral therapies (HAART). Side effects from long-term HAART therapy include loss of muscle mass. Myoblasts when cultured in media low in growth factors withdraw from the cell cycle, express muscle-specific differentiation inducers and proteins, and fuse to form myotubes. The neutral protease, calpain, is required for myotube formation and RTV decreased calpain activity in vitro. We found lower calpain activity, but not protein, in homogenates of RTV-treated L6 cells than in control cultures. Importantly, L6 and C2C12 myoblasts did not form myotubes when cultured with 10 or 20 microM IDV or RTV. Control and drug-related L6 myoblasts showed identical decreases in proliferating cell nuclear antigens expression indicating proliferation arrest. Similarly, muscle differentiation inducers MyoD and myogenin and their downstream target, myosin heavy chain, were expressed at similar levels in control and drug-treated cells. Thus, whereas muscle differentiation was unaffected by protease inhibitors, calpain activity was reduced and myotube formation prevented. We conclude that RTV and IDV reduced myotube formation by reducing calpain activity. Our data suggest that protease inhibitors included in HAART might be directly involved in muscle wasting by reducing muscle remodeling.


Subject(s)
Calpain/metabolism , Cell Differentiation/drug effects , HIV Protease Inhibitors/pharmacology , Indinavir/pharmacology , Muscle Cells/cytology , Ritonavir/pharmacology , Animals , Cell Line , Cells, Cultured , Mice , Muscle Cells/enzymology , Myoblasts/cytology , Myoblasts/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...