Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 57(6): 2712-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545531

ABSTRACT

Several groups have recently reported on the identification of nucleotide-competing reverse transcriptase inhibitors (NcRTIs), a new class of RT inhibitors. NcRTIs reversibly inhibit binding of the incoming nucleotide to the RT active site but do not act as chain terminators, unlike the nucleos(t)ide reverse transcriptase inhibitor (NRTI) class. We identified a novel benzo[4,5]furo[3,2,d]pyrimidin-2-one NcRTI chemical series. Structure-activity relationship evaluation of this series with both RT and viral replication assays led to the identification of compound A, a new NcRTI. Compound A inhibited HIV-1 RT in a primer extension assay (50% inhibitory concentration, 2.6 nM) but had no measurable activity against human DNA polymerase γ at 10 µM. It potently inhibited HIV-1 replication in vitro (50% effective concentration, 1.5 nM). The antiviral potency of compound A was unaffected by the presence of nonnucleotide RT inhibitor (NNRTI) mutations tested (L100I, K103N/Y181C, V106A, or Y188L). Notably, viruses encoding K65R were hypersusceptible to inhibition by compound A. Compound A also retained full activity against viruses encoding M184V. In vitro selection for resistant virus to compound A led to the selection of a single substitution within RT: W153L. A recombinant virus encoding the RT W153L was highly resistant to compound A (fold change, 160). W153 is a highly conserved residue in HIV RT and has not been previously associated with drug resistance. In summary, a novel NcRTI series with optimized antiviral activity, minimal cross-resistance to existing RT inhibitor classes, and a distinct resistance profile has been discovered. These results further establish NcRTIs as an emerging class of antiretroviral agents.


Subject(s)
Anti-HIV Agents/pharmacology , Benzofurans , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , High-Throughput Screening Assays , Pyrimidinones , Reverse Transcriptase Inhibitors , Anti-HIV Agents/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Benzofurans/pharmacology , Drug Resistance, Viral , HIV-1/genetics , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Pyrimidinones/chemical synthesis , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Structure-Activity Relationship , Virus Replication
2.
Antimicrob Agents Chemother ; 51(8): 2948-53, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517847

ABSTRACT

Apricitabine is a novel deoxycytidine analogue reverse transcriptase inhibitor that is under development for the treatment of human immunodeficiency virus type 1 (HIV-1) infection. Apricitabine is phosphorylated to its active triphosphate by deoxycytidine kinase, which is also responsible for the intracellular phosphorylation of lamivudine (3TC) and emtricitabine (FTC); hence, in vitro studies were performed to investigate possible interactions between apricitabine and these agents. Human peripheral blood mononuclear cells (PBMC) were incubated for 24 h with various concentrations of (3)H-labeled or unlabeled apricitabine, 3TC, or FTC. Intracellular concentrations of parent compounds and their phosphorylated derivatives were measured by high-performance liquid chromatography. In other experiments, viral reverse transcriptase activity was measured in PBMC infected with HIV-1 bearing M184V in the presence of various concentrations of apricitabine and 3TC. [(3)H]apricitabine and [(3)H]3TC were metabolized intracellularly to form mono-, di-, and triphosphates. 3TC and FTC (1 to 10 microM) produced concentration-dependent decreases in apricitabine phosphorylation; in contrast, apricitabine at concentrations of up to 30 muM had no effect on the phosphorylation of 3TC or FTC. The combination of apricitabine and 3TC reduced the antiviral activity of apricitabine against HIV-1: apricitabine concentrations producing 50% inhibition of viral reverse transcriptase were increased two- to fivefold in the presence of 3TC. These findings suggest that nucleoside reverse transcriptase inhibitors with similar modes of action may show biochemical interactions that affect their antiviral efficacy. It is therefore essential that potential interactions between combinations of new and existing agents be thoroughly investigated before such combinations are introduced into clinical practice.


Subject(s)
Anti-HIV Agents/metabolism , Deoxycytidine/analogs & derivatives , HIV-1/drug effects , Leukocytes, Mononuclear/metabolism , Reverse Transcriptase Inhibitors/metabolism , Deoxycytidine/metabolism , Drug Interactions , Emtricitabine , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Lamivudine/metabolism , Male , Microbial Sensitivity Tests
3.
Antimicrob Agents Chemother ; 50(2): 625-31, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16436719

ABSTRACT

SPD754 (AVX754) is a deoxycytidine analogue nucleotide reverse transcriptase inhibitor (NRTI) in clinical development. These studies characterized the in vitro activity of SPD754 against NRTI-resistant human immunodeficiency virus type 1 (HIV-1) and non-clade B HIV-1 isolates, its activity in combination with other antiretrovirals, and its potential myelotoxicity and mitochondrial toxicity. SPD754 was tested against 50 clinical HIV-1 isolates (5 wild-type isolates and 45 NRTI-resistant isolates) in MT-4 cells using the Antivirogram assay. SPD754 susceptibility was reduced 1.2- to 2.2-fold against isolates resistant to zidovudine (M41L, T215Y/F, plus a median of three additional nucleoside analogue mutations [NAMs]) and/or lamivudine (M184V) and was reduced 1.3- to 2.8-fold against isolates resistant to abacavir (L74V, Y115F, and M184V plus one other NAM) or stavudine (V75T/M, M41L, T215F/Y, and four other NAMs). Insertions at amino acid position 69 and Q151M mutations (with or without M184V) reduced SPD754 susceptibility 5.2-fold and 14- to 16-fold, respectively (these changes gave values comparable to or less than the corresponding values for zidovudine, lamivudine, abacavir, and didanosine). SPD754 showed similar activity against isolates of group M HIV-1 clades, including A/G, B, C, D, A(E), D/F, F, and H. SPD754 showed additive effects in combination with other NRTIs, tenofovir, nevirapine, or saquinavir. SPD754 had no significant effects on cell viability or mitochondrial DNA in HepG2 or MT-4 cells during 28-day exposure at concentrations up to 200 microM. SPD754 showed a low potential for myelotoxicity against human bone marrow. In vitro, SPD754 retained activity against most NRTI-resistant HIV-1 clinical isolates and showed a low propensity to cause myelotoxicity and mitochondrial toxicity.


Subject(s)
Anti-HIV Agents/pharmacology , Deoxycytidine/analogs & derivatives , Reverse Transcriptase Inhibitors/pharmacology , Bone Marrow/drug effects , DNA, Mitochondrial/analysis , Deoxycytidine/pharmacology , Deoxycytidine/toxicity , HIV-1/drug effects , Humans , Mitochondria/drug effects
4.
J Org Chem ; 66(25): 8599-605, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11735543

ABSTRACT

The palladium-catalyzed heteroannulation of o-iodoanilines with dienyl sulfones provides a convenient route to vinylogous 2-sulfonylindolines 3. The reaction proceeds in DMF/water in the presence of potassium carbonate and catalytic palladium(II) acetate and is compatible with both electron-donating and -withdrawing substituents in the para position of the aniline, and with an alkyl substituent at C-2 of the dienyl sulfone. The indolines underwent oxidation with DDQ to afford the corresponding indoles 4. The latter were then employed as dienes in Diels-Alder reactions with dimethyl acetylenedicarboxylate (DMAD), methyl propiolate, or methyl acrylate. In the case of the latter two dienophiles, the cycloadditions were highly regioselective, affording the corresponding 1,3-products (with respect to the relative positions of the sulfone and ester groups), exclusively. The cycloadducts from acetylenic dienophiles were converted to the corresponding carbazoles by elimination of the sulfone moiety with DBU, and that from methyl acrylate was subjected to reductive desulfonylation and oxidation to the corresponding carbazole with DDQ. The method thus provides access to carbazoles with various substituents at the 3-, 4-, and 6-positions.

5.
Anal Biochem ; 280(2): 291-300, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10790313

ABSTRACT

Determination of the sensitivity of influenza viruses to neuraminidase (NA) inhibitors is presently based on assays of NA function because, unlike available cell culture methods, the results of such assays are predictive of susceptibility in vivo. At present the most widely used substrate in assays of NA function is the fluorogenic reagent 2'-O-(4-methylumbelliferyl)-N-acetylneuraminic acid (MUN). A rapid assay with improved sensitivity is required because a proportion of clinical isolates has insufficient NA to be detectable in the current fluorogenic assay, and because some mutations associated with resistance to NA inhibitors reduce the activity of the enzyme. A chemiluminescence-based assay of NA activity has been developed that uses a 1,2-dioxetane derivative of sialic acid (NA-STAR) as the substrate. When compared with the fluorogenic assay, use of the NA-STAR substrate results in a 67-fold reduction in the limit of detection of the NA assay, from 200 pM (11 fmol) NA to 3 pM (0.16 fmol) NA. A panel of isolates from phase 2 clinical studies of zanamivir, which were undetectable in the fluorogenic assay, was tested for activity using the NA-STAR substrate. Of these 12 isolates with undetectable NA activity, 10 (83%) were found to have detectable NA activity using the NA-STAR substrate. A comparison of sensitivity to zanamivir of a panel of influenza A and B viruses using the two NA assay methods has been performed. IC(50) values for zanamivir using the NA-STAR were in the range 1.0-7.5 nM and those for the fluorogenic assay in the range 1. 0-5.7 nM (n = 6). The NA-STAR assay is a highly sensitive, rapid assay of influenza virus NA activity that is applicable to monitoring the susceptibility of influenza virus clinical isolates to NA inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Microbial Sensitivity Tests/methods , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/drug effects , Sialic Acids/pharmacology , Adamantane/analogs & derivatives , Enzyme Inhibitors/pharmacology , Guanidines , Luminescent Measurements , Neuraminidase/analysis , Neuraminidase/metabolism , Pyrans , Sensitivity and Specificity , Spectrometry, Fluorescence/methods , Sugar Acids , Zanamivir
6.
Methods Mol Med ; 24: 367-74, 2000.
Article in English | MEDLINE | ID: mdl-21331923

ABSTRACT

Viruses that are less sensitive to the influenza neuraminidase (NA)-specific inhibitor 4-guanidino-Neu5Ac2en (zanamavir) (1) can be isolated after several passages in MDCK cells in the presence of the inhibitor. Although there are three reports of a mutation in the NA gene at the same conserved site, glu119 (2-4), most of the variants have mutations in the hemagglutinin (HA) gene (5). Many of these mutations appear to lower the affinity of the HA for the cellular receptor, so there is less requirement for significant NA activity for the newly synthesized progeny virus to elute. In this chapter we describe noncell culture-based methods for characterization of both HA and NA variants.

7.
Methods Mol Med ; 24: 375-82, 2000.
Article in English | MEDLINE | ID: mdl-21331924

ABSTRACT

The compound 4-guanidino-Neu5Ac2en (zanamivir) has been described as a selective inhibitor of the influenza virus neuraminidase (NA) (1). Viruses that are less sensitive to this inhibitor can be isolated after several passages in MDCK cells in the presence of the inhibitor. Variants isolated so far have had mutations predominantly in the hemagglutinin (HA) gene (2). Many of these mutations appear to lower the affinity of the HA for the cellular receptor, so that there is less requirement for significant NA activity for the newly synthesized progeny virus to elute. There are three reports of a mutation in the NA gene, all at the same conserved site, glu 119 (3-5). In this chapter, the authors describe methods for the isolation of the mutants, and for their characterization in cell culture based assays.

8.
Virology ; 265(2): 286-95, 1999 Dec 20.
Article in English | MEDLINE | ID: mdl-10600600

ABSTRACT

We describe the in vitro selection and characterisation of virus derived from B/Beijing/1/87 passaged in the presence of zanamivir. During zanamivir passage, the phenotype of virus isolates was either drug dependent or drug resistant in plaque reduction assays. The susceptibility of the neuraminidase of the drug-dependent isolates was unchanged from that of the wild-type enzyme. The drug-dependent isolates contained two mutations in the viral haemagglutinin: V90A, close to the proposed secondary sialic acid-binding site, and L240Q, close to the primary sialic acid-binding site. Virus isolates that were drug resistant contained the same mutations in the haemagglutinin but also contained the mutation E116G in the neuraminidase. For the drug-dependent viruses, zanamivir susceptibility could not be measured because plaque numbers increased with increasing drug concentration. The in vitro zanamivir susceptibility of drug-resistant viruses was lower than that of the wild-type virus by a factor of 275- to >2532-fold. Neuraminidase containing the E116G mutation has a 33-fold lower affinity for zanamivir than the wild-type enzyme. The finding that the same haemagglutinin mutations are found in both drug-dependent and drug-resistant viruses confirms that the same changes to the receptor binding function can contribute to both phenotypes. This observation demonstrates the interplay between the influenza virus haemagglutinin and neuraminidase in escape from zanamivir inhibition in vitro.


Subject(s)
Antiviral Agents/pharmacology , Influenza B virus/drug effects , Sialic Acids/pharmacology , Animals , Cell Line , Dogs , Drug Resistance, Microbial , Guanidines , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza B virus/genetics , Influenza B virus/growth & development , Influenza B virus/isolation & purification , Models, Molecular , Neuraminidase/genetics , Neuraminidase/metabolism , Protein Conformation , Pyrans , Zanamivir
9.
Virology ; 263(2): 323-38, 1999 Oct 25.
Article in English | MEDLINE | ID: mdl-10544106

ABSTRACT

The basis of differential sensitivity of replication of influenza viruses to the neuraminidase-specific inhibitor zanamivir was examined using four avian influenza viruses and reassortants produced between them. IC(50) values for inhibition of neuraminidase activity by zanamivir were similar for each of the four viruses, whereas the haemagglutinating activity of each of the viruses was relatively insensitive to zanamivir. However, the four viruses showed distinct zanamivir-sensitivity profiles in tissue culture. Analysis of the reassortant viruses showed that sensitivity was determined by the haemagglutinin gene (segment 4) and the neuraminidase gene (segment 6) and was independent of the remaining six RNA segments. Decreased sensitivity to zanamivir was associated with possession of a haemagglutinin that is released from cells with decreased dependence on neuraminidase and with possession of a neuraminidase that has a short stalk region.


Subject(s)
Antiviral Agents/pharmacology , Genes, Viral/physiology , Hemagglutinins, Viral/metabolism , Influenza A virus/drug effects , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Sialic Acids/pharmacology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Chick Embryo , Chickens/blood , Chickens/virology , Dogs , Erythrocytes/virology , Genes, Viral/genetics , Glycosylation , Guanidines , Hemagglutination, Viral/drug effects , Hemagglutinins, Viral/genetics , Influenza A virus/enzymology , Influenza A virus/genetics , Influenza A virus/physiology , Inhibitory Concentration 50 , Lactose/analogs & derivatives , Lactose/metabolism , Molecular Sequence Data , Mutation/genetics , Neuraminidase/chemistry , Neuraminidase/genetics , Pyrans , Reassortant Viruses/drug effects , Reassortant Viruses/enzymology , Reassortant Viruses/genetics , Reassortant Viruses/physiology , Sialic Acids/metabolism , Substrate Specificity , Virus Replication/drug effects , Zanamivir
10.
Bioorg Med Chem Lett ; 9(4): 601-4, 1999 Feb 22.
Article in English | MEDLINE | ID: mdl-10098672

ABSTRACT

Synthesis of 5R-Acetamido-4S-amino-4H-pyran-6R-O-( -ethyl)propyl and 6R-(1-oxo-2-ethyl)butyl 2-carboxylic acids (4 and 5) and their evaluation as inhibitors of influenza virus sialidase is described. Both compounds showed good inhibitory activity with marked selectivity for influenza A sialidase.


Subject(s)
Enzyme Inhibitors/pharmacology , Ketones/pharmacology , Neuraminidase/antagonists & inhibitors , Pyrans/pharmacology , Sialic Acids/pharmacology , Enzyme Inhibitors/chemistry , Guanidines , Ketones/chemistry , Pyrans/chemistry , Sialic Acids/chemistry , Zanamivir
11.
Bioorg Med Chem Lett ; 9(4): 605-10, 1999 Feb 22.
Article in English | MEDLINE | ID: mdl-10098673

ABSTRACT

Several racemic bicyclo[3.2.1]octene derivatives have been synthesised and evaluated as inhibitors of influenza virus sialidases. The 5-acetamido-bicyclo[3.2.1]octenol 4 showed modest activity against influenza A and B virus sialidases.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Influenza A virus/enzymology , Influenza B virus/enzymology , Neuraminidase/antagonists & inhibitors , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/pharmacology
13.
J Infect Dis ; 178(6): 1592-6, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9815209

ABSTRACT

A recent outbreak of influenza in Hong Kong was caused by a highly virulent virus of avian origin. Concern that the appearance of such a virus in the human population may be a harbinger of a new pandemic has brought increased attention to the issue of antivirals available for treatment of influenza. A/HongKong/156/97 (H5N1), the first virus of H5N1 subtype isolated from a human host, is highly virulent in the mouse model and can infect mouse lungs without requiring adaptation. High mortality and evidence of systemic disease, including spread to the brain after intranasal inoculation, are observed. Zanamivir, a novel neuraminidase inhibitor, is effective at decreasing replication of the virus in vitro. In a model of lethal challenge in mice, zanamivir reduces lung titers of the virus and decreases morbidity and mortality.


Subject(s)
Antiviral Agents/therapeutic use , Influenza A Virus, H5N1 Subtype , Influenza A virus/physiology , Influenza, Human/prevention & control , Sialic Acids/therapeutic use , Animals , Chick Embryo , Female , Guanidines , Hong Kong , Humans , Influenza A virus/drug effects , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Influenza, Human/physiopathology , Lung/virology , Mice , Mice, Inbred BALB C , Organ Specificity , Pyrans , Virulence , Virus Replication/drug effects , Zanamivir
14.
J Infect Dis ; 178(5): 1257-62, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9780244

ABSTRACT

Zanamivir, a neuraminidase inhibitor, has shown promise as a drug to control influenza. During prolonged treatment with zanamivir, a mutant virus was isolated from an immunocompromised child infected with influenza B virus. A hemagglutinin mutation (198 Thr-->Ile) reduced the virus affinity for receptors found on susceptible human cells. A mutation in the neuraminidase active site (152 Arg-->Lys) led to a 1000-fold reduction in the enzyme sensitivity to zanamivir. When tested in ferrets, the mutant virus had less virulence than the parent; however, it had a growth preference over the parent in zanamivir-treated animals. Despite these changes, the sensitivity of the mutant virus to zanamivir assessed by a standard test in MDCK cells was unaffected. These data indicate that the current methods for monitoring resistant mutants are potentially flawed because no tissue culture system adequately reflects the receptor specificity of human respiratory tract epithelium.


Subject(s)
Antiviral Agents/therapeutic use , Immunosuppression Therapy/adverse effects , Influenza B virus , Influenza, Human/drug therapy , Mutation , Orthomyxoviridae Infections/drug therapy , Receptors, Virus/genetics , Sialic Acids/therapeutic use , Amino Acid Substitution , Animals , Bone Marrow Transplantation/immunology , Cell Line , Chlorocebus aethiops , Dogs , Drug Resistance, Microbial/genetics , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Ferrets , Guanidines , Hemagglutination Inhibition Tests , Humans , Infant , Influenza B virus/genetics , Influenza B virus/immunology , Influenza, Human/complications , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Pyrans , RNA, Viral/analysis , Receptors, Virus/drug effects , Vero Cells , Zanamivir
15.
Drug Saf ; 19(3): 233-41, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9747669

ABSTRACT

Developing novel compounds with low toxicity may present more difficulties for pharmaceutical companies than developing compounds with known class-related effects. The absence of clearly identified toxicity may be a consequence either of an inadequate or poorly designed toxicity programme or of the very low toxicity of the novel compound. To enable an informed risk assessment to be undertaken prior to registration, regulatory authorities must satisfy themselves that all efforts to fully evaluate the toxicity profile of a novel compound have been made. Zanamivir is a novel antiviral agent developed for the treatment and prevention of influenza when administered by the oral inhaled route. The toxicology programme for zanamivir was designed to support both a short term treatment indication for patients clinically diagnosed with influenza and a longer term treatment indication for the prevention of influenza. The toxicology studies demonstrated that zanamivir has very low toxicity and no drug-specific toxicities were observed in animal toxicity studies. Systemic plasma concentrations 1336-fold those achieved in clinical use were not associated with significant adverse effects. In the absence of dose-limiting toxicity in animal studies and in an attempt to identify target-organ toxicity, the high dosage level in all repeat dose studies was selected to be the maximum practicable. In the rat, nonspecific effects were seen in the respiratory tract following long term inhaled administration and in the kidneys following continuous infusion. However, these nonspecific effects were consequences of the excessive dosages administered and are not related specifically to zanamivir; thus, they are without relevance to the clinical use of this agent.


Subject(s)
Antiviral Agents/therapeutic use , Influenza, Human/drug therapy , Sialic Acids/therapeutic use , Administration, Inhalation , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Dogs , Drug Evaluation, Preclinical , Female , Guanidines , Humans , Influenza, Human/prevention & control , Injections, Intravenous , Male , Mice , Pyrans , Rats , Rats, Wistar , Sialic Acids/administration & dosage , Sialic Acids/adverse effects , Zanamivir
16.
J Med Chem ; 41(6): 787-97, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9526555

ABSTRACT

4-Amino- and 4-guanidino-4H-pyran-6-carboxamides 4 and 5 related to zanamivir (GG167) are a new class of inhibitors of influenza virus sialidases. Structure--activity studies reveal that, in general, secondary amides are weak inhibitors of both influenza A and B viral sialidases. However, tertiary amides, which contain one or more small alkyl groups, show much greater inhibitory activity, particularly against the influenza A virus enzyme. The sialidase inhibitory activities of these compounds correlate well with their in vitro antiviral efficacy, and several of the most potent analogues displayed useful antiviral activity in vivo when evaluated in a mouse model of influenza A virus infection. Carboxamides which were highly active sialidase inhibitors in vitro also showed good antiviral activity in the mouse efficacy model of influenza A infection when administered intranasally but displayed modest activity when delivered by the intraperitoneal route.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Neuraminidase/antagonists & inhibitors , Pyrans/pharmacology , Sialic Acids/pharmacology , Administration, Intranasal , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Guanidines/chemical synthesis , Guanidines/chemistry , Guanidines/pharmacokinetics , Influenza A virus/enzymology , Influenza B virus/enzymology , Injections, Intraperitoneal , Mice , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/enzymology , Pyrans/chemical synthesis , Pyrans/chemistry , Pyrans/pharmacokinetics , Sialic Acids/chemistry , Sialic Acids/pharmacokinetics , Structure-Activity Relationship , Zanamivir
17.
J Med Chem ; 41(6): 798-807, 1998 Mar 12.
Article in English | MEDLINE | ID: mdl-9526556

ABSTRACT

The first paper in this series (see previous article) described structure-activity studies of carboxamide analogues of zanamivir binding to influenza virus sialidase types A and B and showed that inhibitory activity of these compounds was much greater against influenza A enzyme. To understand the large differences in affinities, a number of protein-ligand complexes have been investigated using crystallography and molecular dynamics. The crystallographic studies show that the binding of ligands containing tertiary amide groups is accompanied by the formation of an intramolecular planar salt bridge between two amino acid residues in the active site of the enzyme. It is proposed that the unexpected strong binding of these inhibitors is a result of the burial of hydrophobic surface area and salt-bridge formation in an environment of low dielectric. In sialidase from type A virus, binding of the carboxamide moeity and salt-bridge formation have only a minor effect on the positions of the surrounding residues, whereas in type B enzyme, significant distortion of the protein is observed. The results suggest that the decreased affinity in enzyme from influenza B is directly correlated with the small changes that occur in the amino acid residue interactions accompanying ligand binding. Molecular dynamics calculations have shown that the tendency for salt-bridge formation is greater in influenza A sialidase than influenza B sialidase and that this tendency is a useful descriptor for the prediction of inhibitor potency.


Subject(s)
Acetamides/chemistry , Antiviral Agents/chemistry , Enzyme Inhibitors/chemistry , Influenza A virus/enzymology , Influenza B virus/enzymology , Neuraminidase/chemistry , Pyrans/chemistry , Sialic Acids/chemistry , Acetamides/metabolism , Acetamides/pharmacology , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Guanidines , Models, Molecular , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Protein Conformation , Pyrans/metabolism , Pyrans/pharmacology , Sialic Acids/metabolism , Sialic Acids/pharmacology , Zanamivir
18.
J Virol ; 72(3): 2456-62, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9499107

ABSTRACT

The influenza virus neuraminidase (NA)-specific inhibitor zanamivir (4-guanidino-Neu5Ac2en) is effective in humans when administered topically within the respiratory tract. The search for compounds with altered pharmacological properties has led to the identification of a novel series of influenza virus NA inhibitors in which the triol group of zanamivir has been replaced by a hydrophobic group linked by a carboxamide at the 6 position (6-carboxamide). NWS/G70C variants generated in vitro, with decreased sensitivity to 6-carboxamide, contained hemagglutinin (HA) and/or NA mutations. HA mutants bound with a decreased efficiency to the cellular receptor and were cross-resistant to all the NA inhibitors tested. The NA mutation, an Arg-to-Lys mutation, was in a previously conserved site, Arg292, which forms part of a triarginyl cluster in the catalytic site. In enzyme assays, the NA was equally resistant to zanamivir and 4-amino-Neu5Ac2en but showed greater resistance to 6-carboxamide and was most resistant to a new carbocyclic NA inhibitor, GS4071, which also has a hydrophobic side chain at the 6 position. Consistent with enzyme assays, the lowest resistance in cell culture was seen to zanamivir, more resistance was seen to 6-carboxamide, and the greatest resistance was seen to GS4071. Substrate binding and enzyme activity were also decreased in the mutant, and consequently, virus replication in both plaque assays and liquid culture was compromised. Altered binding of the hydrophobic side chain at the 6 position or the triol group could account for the decreased binding of both the NA inhibitors and substrate.


Subject(s)
Conserved Sequence , Enzyme Inhibitors/pharmacology , Influenza A virus/enzymology , Mutation , N-Acetylneuraminic Acid/analogs & derivatives , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Acetamides/chemistry , Acetamides/pharmacology , Adsorption , Animals , Binding Sites , Birds , Cell Line , Dogs , Drug Resistance, Microbial , Enzyme Inhibitors/chemistry , Guanidines , Heating , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A virus/growth & development , Influenza A virus/metabolism , Kinetics , Molecular Structure , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/pharmacology , Oseltamivir , Phenotype , Pyrans , Sialic Acids/chemistry , Sialic Acids/pharmacology , Substrate Specificity , Viral Plaque Assay , Virus Replication , Zanamivir
19.
Bioorg Med Chem Lett ; 8(19): 2623-8, 1998 Oct 06.
Article in English | MEDLINE | ID: mdl-9873592

ABSTRACT

A series of biaryl acids has been found to show micromolar inhibition of the HIV reverse transcriptase (RT) from types 1 and 2 with IC50S in the micromolar range. The series was discovered by consideration of the polymerase active site and sub-structure searching of the company compound collection. Synthesis of analogues to investigate the SAR is described. Two of these compounds have shown inhibition of HIV-2 RT only.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , RNA-Directed DNA Polymerase/drug effects , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Anti-HIV Agents/chemical synthesis , Carboxylic Acids/chemical synthesis , Hydrocarbons, Aromatic/chemical synthesis , Hydrocarbons, Aromatic/chemistry , Hydrocarbons, Aromatic/pharmacology , Reverse Transcriptase Inhibitors/chemical synthesis , Structure-Activity Relationship
20.
Virology ; 238(2): 265-72, 1997 Nov 24.
Article in English | MEDLINE | ID: mdl-9400599

ABSTRACT

4-Guanidino-2,4-dideoxy-2,3-dehydro-N-acetylneuraminic acid (4-guanidino-Neu5Ac2en) specifically inhibits the influenza virus neuraminidase (NA) through interaction of the guanidino group with conserved Glu 119 and Glu 227 residues in the substrate binding pocket of the enzyme. To understand the mechanism by which influenza viruses become resistant to 4-guanidino-Neu5Ac2en, we investigated mutations at amino acid residues 119 and 227 in the influenza virus NA for their effects on this compound and on NA activity. The NA gene was cloned from the NWS-G70c virus, and mutations were introduced at the codon for amino acid residue 119 or 227. All of the 13 mutants containing a change at residue 119 were transported to the cell surface, although their expression levels ranged from 68.2 to 91.3% of wild type. Mutant NAs that retained at least 20% of the wild-type enzymatic activity were tested for their sensitivity to 4-guanidino-Neu5Ac2en and found to be sevenfold less sensitive to this compound than was the wild-type NA. By contrast, only 6 of 13 mutants defined by modifications at residue 227 were transported to the cell surface, and those NAs lacked substantial enzymatic activity (9% of wild type, at most). These results suggest that only a limited number of resistant viruses arise through mutations at Glu 119 and Glu 227 under selective pressure from 4-guanidino-Neu5Ac2en and that the development of compounds which interact with 227 Glu more strongly than does 4-guanidino-Neu5Ac2en may reduce the likelihood of drug-resistant viruses still further.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Influenza A virus/enzymology , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Sialic Acids/pharmacology , Cell Line, Transformed , Gene Expression , Glutamic Acid/genetics , Glutamic Acid/metabolism , Guanidines , Humans , Influenza A virus/drug effects , Influenza A virus/genetics , Mutagenesis, Site-Directed , Neuraminidase/biosynthesis , Pyrans , Zanamivir
SELECTION OF CITATIONS
SEARCH DETAIL
...