Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Antimicrob Agents Chemother ; 51(12): 4366-73, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17908951

ABSTRACT

Rupintrivir (formerly AG7088) is an irreversible inhibitor of the human rhinovirus (HRV) 3C protease that has been demonstrated to have in vitro activity against all HRVs tested, consistent with its interaction with a strictly conserved subset of amino acids in the 3C protease. The potential for resistance was studied following in vitro serial passage of HRV serotypes 14, 2, 39, and Hanks in the presence of increasing rupintrivir concentrations. HRV variants with reduced susceptibilities to rupintrivir (sevenfold for HRV 14) or with no significant reductions in susceptibility but genotypic changes (HRV 2, 39, and Hanks) were initially isolated following 14 to 40 cumulative days in culture (three to six passages). Sequence analysis of the 3C protease identified one to three substitutions in diverse patterns but with common features (T129T/A, T131T/A, and T143P/S in HRV 14; N165T in HRV 2; N130N/K and L136L/F in HRV 39; T130A in HRV Hanks). Notably, three of the four HRV variants contained a substitution at residue 130 (residue 129 in HRV 14). Continued selection in the presence of escalating concentrations of rupintrivir (40 to 72 days) resulted in the accumulation of additional mutations (A121A/V and Y139Y/H in HRV 14, E3E/G and A103A/V in HRV 2, S105T in HRV 39), with only minimal further reductions in susceptibility (up to fivefold). The ability of specific substitutions to confer resistance was examined by susceptibility testing of HRV 14 variants constructed to contain 3C protease mutations. In summary, the slow accumulation of multiple amino acid substitutions with only minimal to moderate reductions in susceptibility highlight the advantages of 3C protease as an antiviral target.


Subject(s)
Antiviral Agents/pharmacology , Isoxazoles/pharmacology , Pyrrolidinones/pharmacology , Rhinovirus/drug effects , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Amino Acid Sequence , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Drug Resistance, Viral/genetics , Genotype , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Mutation , Phenotype , Phenylalanine/analogs & derivatives , Rhinovirus/enzymology , Rhinovirus/genetics , Sequence Homology, Amino Acid , Valine/analogs & derivatives , Viral Proteins/genetics , Viral Proteins/metabolism
2.
Antimicrob Agents Chemother ; 49(2): 619-26, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15673742

ABSTRACT

The picornavirus 3C protease is required for the majority of proteolytic cleavages that occur during the viral life cycle. Comparisons of published amino acid sequences from 6 human rhinoviruses (HRV) and 20 human enteroviruses (HEV) show considerable variability in the 3C protease-coding region but strict conservation of the catalytic triad residues. Rupintrivir (formerly AG7088) is an irreversible inhibitor of HRV 3C protease with potent in vitro activity against all HRV serotypes (48 of 48), HEV strains (4 of 4), and untyped HRV field isolates (46 of 46) tested. To better understand the relationship between in vitro antiviral activity and 3C protease-rupintrivir binding interactions, we performed nucleotide sequence analyses on an additional 21 HRV serotypes and 11 HRV clinical isolates. Antiviral activity was also determined for 23 HRV clinical isolates and four additional HEV strains. Sequence comparison of 3C proteases (n = 58) show that 13 and 11 of the 14 amino acids that are involved in side chain interactions with rupintrivir are strictly conserved among HRV and HEV, respectively. These sequence analyses are consistent with the comparable in vitro antiviral potencies of rupintrivir against all HRV serotypes, HRV isolates, and HEV strains tested (50% effective concentration range, 3 to 183 nM; n = 125). In summary, the conservation of critical amino acid residues in 3C protease and the observation of potent, broad-spectrum antipicornavirus activity of rupintrivir highlight the advantages of 3C protease as an antiviral target.


Subject(s)
Amino Acids/metabolism , Antiviral Agents/pharmacology , Cysteine Endopeptidases/metabolism , Isoxazoles/pharmacology , Protease Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Rhinovirus/enzymology , Rhinovirus/genetics , Viral Proteins/metabolism , 3C Viral Proteases , Conserved Sequence , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/genetics , HeLa Cells , Humans , Molecular Sequence Data , Phenylalanine/analogs & derivatives , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Valine/analogs & derivatives , Viral Proteins/drug effects , Viral Proteins/genetics
3.
Bioorg Med Chem Lett ; 11(20): 2683-6, 2001 Oct 22.
Article in English | MEDLINE | ID: mdl-11591501

ABSTRACT

Novel tripeptidyl C-terminal Michael acceptors with an ester replacement of the P(2)-P(3) amide bond were investigated as irreversible inhibitors of the human rhinovirus (HRV) 3C protease (3CP). When screened against HRV serotype-14 the best compound was shown to have very good 3CP inhibition (k(obs)/[I]=270,000M(-1)s(-1)) and potent in vitro antiviral activity (EC(50)=7.0nM).


Subject(s)
Peptides/chemical synthesis , Protease Inhibitors/chemical synthesis , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cysteine Endopeptidases/metabolism , Microbial Sensitivity Tests , Models, Molecular , Peptides/chemistry , Peptides/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Rhinovirus/drug effects , Structure-Activity Relationship , Viral Proteins/metabolism
4.
J Clin Virol ; 21(2): 105-17, 2001 May.
Article in English | MEDLINE | ID: mdl-11378491

ABSTRACT

BACKGROUND: More than 100 immunologically distinct serotypes of human rhinoviruses (HRV) have been discovered, making detection of surface exposed capsid antigens impractical. However, the non-structural protein 3C protease (3Cpro) is essential for viral replication and is relatively highly conserved among serotypes, making it a potential target for diagnostic testing. The thin film biosensor is an assay platform that can be formatted into a sensitive immunoassay for viral proteins in clinical specimens. The technology utilizes an optically coated silicon surface to convert specific molecular binding events into visual color changes by altering the reflective properties of light through molecular thin films. OBJECTIVE: To develop a rapid test for detection of HRV by developing broadly serotype reactive antibodies to 3Cpro and utilizing them in the thin film biosensor format. STUDY DESIGN: Polyclonal antibodies to 3Cpro were purified and incorporated into the thin film assay. The in vitro sensitivity, specificity and multiserotype cross-reactivity of the 3Cpro assay were tested. Nasal washes from naturally infected individuals were also tested to verify that 3Cpro was detectable in clinical specimens. RESULTS: The 3Cpro assay is a 28-min, non-instrumented room temperature test with a visual limit of detection of 12 pM (picomolar) 3Cpro. In terms of viral titer, as few as 1000 TCID(50) equivalents of HRV2 were detectable. The assay detected 45/52 (87%) of the HRV serotypes tested but showed no cross-reactivity to common respiratory viruses or bacteria. The thin film assay detected 3Cpro in HRV-infected cell culture supernatants coincident with first appearance of cytopathic effect. Data are also presented demonstrating 3Cpro detection from clinical samples collected from HRV-infected individuals. The assay detected 3Cpro in expelled nasal secretions from a symptomatic individual on the first day of illness. In addition, 9/11 (82%) concentrated nasal wash specimens from HRV infected children were positive in the 3Cpro test. CONCLUSION: We have described a novel, sensitive thin film biosensor for rapid detection of HRV 3Cpro. This test may be suitable for the point of care setting, where rapid HRV diagnostic test results could contribute to clinical decisions regarding appropriate antibiotic or antiviral therapy.


Subject(s)
Cysteine Endopeptidases/analysis , Immunoenzyme Techniques/methods , Rhinovirus/isolation & purification , Viral Proteins , 3C Viral Proteases , Antibodies, Viral/immunology , Biosensing Techniques , Common Cold/diagnosis , Common Cold/virology , Cross Reactions , Cysteine Endopeptidases/immunology , HeLa Cells , Humans , Immune Sera , Nasal Lavage Fluid/virology , Optics and Photonics , Picornaviridae Infections/diagnosis , Picornaviridae Infections/virology , Rhinovirus/enzymology , Rhinovirus/immunology , Rhinovirus/physiology , Sensitivity and Specificity , Serotyping , Silicon , Virus Replication
5.
Antimicrob Agents Chemother ; 45(4): 1086-93, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11257019

ABSTRACT

Nelfinavir mesylate (Viracept, formally AG1343) is a potent and orally bioavailable human immunodeficiency virus (HIV) type 1 (HIV-1) protease inhibitor (K(i) = 2 nM) and is being widely prescribed in combination with HIV reverse transcriptase inhibitors for the treatment of HIV infection. The current studies evaluated the presence of metabolites circulating in plasma following the oral administration of nelfinavir to healthy volunteers and HIV-infected patients, as well as the levels in plasma and antiviral activities of these metabolites. The results showed that the parent drug was the major circulating chemical species, followed in decreasing abundance by its hydroxy-t-butylamide metabolite (M8) and 3'-methoxy-4'-hydroxynelfinavir (M1). Antiviral assays with HIV-1 strain RF-infected CEM-SS cells showed that the 50% effective concentrations (EC50) of nelfinavir, M8, and M1 were 30, 34, and 151 nM, respectively, and that the corresponding EC50 against another HIV-1 strain, IIIB, in MT-2 cells were 60, 86, and 653 nM. Therefore, apparently similar in vitro antiviral activities were demonstrated for nelfinavir and M8, whereas an approximately 5- to 11-fold-lower level of antiviral activity was observed for M1. The active metabolite, M8, showed a degree of binding to human plasma proteins similar to that of nelfinavir (ca. 98%). Concentrations in plasma of nelfinavir and its metabolites in 10 HIV-positive patients receiving nelfinavir therapy (750 mg three times per day) were determined by a liquid chromatography tandem mass spectrometry assay. At steady state (day 28), the mean plasma nelfinavir concentrations ranged from 1.73 to 4.96 microM and the M8 concentrations ranged from 0.55 to 1.96 microM, whereas the M1 concentrations were low and ranged from 0.09 to 0.19 microM. In conclusion, the findings from the current studies suggest that, in humans, nelfinavir forms an active metabolite circulating at appreciable levels in plasma. The active metabolite M8 may account for some of the antiviral activity associated with nelfinavir in the treatment of HIV disease.


Subject(s)
HIV Protease Inhibitors/blood , HIV Seropositivity/metabolism , HIV-1/drug effects , Nelfinavir/blood , Blood Proteins/metabolism , Cell Line , Chromatography, Liquid , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Humans , Kinetics , Male , Mass Spectrometry , Microbial Sensitivity Tests , Nelfinavir/chemistry , Nelfinavir/pharmacology
6.
J Infect Dis ; 182(2): 420-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10915071

ABSTRACT

The relationship between detectable human immunodeficiency virus (HIV) genotypic resistance and virologic response was compared in patients receiving nelfinavir as monotherapy (16 weeks) or in combination with lamuvidine and zidovudine (48 weeks). Two patient groups were defined on the basis of the presence or absence of substitutions associated with nelfinavir, a protease (PR) inhibitor, and/or a reverse transcriptase (RT) inhibitor. HIV RNA levels <50 copies/mL were achieved in 17 (85%) of 20 combination-therapy patients without genotypic resistance (PR-RT(-)) versus only 1 (17%) of 6 patients with genotypic resistance (PR-RT(+)). PR-RT(-) patients exhibited greater and more durable virus suppression compared with PR-RT(+) patients. All 6 PR-RT(+) patients had virus with M184V (lamuvidine resistance); 3 isolates also contained D30N (nelfinavir resistance). M184V preceded D30N in all determinable instances. In this study, suppression of HIV replication to <50 copies/mL was associated with durable response and reduced incidence of resistance. Results also indicate that combination regimens can fail despite the absence of detectable genotypic PR resistance.


Subject(s)
HIV Infections/drug therapy , HIV/genetics , Lamivudine/therapeutic use , Nelfinavir/therapeutic use , Zidovudine/therapeutic use , Anti-HIV Agents/therapeutic use , CD4 Lymphocyte Count , Clinical Trials as Topic , Drug Resistance, Microbial/genetics , Drug Therapy, Combination , Genotype , HIV Protease Inhibitors/therapeutic use , Humans , Mutation , RNA, Viral/blood , RNA-Directed DNA Polymerase/genetics , Reverse Transcriptase Inhibitors/therapeutic use
7.
J Med Chem ; 43(9): 1670-83, 2000 May 04.
Article in English | MEDLINE | ID: mdl-10794684

ABSTRACT

A series of nonpeptide benzamide-containing inhibitors of human rhinovirus (HRV) 3C protease was identified using structure-based design. The design, synthesis, and biological evaluation of these inhibitors are reported. A Michael acceptor was combined with a benzamide core mimicking the P1 recognition element of the natural 3CP substrate. alpha,beta-Unsaturated cinnamate esters irreversibly inhibited the 3CP and displayed antiviral activity (EC(50) 0.60 microM, HRV-16 infected H1-HeLa cells). On the basis of cocrystal structure information, a library of substituted benzamide derivatives was prepared using parallel synthesis on solid support. A 1.9 A cocrystal structure of a benzamide inhibitor in complex with the 3CP revealed a binding mode similar to that initially modeled wherein covalent attachment of the nucleophilic cysteine residue is observed. Unsaturated ketones displayed potent reversible inhibition but were inactive in the cellular antiviral assay and were found to react with nucleophilic thiols such as DTT.


Subject(s)
Benzamides/chemical synthesis , Benzamides/pharmacology , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Drug Design , Humans , Protein Conformation , Rhinovirus/drug effects , Structure-Activity Relationship
8.
Antimicrob Agents Chemother ; 44(5): 1236-41, 2000 May.
Article in English | MEDLINE | ID: mdl-10770757

ABSTRACT

Symptom severity in patients with human rhinovirus (HRV)-induced respiratory illness is associated with elevated levels of the inflammatory cytokines interleukin-6 (IL-6) and IL-8. AG7088 is a novel, irreversible inhibitor of the HRV 3C protease. In this study, AG7088 was tested for its antiviral activity and ability to inhibit the production of IL-6 and IL-8 in a human bronchial epithelial cell line, BEAS-2B. Infection of BEAS-2B cells with HRV 14 resulted in the production of both infectious virus and the cytokines IL-6 and IL-8. Treatment of HRV 14-infected cells with AG7088 resulted in a statistically significant (P, <0.05) dose-dependent reduction in the levels of infectious virus as well as IL-6 and IL-8 released into the cell supernatant compared to the results obtained for compound-free infected cells. AG7088 was also able to inhibit the replication of HRV 2 and 16 in BEAS-2B cells. In time-of-addition studies, AG7088 could be added as late as 14 to 26 h after HRV 14 infection of BEAS-2B cells and still result in a statistically significant (P, <0.05) reduction in the levels of infectious virus, IL-6, and IL-8 compared to the results obtained for compound-free infected cells. These findings have implications for the development of an antirhinovirus agent that may not only block virus replication but also diminish symptoms.


Subject(s)
Antiviral Agents/pharmacology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytokines/metabolism , Isoxazoles/pharmacology , Pyrrolidinones/pharmacology , Rhinovirus/drug effects , Viral Proteins , 3C Viral Proteases , Cells, Cultured , Cysteine Endopeptidases/drug effects , Cytokines/antagonists & inhibitors , HeLa Cells , Humans , Microbial Sensitivity Tests , Phenylalanine/analogs & derivatives , Rhinovirus/enzymology , Rhinovirus/metabolism , Time Factors , Valine/analogs & derivatives
9.
Bioorg Med Chem Lett ; 10(1): 45-8, 2000 Jan 03.
Article in English | MEDLINE | ID: mdl-10636240

ABSTRACT

Tripeptide-derived molecules incorporating C-terminal ketone electrophiles were evaluated as reversible inhibitors of the cysteine-containing human rhinovirus 3C protease (3CP). An optimized example of such compounds displayed potent 3CP inhibition activity (K = 0.0045 microM) and in vitro antiviral properties (EC50=0.34 microM) when tested against HRV serotype-14.


Subject(s)
Antiviral Agents/chemical synthesis , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Ketones/chemical synthesis , Oligopeptides/chemical synthesis , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Antiviral Agents/pharmacology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Drug Design , Humans , Inhibitory Concentration 50 , Ketones/pharmacology , Kinetics , Oligopeptides/pharmacology , Rhinovirus/drug effects , Structure-Activity Relationship
10.
Antimicrob Agents Chemother ; 43(10): 2444-50, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508022

ABSTRACT

AG7088 is a potent, irreversible inhibitor of human rhinovirus (HRV) 3C protease (inactivation rate constant (k(obs)/[I]) = 1,470,000 +/- 440,000 M(-1) s(-1) for HRV 14) that was discovered by protein structure-based drug design methodologies. In H1-HeLa and MRC-5 cell protection assays, AG7088 inhibited the replication of all HRV serotypes (48 of 48) tested with a mean 50% effective concentration (EC(50)) of 0.023 microM (range, 0.003 to 0.081 microM) and a mean EC(90) of 0.082 microM (range, 0.018 to 0.261 microM) as well as that of related picornaviruses including coxsackieviruses A21 and B3, enterovirus 70, and echovirus 11. No significant reductions in the antiviral activity of AG7088 were observed when assays were performed in the presence of alpha(1)-acid glycoprotein or mucin, proteins present in nasal secretions. The 50% cytotoxic concentration of AG7088 was >1,000 microM, yielding a therapeutic index of >12,346 to >333,333. In a single-cycle, time-of-addition assay, AG7088 demonstrated antiviral activity when added up to 6 h after infection. In contrast, a compound targeting viral attachment and/or uncoating was effective only when added at the initiation of virus infection. Direct inhibition of 3C proteolytic activity in infected cells treated with AG7088 was demonstrated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis analysis of radiolabeled proteins, which showed a dose-dependent accumulation of viral precursor polyproteins and reduction of processed protein products. The broad spectrum of antiviral activity of AG7088, combined with its efficacy even when added late in the virus life cycle, highlights the advantages of 3C protease as a target and suggests that AG7088 will be a promising clinical candidate.


Subject(s)
Antiviral Agents/pharmacology , Cysteine Endopeptidases/drug effects , Isoxazoles/pharmacology , Pyrrolidinones/pharmacology , Rhinovirus/drug effects , Viral Proteins , 3C Viral Proteases , Cell Division/drug effects , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , HeLa Cells , Humans , Microbial Sensitivity Tests , Phenylalanine/analogs & derivatives , Proteins/pharmacology , Rhinovirus/physiology , Serotyping , Valine/analogs & derivatives
11.
Proc Natl Acad Sci U S A ; 96(20): 11000-7, 1999 Sep 28.
Article in English | MEDLINE | ID: mdl-10500114

ABSTRACT

Human rhinoviruses, the most important etiologic agents of the common cold, are messenger-active single-stranded monocistronic RNA viruses that have evolved a highly complex cascade of proteolytic processing events to control viral gene expression and replication. Most maturation cleavages within the precursor polyprotein are mediated by rhinovirus 3C protease (or its immediate precursor, 3CD), a cysteine protease with a trypsin-like polypeptide fold. High-resolution crystal structures of the enzyme from three viral serotypes have been used for the design and elaboration of 3C protease inhibitors representing different structural and chemical classes. Inhibitors having alpha,beta-unsaturated carbonyl groups combined with peptidyl-binding elements specific for 3C protease undergo a Michael reaction mediated by nucleophilic addition of the enzyme's catalytic Cys-147, resulting in covalent-bond formation and irreversible inactivation of the viral protease. Direct inhibition of 3C proteolytic activity in virally infected cells treated with these compounds can be inferred from dose-dependent accumulations of viral precursor polyproteins as determined by SDS/PAGE analysis of radiolabeled proteins. Cocrystal-structure-assisted optimization of 3C-protease-directed Michael acceptors has yielded molecules having extremely rapid in vitro inactivation of the viral protease, potent antiviral activity against multiple rhinovirus serotypes and low cellular toxicity. Recently, one compound in this series, AG7088, has entered clinical trials.


Subject(s)
Antiviral Agents/pharmacology , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Isoxazoles/pharmacology , Pyrrolidinones/pharmacology , Rhinovirus/drug effects , Viral Proteins , 3C Viral Proteases , Amino Acid Sequence , Binding Sites , Crystallization , Drug Design , Humans , Isoxazoles/chemistry , Molecular Sequence Data , Phenylalanine/analogs & derivatives , Pyrrolidinones/chemistry , Rhinovirus/enzymology , Structure-Activity Relationship , Valine/analogs & derivatives
12.
Bioorg Med Chem Lett ; 9(15): 2189-94, 1999 Aug 02.
Article in English | MEDLINE | ID: mdl-10465543

ABSTRACT

Tripeptide-derived molecules incorporating N-methyl amino acid residues and C-terminal Michael acceptor moieties were evaluated as irreversible inhibitors of the cysteine-containing human rhinovirus 3C protease (3CP). Such compounds displayed good 3CP inhibition activity (k(obs)/[I] up to 610,000 M(-1) s(-1)) and potent in vitro antiviral properties (EC50 approaching 0.03 microM) when tested against HRV serotype-14.


Subject(s)
Cysteine Endopeptidases/metabolism , Protease Inhibitors/chemical synthesis , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Cysteine Endopeptidases/drug effects , Drug Design , Humans , Peptides/chemical synthesis , Peptides/pharmacology , Protease Inhibitors/pharmacology , Rhinovirus/drug effects , Structure-Activity Relationship
13.
Bioorg Med Chem ; 7(4): 589-98, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10353638

ABSTRACT

The optimization of a series of irreversible human rhinovirus (HRV) 3C protease (3CP) inhibitors is described. These inhibitors are comprised of an L-Leu-L-Phe-L-Gln tripeptide containing an N-terminal amide moiety and a C-terminal ethyl propenoate Michael acceptor. Examination of approximately 500 compounds with varying N-terminal amides utilizing solid-phase synthesis and high-throughput assay techniques is described along with the solution phase preparation of several highly active molecules. A tripeptide Michael acceptor containing an N-terminal amide derived from 5-methylisoxazole-3-carboxylic acid is shown to exhibit potent, irreversible anti-3CP activity (k(obs)/[I] = 260,000 M(-1) s(-1); type-14 3CP) and broad-spectrum antirhinoviral properties (average EC50 = 0.47 microM against four different HRV serotypes).


Subject(s)
Amides/chemistry , Cysteine Endopeptidases/chemistry , Peptides/chemistry , Protease Inhibitors/chemical synthesis , Viral Proteins , 3C Viral Proteases , HeLa Cells , Humans , Kinetics
14.
J Virol ; 73(7): 6197-202, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364383

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) amino acid substitutions observed during antiretroviral drug therapy may be caused by drug selection, non-drug-related evolution, or sampling error introduced by the sequencing process. We analyzed HIV-1 sequences from 371 untreated patients and from 178 patients receiving a single protease inhibitor. Amino acid substitution patterns during treatment were compared with inferred substitution patterns arising evolutionarily without treatment. Our results suggest that most treatment-associated amino acid substitutions are caused by selective drug pressure, including substitutions not previously associated with drug resistance.


Subject(s)
Amino Acid Substitution , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/therapeutic use , HIV Protease/genetics , HIV-1/genetics , Amino Acid Sequence , Genetic Variation , HIV Protease/drug effects , HIV-1/drug effects , HIV-1/isolation & purification , Humans , Indinavir/therapeutic use , Molecular Sequence Data , Nelfinavir/therapeutic use , Ritonavir/therapeutic use , Saquinavir/therapeutic use
15.
AIDS ; 13(2): F23-8, 1999 Feb 04.
Article in English | MEDLINE | ID: mdl-10202820

ABSTRACT

OBJECTIVES: The effectiveness of a second protease inhibitor in patients who failed an initial protease inhibitor is unclear but believed to be low. It has been postulated, however, that patients who fail nelfinavir may respond differently. We therefore assessed the virologic response to a ritonavir-saquinavir-containing regimen in patients who had previously failed nelfinavir. METHODS: A total of 26 patients enrolled in the nelfinavir clinical trials AG506 and AG511 at our two sites who failed (two consecutive HIV viral loads > 5000 copies/ml; branched DNA assay) were switched to a combination of stavudine 40 mg twice daily, lamivudine 150 mg twice daily, ritonavir 400 mg twice daily and saquinavir 400 mg twice daily. RESULTS: The mean viral load at enrollment in this study was 46 674 copies/ml (range, 1075-146400 copies/ml). The median CD4 cell count was 222 x 10(6)/l (range, 82-448 x 10(6)/l). The median duration of nelfinavir use with a detectable viral load before the switch occurred was 48 weeks. Two patients discontinued the study at 3 weeks. All of the remaining patients (n = 24) reached undetectable viral loads (< 500 copies/ml) that were sustained at week 24 in 17 (71%) out of 24 subjects. The most frequent baseline mutations in the protease gene prior to switching were D30N (13 out of 18), N88D (eight out of 18) and M36I (eight out of 18). The presence or absence of these mutations was not predictive of a short-term virologic response. CONCLUSIONS: Most patients who failed a nelfinavir-containing regimen responded to a switch to a combination regimen with saquinavir-ritonavir.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/therapeutic use , HIV-1 , Nelfinavir/therapeutic use , Ritonavir/therapeutic use , Saquinavir/therapeutic use , Viral Load , CD4 Lymphocyte Count , Drug Therapy, Combination , HIV Infections/immunology , HIV Protease/genetics , HIV-1/genetics , Humans , Predictive Value of Tests , Prospective Studies , RNA, Viral , Treatment Failure
16.
J Med Chem ; 42(7): 1203-12, 1999 Apr 08.
Article in English | MEDLINE | ID: mdl-10197964

ABSTRACT

The structure-based design, chemical synthesis, and biological evaluation of various ketomethylene-containing human rhinovirus (HRV) 3C protease (3CP) inhibitors are described. These compounds are comprised of a peptidomimetic binding determinant and an ethyl propenoate Michael acceptor moiety which forms an irreversible covalent adduct with the active site cysteine residue of the 3C enzyme. The ketomethylene-containing inhibitors typically display slightly reduced 3CP inhibition activity relative to the corresponding peptide-derived molecules, but they also exhibit significantly improved antiviral properties. Optimization of the ketomethylene-containing compounds is shown to provide several highly active 3C protease inhibitors which function as potent antirhinoviral agents (EC90 = <1 microM) against multiple virus serotypes in cell culture.


Subject(s)
Antiviral Agents/chemical synthesis , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Dipeptides/chemical synthesis , Ketones/chemical synthesis , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/chemistry , Dipeptides/pharmacology , Drug Design , Humans , Ketones/chemistry , Ketones/pharmacology , Molecular Mimicry , Rhinovirus/drug effects , Structure-Activity Relationship
17.
J Med Chem ; 42(7): 1213-24, 1999 Apr 08.
Article in English | MEDLINE | ID: mdl-10197965

ABSTRACT

The structure-based design, chemical synthesis, and biological evaluation of various human rhinovirus (HRV) 3C protease (3CP) inhibitors which incorporate P1 lactam moieties in lieu of an L-glutamine residue are described. These compounds are comprised of a tripeptidyl or peptidomimetic binding determinant and an ethyl propenoate Michael acceptor moiety which forms an irreversible covalent adduct with the active site cysteine residue of the 3C enzyme. The P1-lactam-containing inhibitors display significantly increased 3CP inhibition activity along with improved antirhinoviral properties relative to corresponding L-glutamine-derived molecules. In addition, several lactam-containing compounds exhibit excellent selectivity for HRV 3CP over several other serine and cysteine proteases and are not appreciably degraded by a variety of biological agents. One of the most potent inhibitors (AG7088, mean antirhinoviral EC90 approximately 0.10 microM, n = 46 serotypes) is shown to warrant additional preclinical development to explore its potential for use as an antirhinoviral agent.


Subject(s)
Antiviral Agents/chemical synthesis , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Glutamine/chemistry , Isoxazoles/chemical synthesis , Lactams/chemical synthesis , Oligopeptides/chemical synthesis , Pyrrolidinones/chemical synthesis , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Drug Design , Drug Evaluation, Preclinical , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , Lactams/chemistry , Lactams/pharmacology , Models, Molecular , Molecular Mimicry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Phenylalanine/analogs & derivatives , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Rhinovirus/drug effects , Structure-Activity Relationship , Valine/analogs & derivatives
18.
Clin Microbiol Rev ; 11(4): 614-27, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9767059

ABSTRACT

Currently, there are a number of approved antiviral agents for use in the treatment of viral infections. However, many instances exist in which the use of a second antiviral agent would be beneficial because it would allow the option of either an alternative or a combination therapeutic approach. Accordingly, virus-encoded proteases have emerged as new targets for antiviral intervention. Molecular studies have indicated that viral proteases play a critical role in the life cycle of many viruses by effecting the cleavage of high-molecular-weight viral polyprotein precursors to yield functional products or by catalyzing the processing of the structural proteins necessary for assembly and morphogenesis of virus particles. This review summarizes some of the important general features of virus-encoded proteases and highlights new advances and/or specific challenges that are associated with the research and development of viral protease inhibitors. Specifically, the viral proteases encoded by the herpesvirus, retrovirus, hepatitis C virus, and human rhinovirus families are discussed.


Subject(s)
Antiviral Agents/therapeutic use , Protease Inhibitors/therapeutic use , Virus Diseases/drug therapy , Gene Products, pol , Humans
19.
Antimicrob Agents Chemother ; 42(10): 2637-44, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9756769

ABSTRACT

Nelfinavir mesylate (formerly AG1343) is a potent and selective inhibitor of human immunodeficiency virus (HIV) protease approved for the treatment of individuals infected with HIV. Nucleotide sequence analysis of protease genes from plasma HIV type 1 (HIV-1) RNA revealed a unique aspartic acid (D)-to-asparagine (N) substitution at residue 30 (D30N) in 25 of 55 patients treated with nelfinavir for a median of 13 weeks. Although the appearance of D30N was occasionally associated with concurrent or sequential emergence of other changes (e.g., at residues 35, 36, 46, 71, 77, and 88), genotypic changes associated with phenotypic resistance to other protease inhibitors were not observed (e.g., at residues 48, 50, 82, and 84) or were only rarely observed (e.g., at residue 90). In phenotypic assays, viral isolates with high-level resistance to nelfinavir remained susceptible to indinavir, saquinavir, ritonavir, and amprenavir (formerly VX-478/141W94). Similar results were observed in phenotypic assays utilizing HIV-1 NL4-3, which contained the D30N substitution alone or in combination with substitutions at other residues (e.g., residues 46, 71, and 88). These data indicate that the initial pathway of resistance to nelfinavir is unique and suggest that individuals failing short courses of nelfinavir-containing regimens may respond to regimens containing other protease inhibitors.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/pharmacology , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Nelfinavir/therapeutic use , Acquired Immunodeficiency Syndrome/virology , Drug Resistance , Genotype , HIV Protease/chemistry , HIV Protease/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Phenotype , Polymorphism, Genetic , RNA, Viral/blood
20.
J Med Chem ; 41(15): 2786-805, 1998 Jul 16.
Article in English | MEDLINE | ID: mdl-9667969

ABSTRACT

The investigation of tripeptide aldehydes as reversible covalent inhibitors of human rhinovirus (HRV) 3C protease (3CP) is reported. Molecular models based on the apo crystal structure of HRV-14 3CP and other trypsin-like serine proteases were constructed to approximate the binding of peptide substrates, generate transition state models of P1-P1' amide cleavage, and propose novel tripeptide aldehydes. Glutaminal derivatives have limitations since they exist predominantly in the cyclic hemiaminal form. Therefore, several isosteric replacements for the P1 carboxamide side chain were designed and incorporated into the tripeptide aldehydes. These compounds were found to be potent inhibitors of purified HRV-14 3CP with Kis ranging from 0.005 to 0.64 microM. Several have low micromolar antiviral activity when tested against HRV-14-infected H1-HeLa cells. The N-acetyl derivative 3 was also shown to be active against HRV serotypes 2, 16, and 89. High-resolution cocrystal structures of HRV-2 3CP, covalently bound to compounds 3, 15, and 16, were solved. These cocrystal structures were analyzed and compared with our original HRV-14 3CP-substrate and inhibitor models.


Subject(s)
Antiviral Agents , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors , Drug Design , Glutamine/chemistry , Oligopeptides , Rhinovirus/drug effects , Viral Proteins , 3C Viral Proteases , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Binding Sites , Cell Line, Transformed , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , HeLa Cells , Humans , Models, Molecular , Molecular Conformation , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Oligopeptides/pharmacology , Protein Conformation , Rhinovirus/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...