Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioorg Med Chem Lett ; 18(15): 4360-3, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18606544

ABSTRACT

We report the synthesis and in vitro activity of a series of novel pyrrolidinyl pyridones and pyrazinones as potent inhibitors of prolyl oligopeptidase (POP). Within this series, compound 39 was co-crystallized within the catalytic site of a human chimeric POP protein which provided a more detailed understanding of how these inhibitors interacted with the key residues within the catalytic pocket.


Subject(s)
Pyridones/chemical synthesis , Pyridones/pharmacology , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Serine Endopeptidases/drug effects , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacology , Brain/drug effects , Brain/metabolism , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Drug Design , Humans , Molecular Conformation , Molecular Structure , Prolyl Oligopeptidases , Pyridones/blood , Pyridones/chemistry , Pyrrolidines/blood , Pyrrolidines/chemistry , Serine Proteinase Inhibitors/blood , Serine Proteinase Inhibitors/chemistry
2.
Bioorg Med Chem Lett ; 16(7): 1788-94, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16458505

ABSTRACT

A novel series of P1 modified HIV protease inhibitors was synthesized and evaluated for in vitro antiviral activity against wild-type virus and protease inhibitor-resistant viruses. Optimization of the P1 moiety resulted in compounds with femtomolar enzyme activities and cellular antiviral activities in the low nanomolar range culminating in the identification of clinical candidate GW0385.


Subject(s)
HIV Protease Inhibitors/pharmacology , Sulfonamides/pharmacology , HIV Protease Inhibitors/chemistry , Molecular Structure , Sulfonamides/chemistry
3.
Biochemistry ; 43(45): 14500-7, 2004 Nov 16.
Article in English | MEDLINE | ID: mdl-15533054

ABSTRACT

The arylsulfonamide derivatives described herein were such potent inhibitors of human immunodeficiency virus type 1 (HIV-1) protease (enzyme, E) that values for the inhibition constants (K(i)) could not be determined by conventional steady-state kinetic techniques (i.e., the minimal enzyme concentration usable for the activity assay was much greater than the value of the dissociation constant). Consequently, two alternative methods were developed for estimation of K(i) values. The first method employed kinetic determinations of values for k(1) and k(-1), from which K(i) was determined (k(-1)/k(1)). The second method was a competitive displacement assay used to determine binding affinities of other inhibitors relative to that of GW0385. In these assays, the inhibitor of unknown affinity was used to displace [(3)H]GW0385 from E.[(3)H]GW0385. From the concentration of E.[(3)H]GW0385 at equilibrium, the concentrations of enzyme-bound and free inhibitors were calculated, and the ratio of the K(i) value of the unknown to that of GW0385 was determined (K(i,unknown)/K(i,GW0385)). The values of k(1) were calculated from data in which changes in the intrinsic protein fluorescence of the enzyme associated with inhibitor binding were directly or indirectly monitored. In the case of saquinavir, the fluorescence changes associated with complex formation were large enough to monitor directly. The value of k(1) for saquinavir was 62 +/- 2 microM(-1) s(-1). In the case of GW0385, the fluorescence changes associated with complex formation were too small to monitor directly. Consequently, the value of k(1) was estimated from a competition experiment in which the effect of GW0385 on the binding of E to saquinavir was determined. The value of k(1) for GW0385 was estimated from these experiments to be 137 +/- 4 microM(-1) s(-1). Because E.[(3)H]GW0385 was stable in the standard buffer at room temperature for greater than 33 days, the value of the first-order rate constant for dissociation of E.[(3)H]GW0385 (k(-1)) could be estimated from the time-course for exchange of E.[(3)H]GW0385 with excess unlabeled GW0385. The value of k(-1) calculated from these data was (2.1 +/- 0.1) x10(-6) s(-1) (t(1/2) = 91 h). The K(i) value of wild-type HIV-1 protease for GW0385, calculated from these values for k(1) and k(-1), was 15 +/- 1 fM. Three multidrug resistant enzymes had K(i) values for GW0385 that were less than 5 pM.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV-1/enzymology , Sulfonamides/chemistry , Amino Acid Substitution/genetics , Binding, Competitive/genetics , Carbamates , Chromatography, Affinity , Chromatography, Gel , Dextrans/chemistry , Dimerization , Furans , HIV Protease/chemistry , HIV Protease/genetics , HIV Protease/metabolism , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , Hydrolysis , Kinetics , Protein Binding/genetics , Saquinavir/chemistry , Saquinavir/metabolism , Saquinavir/pharmacology , Spectrometry, Fluorescence , Substrate Specificity , Sulfonamides/metabolism , Sulfonamides/pharmacology
4.
Bioorg Med Chem Lett ; 14(4): 959-63, 2004 Feb 23.
Article in English | MEDLINE | ID: mdl-15013001

ABSTRACT

A novel series of P1' chain-extended arylsufonamides was synthesized and evaluated for wild-type HIV protease inhibitory activity and in vitro antiviral activity against wild type virus and two protease inhibitor-resistant mutant viruses. All of the compounds showed dramatic increases in enzyme activity as compared to the currently marketed HIV protease inhibitors amprenavir, indinavir, and nelfinavir. In addition, significant improvements in antiviral potencies against wild type and the two mutant viruses were also realized.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Multiple, Viral/genetics , HIV Protease Inhibitors/pharmacology , HIV Protease/drug effects , HIV/genetics , Sulfonamides/pharmacology , Animals , Anti-HIV Agents/chemical synthesis , Cell Line , Drug Resistance, Multiple, Viral/drug effects , HIV/drug effects , HIV Protease/metabolism , HIV Protease Inhibitors/chemical synthesis , Humans , Molecular Structure , Mutation , Rats , Structure-Activity Relationship , Sulfonamides/chemical synthesis
5.
J Virol ; 76(15): 7398-406, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12097552

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Gag protease cleavage sites (CS) undergo sequence changes during the development of resistance to several protease inhibitors (PIs). We have analyzed the association of sequence variation at the p7/p1 and p1/p6 CS in conjunction with amprenavir (APV)-specific protease mutations following PI combination therapy with APV. Querying a central resistance data repository resulted in the detection of significant associations (P < 0.001) between the presence of APV protease signature mutations and Gag L449F (p1/p6 LP1'F) and P453L (p1/p6 PP5'L) CS changes. In population-based sequence analyses the I50V mutant was invariably linked to either L449F or P453L. Clonal analysis revealed that both CS mutations were never present in the same genome. Sequential plasma samples from one patient revealed a transition from I50V M46L P453L viruses at early time points to I50V M46I L449F viruses in later samples. Various combinations of the protease and Gag mutations were introduced into the HXB2 laboratory strain of HIV-1. In both single- and multiple-cycle assay systems and in the context of I50V, the L449F and P453L changes consistently increased the 50% inhibitory concentration of APV, while the CS changes alone had no measurable effect on inhibitor sensitivity. The decreased in vitro fitness of the I50V mutant was only partially improved by addition of either CS change (I50V M46I L449F mutant replicative capacity approximately 16% of that of wild-type virus). Western blot analysis of Pr55 Gag precursor cleavage products from infected-cell cultures indicated accumulation of uncleaved Gag p1-p6 in all I50V viruses without coexisting CS changes. Purified I50V protease catalyzed cleavage of decapeptides incorporating the L449F or P453L change 10-fold and 22-fold more efficiently than cleavage of the wild-type substrate, respectively. HIV-1 protease CS changes are selected during PI therapy and can have effects on both viral fitness and phenotypic resistance to PIs.


Subject(s)
Drug Resistance, Viral , Gene Products, gag/genetics , HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , HIV-1/drug effects , Sulfonamides/pharmacology , Binding Sites , Carbamates , Drug Therapy, Combination , Furans , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HIV Protease/metabolism , HIV Protease Inhibitors/therapeutic use , HIV-1/enzymology , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Mutation , Substrate Specificity , Sulfonamides/therapeutic use , Virus Replication , gag Gene Products, Human Immunodeficiency Virus
6.
Biochemistry ; 41(4): 1302-7, 2002 Jan 29.
Article in English | MEDLINE | ID: mdl-11802730

ABSTRACT

The steady-state and pre-steady-state kinetic parameters for the interaction of E with the fluorogenic substrate 2-aminobenzoyl-Thr-Ile-Nle-Phe(p-NO(2))-Gln-Arg-NH(2) were determined in 1.25 M NaCl, 0.1 M MES-TRIS at pH 6.0 at 25 degrees C. At low concentrations of enzyme, the values of the K(m) and k(cat) calculated from steady-state data were 2.1 microM and 7.4 s(-1), respectively. At high concentrations of enzyme, the time-courses of fluorescence enhancement associated with catalysis were very dependent on the excitation wavelength used to monitor the reaction. Because the absorbance spectrum of the substrate overlapped the fluorescence emission spectrum of the enzyme, these abnormalities were attributed to fluorescence energy transfer between the enzyme and the substrate in an enzyme-substrate intermediate. The kinetic data collected with lambda(ex) = 280 nm and lambda(em) > 435 nm were analyzed according to the following mechanism in which EX was the species with enhanced fluorescence relative to substrate or products: [formula see text]. The values of the kinetic parameters with (1)H(2)O as the solvent were K = 13 microM, k(2) = 150 s(-1), k(-2) = 25 s(-1), and k(3) = 11 s(-1). The values of the kinetic parameters with (2)H(2)O as the solvent were K = 13 microM, k(2) = 210 s(-1), k(-2) = 12 s(-1), and k(3) = 4.4 s(-1). These values yielded solvent isotope effects of 2 on k(cat) and 0.9 on k(cat)/K(m). From analysis of the complete time-course of the fluorescence change (lambda(ex) = 280 nm and lambda(em) > 435 nm) during the course of substrate hydrolysis, the intermediate EX was determined to be 6.3-fold more fluorescent than the product, which, in turn, was 4.5-fold more fluorescent than ES or S. Rapid quench experiments with 2 N HCl as the quenching reagent confirmed that EX was a complex between enzyme and substrate. Consequently, the small burst in fluorescence observed when monitoring with lambda(ex) = 340 nm (0.3 product equiv per enzyme equivalent) was attributed to the fluorescence change upon transfer of substrate from an aqueous environment to a nonaqueous environment in the enzyme. These results were consistent with carbon-nitrogen bond cleavage being the major contributor to k(cat).


Subject(s)
HIV Protease/metabolism , Catalysis , HIV Protease/chemistry , Hydrolysis , Kinetics , Spectrometry, Fluorescence , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL