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










Publication year range
1.
Mucosal Immunol ; 9(5): 1218-33, 2016 09.
Article in English | MEDLINE | ID: mdl-26813339

ABSTRACT

Intestinal goblet cells are potentially key players in controlling susceptibility to ulcerative colitis (UC). Although impaired mucin (Muc2) production by goblet cells increases microbial stimulation of the colonic mucosa, goblet cells secrete other mediators that may influence or promote UC development. Correspondingly, Muc2-deficient ((-/-)) mice develop spontaneous colitis, concurrent with the dramatic upregulation of the goblet cell mediator, resistin-like molecule-beta (RELM-ß). Testing RELM-ß's role, we generated Muc2(-/-)/Retnlb(-/-) mice, finding that RELM-ß deficiency significantly attenuated colitis development and symptoms compared with Muc2(-/-) mice. RELM-ß expression in Muc2(-/-) mice strongly induced the production/secretion of the antimicrobial lectin RegIIIß, that exerted its microbicidal effect predominantly on Gram-positive Lactobacillus species. Compared with Muc2(-/-)/Retnlb(-/-) mice, this worsened intestinal microbial dysbiosis with a selective loss of colonic Lactobacilli spp. in Muc2(-/-) mice. Orally replenishing Muc2(-/-) mice with murine Lactobacillus spp., but not with a probiotic formulation containing several human Lactobacillus spp. (VSL#3), ameliorated their spontaneous colitis in concert with increased production of short-chain fatty acids. These studies demonstrate that the goblet cell mediator RELM-ß drives colitis in Muc2(-/-) mice by depleting protective commensal microbes. The ability of selective commensal microbial replacement to ameliorate colitis suggests that personalized bacterial therapy may prove beneficial for treatment of UC.


Subject(s)
Colitis, Ulcerative/immunology , Goblet Cells/immunology , Hormones, Ectopic/immunology , Intestinal Mucosa/immunology , Lactobacillus/immunology , Mucin-2/immunology , Animals , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/prevention & control , Colon/immunology , Colon/microbiology , Dysbiosis , Fatty Acids, Volatile/biosynthesis , Gene Expression Regulation , Goblet Cells/microbiology , Hormones, Ectopic/genetics , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Mucin-2/deficiency , Mucin-2/genetics , Pancreatitis-Associated Proteins , Probiotics/administration & dosage , Proteins/genetics , Proteins/immunology , Severity of Illness Index , Signal Transduction , Symbiosis/immunology
2.
Mucosal Immunol ; 9(5): 1278-87, 2016 09.
Article in English | MEDLINE | ID: mdl-26647716

ABSTRACT

Bacterial pneumonia is a leading cause of morbidity and mortality worldwide. Host responses to contain infection and mitigate pathogen-mediated lung inflammation are critical for pneumonia resolution. Aspirin-triggered resolvin D1 (AT-RvD1; 7S,8R,17R-trihydroxy-4Z,9E,11E,13Z,15E,19Z-docosahexaenoic acid) is a lipid mediator (LM) that displays organ-protective actions in sterile lung inflammation, and regulates pathogen-initiated cellular responses. Here, in a self-resolving murine model of Escherichia coli pneumonia, LM metabololipidomics performed on lungs obtained at baseline, 24, and 72 h after infection uncovered temporal regulation of endogenous AT-RvD1 production. Early treatment with exogenous AT-RvD1 (1 h post infection) enhanced clearance of E. coli and Pseudomonas aeruginosa in vivo, and lung macrophage phagocytosis of fluorescent bacterial particles ex vivo. Characterization of macrophage subsets in the alveolar compartment during pneumonia identified efferocytosis by infiltrating macrophages (CD11b(Hi) CD11c(Low)) and exudative macrophages (CD11b(Hi) CD11c(Hi)). AT-RvD1 increased efferocytosis by these cells ex vivo, and accelerated neutrophil clearance during pneumonia in vivo. These anti-bacterial and pro-resolving actions of AT-RvD1 were additive to antibiotic therapy. Taken together, these findings suggest that the pro-resolving actions of AT-RvD1 during pneumonia represent a novel host-directed therapeutic strategy to complement the current antibiotic-centered approach for combatting infections.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Aspirin/pharmacokinetics , Docosahexaenoic Acids/biosynthesis , Escherichia coli Infections/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/immunology , Aspirin/administration & dosage , Aspirin/immunology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Docosahexaenoic Acids/immunology , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Gene Expression , Lipid Metabolism/drug effects , Lipids/analysis , Lipids/immunology , Lipocalin-2/genetics , Lipocalin-2/immunology , Lung/drug effects , Lung/immunology , Lung/microbiology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/pathology , Phagocytosis/drug effects , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/immunology
3.
Diabetologia ; 53(4): 741-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20012858

ABSTRACT

AIMS/HYPOTHESIS: Increased exposure to enteric microbes as a result of intestinal barrier disruption is thought to contribute to the development of several intestinal inflammatory diseases; however, it less clear whether such exposure modulates the development of extra-intestinal inflammatory and autoimmune diseases. The goal of this study was to examine the potential role of pathogenic enteric microbes and intestinal barrier dysfunction in the pathogenesis of type 1 diabetes. METHODS: Using NOD mice, we assessed: (1) intrinsic barrier function in mice at different ages by measuring serum levels of FITC-labelled dextran; and (2) the impact on insulitis development of infection by strains of an enteric bacterial pathogen (Citrobacter rodentium) either capable (wild-type) or incapable (lacking Escherichia coli secreted protein F virulence factor owing to deletion of the gene [DeltaespF]) of causing intestinal epithelial barrier disruption. RESULTS: Here we demonstrate that prediabetic (12-week-old) NOD mice display increased intestinal permeability compared with non-obese diabetes-resistant and C57BL/6 mice. We also found that young (4-week-old) NOD mice infected with wild-type C. rodentium exhibited accelerated development of insulitis in concert with infection-induced barrier disruption. In contrast, insulitis development was not altered in NOD mice infected with the non-barrier-disrupting DeltaespF strain. Moreover, C. rodentium-infected NOD mice demonstrated increased activation and proliferation of pancreatic-draining lymph node T cells, including diabetogenic CD8(+) T cells, compared with uninfected NOD mice. CONCLUSIONS/INTERPRETATION: This is the first demonstration that a loss of intestinal barrier integrity caused by an enteric bacterial pathogen results in the activation of diabetogenic CD8(+) T cells and modulates insulitis.


Subject(s)
Bacterial Infections/complications , Animals , Bacterial Infections/microbiology , CD8-Positive T-Lymphocytes/immunology , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Enterobacteriaceae/immunology , Enterobacteriaceae/pathogenicity , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/pathology , Flow Cytometry , Gene Rearrangement , Hyperinsulinism/microbiology , Inflammation/immunology , Intestines/microbiology , Intestines/physiology , Intestines/physiopathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Prediabetic State/microbiology , Prediabetic State/physiopathology , Receptors, Antigen, T-Cell/genetics , Species Specificity
4.
Cancer Res ; 61(14): 5480-5, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454695

ABSTRACT

Drug resistance is a prevalent problem in the treatment of neoplastic disease, and the effectiveness of many clinically useful drugs is limited by the fact that they are substrates for the efflux pump, P-glycoprotein. Because there is a need for new compounds that are effective in treating drug-resistant tumors, we tested A-204197 (4-[4-acetyl-4,5-dihydro-5-(3,4,5-trimethoxyphenyl)-1,3,4-oxadiazol-2-yl]-N,N-dimethylbenzeneamine), a novel oxadiazoline derivative with antiproliferative properties, on cell lines that were either sensitive or resistant to known microtubule inhibitors. Cell lines that were resistant to paclitaxel, vinblastine, or colchicine were equally sensitive to A-204197 (proliferation IC50s ranging from 36 to 48 nM) despite their expression levels of P-glycoprotein. The effect of A-204197 on cell growth was associated with cell cycle arrest in G2-M, increased phosphorylation of select G2-M checkpoint proteins, and apoptosis. In competition-binding assays, A-204197 competed with [3H]-labeled colchicine for binding to tubulin (K(i) = 0.75 microM); however, it did not compete with [3H]-labeled paclitaxel. A-204197 prevented tubulin polymerization in a dose-dependent manner (IC50 = 4.5 microM) in vitro and depolymerized microtubules in a time-dependent manner in cultured cells. These findings indicate A-204197 is a promising new tubulin-binding compound with antimitotic activity that has potential for treating neoplastic diseases with greater efficacy than currently used antimitotic agents.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Oxadiazoles/pharmacology , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Binding Sites , Cell Cycle/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Colchicine/metabolism , Colchicine/pharmacology , Drug Interactions , Drug Resistance, Multiple , G2 Phase/drug effects , Humans , Microtubules/metabolism , Mitosis/drug effects , Oxadiazoles/metabolism , Paclitaxel/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Time Factors , Tubulin/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Vinblastine/pharmacology
5.
Bioorg Med Chem Lett ; 11(13): 1671-3, 2001 Jul 09.
Article in English | MEDLINE | ID: mdl-11425534

ABSTRACT

The synthesis and biological evaluation of novel sulfonate analogues of E-7010 are reported. Several of the compounds are potent inhibitors of cell proliferation and tubulin polymerization. Importantly, these compounds are also active against P-glycoprotein positive (+) cancer cells, which are resistant to many other antitumor agents.


Subject(s)
Aminophenols/chemistry , Antineoplastic Agents/pharmacology , Mitosis/drug effects , Sulfonamides/chemistry , Sulfonic Acids/pharmacology , Antineoplastic Agents/chemistry , Humans , Sulfonic Acids/chemistry , Tumor Cells, Cultured
7.
J Med Chem ; 44(8): 1192-201, 2001 Apr 12.
Article in English | MEDLINE | ID: mdl-11312919

ABSTRACT

The discovery of (+/-)-(2S,3R,4R)-2-(trifluoroacetamido)methyl-3-amino-1-(N'-ethyl-N'-isopropylcarbamyl)pyrrolidine-4-carboxylic acid (A-192558, 20e) as a potent inhibitor of influenza neuraminidase (NA) is described. Efficient syntheses of two core structures, cis-3-(allyloxycarbonyl)amino-1-(9'-fluorenylmethoxycarbonyl)pyrrolidine-4-carboxylic acid (7) and tert-butyl (+/-)-(2S,3R,4R)-2-aminomethyl-3-bis(tert-butyloxycarbonyl)amino-1-(N'-ethyl-N'-isopropylcarbamyl)pyrrolidine-4-carboxylate (18b), were developed. Starting with these core structures and using available structural information of the NA active site as the guide, analogues were synthesized in both the tri- and tetrasubstituted pyrrolidine series by means of high-throughput parallel synthesis in solid or solution phase for expeditious SAR. These studies accelerated the identification of (+/-)-(2S,3R,4R)-2-(trifluoroacetamido)methyl-3-amino-1-(N-ethyl-N-isopropylcarbamyl)pyrrolidine-4-carboxylate (20e, A-192558) as the most potent NA inhibitor in this series (IC50 = 0.2 microM against NA A and 8 microM against NA B). The X-ray crystallographic structure of A-192558 bound to NA revealed the predicted interaction of the carboxylic group with the positively charged pocket (Arg118, Arg292, Arg371) and interaction of the trifluoroacetamino residue with the hydrophobic pocket (Ile222, Trp178) of the enzyme active site. Surprisingly, the ethyl and isopropyl groups of the urea functionality induced a conformational change of Glu276, turning the Glu276/Glu277 hydrophilic pocket, which normally accommodates the triglycerol side chain of substrate sialic acid, into an induced hydrophobic pocket.


Subject(s)
Antiviral Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Neuraminidase/antagonists & inhibitors , Orthomyxoviridae/enzymology , Pyrrolidines/chemical synthesis , Antiviral Agents/chemistry , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Pyrrolidines/chemistry
8.
Bioorg Med Chem Lett ; 11(7): 871-4, 2001 Apr 09.
Article in English | MEDLINE | ID: mdl-11294380

ABSTRACT

Sulfonate analogues of combretastatin A-4 have been prepared. These compounds compete with colchicine and combretastatin A-4 for the colchicine binding site on tubulin and are potent inhibitors of tubulin polymerization and cell proliferation. Importantly, these compounds also inhibit the proliferation of P-glycoprotein positive (+) cancer cells, which are resistant to many other antitumor agents.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Colchicine/antagonists & inhibitors , Stilbenes/chemistry , Stilbenes/pharmacology , Tubulin Modulators , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Binding Sites/physiology , Binding, Competitive , Cell Division/drug effects , Colchicine/metabolism , Humans , Polymers/metabolism , Tubulin/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
9.
Eur J Cancer ; 36(12): 1565-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10930805

ABSTRACT

Beta(beta)-tubulin isotype variation has recently been implicated in the modulation of resistance to paclitaxel in human lung cancer cells and in primary human ovarian tumour samples. Whether alpha-tubulin is involved in drug resistance has not been reported. We have generated a paclitaxel-resistant cell line (H460/T800) from the sensitive human lung carcinoma parental cell line NCI-H460. The resistant cells are more than 1000-fold resistant to taxol and overexpress P-glycoprotein. Interestingly, H460/T800 cells also overexpress alpha- and beta-tubulin as detected by Western blot analysis. From Northern blot analysis, the mechanism of tubulin overexpression appears to be post-transcriptional. To understand whether alpha-tubulin plays a role in drug resistance, we transfected antisense human kalpha1 cDNA construct into the H460/T800 paclitaxel-resistant cells. The antisense clones displayed a reduced alpha-tubulin expression, and the cells were 45-51% more sensitive to paclitaxel and other known antimitotic drugs, compared with vector transfected controls. Complementary experiments of transfecting the sense kalpha1 cDNA into H460 cells conferred a 1.8- to 3.3-fold increase in the IC(50) of several antimitotic agents. Our study suggests that alpha-tubulin is one of the factors that contributes to drug resistance.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Lung Neoplasms/drug therapy , Paclitaxel/therapeutic use , Tubulin/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Division , DNA, Antisense/genetics , DNA, Complementary/genetics , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Transfection , Tubulin/metabolism , Tumor Cells, Cultured
10.
J Med Chem ; 42(23): 4844-52, 1999 Nov 18.
Article in English | MEDLINE | ID: mdl-10579847

ABSTRACT

Synthesis of a library of secondary benzylic amines based on the Sebti-Hamilton type peptidomimetic farnesyltransferase (FTase) inhibitor FTI-276 (1) led to the identification of 6 as a potent enzyme inhibitor (IC(50) of 8 nM) which lacked the problematic thiol residue which had been a common theme in many of the more important FTase inhibitors reported to date. It has previously been disclosed that addition of o-tolyl substitution to FTase inhibitors of the general description 2 had a salutary effect on both FTase inhibition and inhibition of Ras prenylation in whole cells. Combination of these two observations led us to synthesize 7, a potent FTase inhibitor which displayed an IC(50) of 0.16 nM for in vitro inhibition of FTase and an EC(50) of 190 nM for inhibition of whole cell Ras prenylation. Modification of 7 by classical medicinal chemistry led to the discovery of a series of potent FTase inhibitors, culminating in the identification of 25 which exhibited an IC(50) of 0.20 nM and an EC(50) of 4.4 nM. In vivo tests in a nude mouse xenograft model of human pancreatic cancer (MiaPaCa cells) showed that oral dosing of 25 gave rise to impressive attenuation of the growth of this aggressive tumor cell line.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Methionine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Methionine/chemical synthesis , Methionine/chemistry , Methionine/pharmacology , Mice , Mice, Nude , Molecular Mimicry , Neoplasm Transplantation , Peptides/chemistry , Protein Prenylation , Structure-Activity Relationship , Transplantation, Heterologous , Tumor Cells, Cultured
11.
J Med Chem ; 42(18): 3701-10, 1999 Sep 09.
Article in English | MEDLINE | ID: mdl-10479301

ABSTRACT

The synthesis and evaluation of analogues of previously reported farnesyltransferase inhibitors, pyridyl benzyl ether 3 and pyridylbenzylamine 4, are described. Substitution of 3 at the 5-position of the core aryl ring resulted in inhibitors of equal or less potency against the enzyme and decreased efficacy in a cellular assay against Ras processing by the enzyme. Substitution of 4 at the benzyl nitrogen yielded 26, which showed improved efficacy and potency and yet presented a poor pharmacokinetic profile. Further modification afforded 30, which demonstrated a dramatically improved pharmacokinetic profile. Compounds 26 and 29 demonstrated significant in vivo efficacy in nude mice inoculated with MiaPaCa-2, a human pancreatic tumor-derived cell line.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Receptors, Tumor Necrosis Factor , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzylamines/chemical synthesis , Benzylamines/pharmacology , Enzyme Inhibitors/pharmacology , Ethers/chemical synthesis , Ethers/pharmacology , Humans , Mice , Mice, Nude , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , fas Receptor
12.
Bioorg Med Chem Lett ; 9(8): 1069-74, 1999 Apr 19.
Article in English | MEDLINE | ID: mdl-10328287

ABSTRACT

Potent and orally bioavailable nonthiol-containing inhibitors of protein farnesyltransferase are described. Oral bioavailability was achieved by replacement of the pyridyl ether moiety of 1 with a 2-substituted furan ether to give 4. Potency was regained with 2,5-disubstituted furan ethers while maintaining the bioavailability inherent in 4. p-Chlorophenylfuran ether 24 is 0.7 nM in vitro (FTase) and is 32% bioavailable in the mouse, 30% bioavailable in rats, and 21% bioavailable in dogs.


Subject(s)
Alkyl and Aryl Transferases/administration & dosage , Alkyl and Aryl Transferases/antagonists & inhibitors , Cysteine/chemistry , Alkyl and Aryl Transferases/chemical synthesis , Alkyl and Aryl Transferases/pharmacokinetics , Animals , Biological Availability , Dogs , Mice , Models, Chemical , Rats
13.
Bioorg Med Chem Lett ; 9(5): 703-8, 1999 Mar 08.
Article in English | MEDLINE | ID: mdl-10201832

ABSTRACT

Synthesis and biological evaluation of heteroarenes as reduced cysteine replacements are described. Of the heteroaryl groups examined with respect to FT inhibitor FTI-276 (1), pyridyl was the replacement found to be most effective. Substitutions at C4 of the pyridyl moiety did not affect the in vitro activity. Compound 9a was found to have moderate in vivo bioavailability.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Epoxy Compounds/chemical synthesis , 3T3 Cells , Animals , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacokinetics , Epoxy Compounds/pharmacology , Genes, ras/drug effects , Mice , Rats , Structure-Activity Relationship
14.
Arch Biochem Biophys ; 362(2): 363-75, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9989947

ABSTRACT

The gene coding for the 3C protease from human rhinovirus strain 1B was efficiently expressed in an Escherichia coli strain which also overexpressed the rare argU tRNA. The protease was isolated from inclusion bodies, refolded, and exhibited a kcat/Km = 3280 M-1 s-1 using an internally quenched peptidyl fluorogenic substrate. This continuous fluorogenic assay was used to measure the kinetics of 3C protease inhibition by several conventional peptidyl chloromethylketones as well as a novel series of compounds, the bromomethylketonehydrazides. Compounds containing the bromomethylketonehydrazide backbone and a glutamine-like side chain at the P1 position were potent, time-dependent inhibitors of rhinovirus 3C protease with kinact/Kinact values as high as 23,400 M-1 s-1. The inhibitory activity of compounds containing modified P1 side chains suggests that the interactions between the P1 carboxamide group and the 3C protease contributes at least 30-fold to the kinact/Kinact rate constants for bromomethylketonehydrazide inhibition of 3C protease. Electrospray ionization mass spectrometry measurements of the molecular weights of native and inhibited 3C protease have established an inhibitory mechanism involving formation of a covalent adduct between the enzyme and the inhibitor with the loss of a bromide ion from the bromomethylketonehydrazide. Tryptic digestion of bromomethylketonehydrazide-inhibited 3C protease established adduct formation to a peptide corresponding to residues 145-154, a region which contains the active site cysteine-148 residue. The bromomethylketonehydrazides were fairly weak inhibitors of chymotrypsin, human elastase, and cathepsin B and several of these compounds also showed evidence for inhibition of human rhinovirus 1B replication in cell culture.


Subject(s)
Cysteine Endopeptidases/metabolism , Hydrazines/pharmacology , Protease Inhibitors/pharmacology , Rhinovirus/enzymology , Viral Proteins , 3C Viral Proteases , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Cathepsin B/antagonists & inhibitors , Chymotrypsin/antagonists & inhibitors , Cysteine/metabolism , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/isolation & purification , Escherichia coli/genetics , Fluorescent Dyes , Glutamine/analogs & derivatives , Glutamine/metabolism , Humans , Hydrazines/chemical synthesis , Kinetics , Models, Chemical , Molecular Weight , Pancreatic Elastase/antagonists & inhibitors , Protease Inhibitors/chemistry , RNA, Transfer/biosynthesis , RNA, Transfer/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Solubility , Substrate Specificity , Trypsin/metabolism , Virus Replication/drug effects
15.
Drug Metab Dispos ; 27(1): 86-91, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9884314

ABSTRACT

HIV protease inhibitor ABT-378 (ABT-378) was metabolized very extensively and rapidly by liver microsomes from mouse, rat, dog, monkey, and humans. The rates of NADPH-dependent metabolism of ABT-378 ranged from 2.39 to 9.80 nmol.mg microsomal protein-1.min-1, with monkey liver microsomes exhibiting the highest rates of metabolism. ABT-378 was metabolized to 12 metabolites (M-1 to M-12), which were characterized by mass and NMR spectroscopy. The metabolite profile of ABT-378 in liver microsomes from all five species was similar, except that the mouse liver microsomes did not form M-9, a minor secondary metabolite. The predominant site of metabolism was the cyclic urea moiety of ABT-378. In all five species, the major metabolites were M-1 (4-oxo-ABT-378) and M-3 and M-4 (4-hydroxy-ABT-378). Metabolite M-2 (6-hydroxy-ABT-378) was formed by rodents at a faster rate than by dog, monkey, and human liver microsomes. Metabolites M-5 to M-8 were identified as monohydroxylated derivatives of ABT-378. Metabolites M-9 and M-10 were identified as hydroxylated products of M-1. Metabolites M-11 and M-12 were identified as dihydroxylated derivatives of ABT-378. The metabolite profile in human hepatocytes and liver slices was similar to that of human liver microsomes. The results of the current study indicate that ABT-378 is highly susceptible to oxidative metabolism in vitro, and possibly in vivo, in humans.


Subject(s)
Anti-HIV Agents/metabolism , HIV Protease Inhibitors/metabolism , HIV-1/enzymology , Liver/metabolism , Pyrimidinones/metabolism , Animals , Chromatography, High Pressure Liquid , Dogs , Female , Gas Chromatography-Mass Spectrometry , Humans , Liver/cytology , Lopinavir , Macaca fascicularis , Magnetic Resonance Spectroscopy , Male , Mice , Microsomes, Liver/metabolism , Middle Aged , Rats , Rats, Sprague-Dawley
16.
Antimicrob Agents Chemother ; 42(12): 3218-24, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9835517

ABSTRACT

The valine at position 82 (Val 82) in the active site of the human immunodeficiency virus (HIV) protease mutates in response to therapy with the protease inhibitor ritonavir. By using the X-ray crystal structure of the complex of HIV protease and ritonavir, the potent protease inhibitor ABT-378, which has a diminished interaction with Val 82, was designed. ABT-378 potently inhibited wild-type and mutant HIV protease (Ki = 1.3 to 3.6 pM), blocked the replication of laboratory and clinical strains of HIV type 1 (50% effective concentration [EC50], 0.006 to 0.017 microM), and maintained high potency against mutant HIV selected by ritonavir in vivo (EC50, 50-fold after 8 h. In healthy human volunteers, coadministration of a single 400-mg dose of ABT-378 with 50 mg of ritonavir enhanced the area under the concentration curve of ABT-378 in plasma by 77-fold over that observed after dosing with ABT-378 alone, and mean concentrations of ABT-378 exceeded the EC50 for >24 h. These results demonstrate the potential utility of ABT-378 as a therapeutic intervention against AIDS.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Protease Inhibitors/pharmacology , Pyrimidinones/pharmacology , Animals , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacokinetics , Area Under Curve , Crystallography, X-Ray , Dogs , Drug Interactions , Female , HIV Protease/chemistry , HIV Protease Inhibitors/metabolism , HIV Protease Inhibitors/pharmacokinetics , HIV-1/drug effects , Humans , In Vitro Techniques , Lopinavir , Macaca fascicularis , Male , Microsomes, Liver/metabolism , Models, Molecular , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Rats , Rats, Sprague-Dawley , Ritonavir/chemistry , Ritonavir/pharmacology
17.
Biochemistry ; 37(49): 17192-8, 1998 Dec 08.
Article in English | MEDLINE | ID: mdl-9860832

ABSTRACT

The binding of several different active site mutants of Bacillus circulans cyclodextrin glycosyltransferase to the inhibitor acarbose has been investigated through measurement of Ki values. The mutations represent several key amino acid positions, most of which are believed to play important roles in governing the product specificity of cyclodextrin glycosyltransferase. Michaelis-Menten parameters for the substrates alpha-maltotriosyl fluoride (alphaG3F) and alpha-glucosyl fluoride (alphaGF) with each mutant have been determined by following the enzyme-catalyzed release of fluoride with an ion-selective fluoride electrode. In both cases, reasonable correlations are observed in logarithmic plots relating the Ki value for acarbose with each mutant and both kcat/Km and Km for the hydrolysis of either substrate by the corresponding mutants. This indicates that acarbose, as an inhibitor, is mimicking aspects of both the ground state and the transition state. A better correlation is observed for alphaGF (r = 0.98) than alphaG3F (r = 0.90), which can be explained in terms of the modes of binding of these substrates and acarbose. Re-refinement of the previously determined crystal structure of wild-type CGTase complexed with acarbose [Strokopytov, B., Penninga, D., Rozeboom, H. J., Kalk, K. H., Dijhuizen, L., and Dijkstra, B. W. (1995) Biochemistry 34, 2234-2240] reveals a binding mode consistent with the transition state analogue character of this inhibitor.


Subject(s)
Cyclodextrins/metabolism , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Trisaccharides/pharmacology , Acarbose , Bacillus/enzymology , Bacillus/genetics , Binding Sites/genetics , Crystallography, X-Ray , Fluorides/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Substrate Specificity/genetics , Trisaccharides/metabolism
18.
Virology ; 250(2): 255-62, 1998 Oct 25.
Article in English | MEDLINE | ID: mdl-9792836

ABSTRACT

The potency of therapeutic regimens containing human immunodeficiency virus (HIV) protease inhibitors is related to the ability to maintain concentrations of drug in the plasma of patients that are sufficient for blocking viral replication. The estimation of concentrations required for in vivo activity using in vitro assays is complicated by the fact that extensive binding of many protease inhibitors to serum proteins attenuates their antiviral potency. To provide insight into the relative in vivo potency of current protease inhibitors, we assayed their in vitro activity against wild-type and mutant HIV in the presence of human serum (HS). Using this assay, ABT-378, a new protease inhibitor with trough levels in humans far in excess of the EC50 in the presence of 50% HS, was identified. The antiviral activity of ABT-378 was only modestly attenuated by HS, in contrast to ritonavir, saquinavir, and nelfinavir. Examination of the effect of individual serum components suggested that the activity of ABT-378 is affected predominantly by binding to alpha1-acid glycoprotein (AGP) while the activity of ritonavir is modulated by both AGP and albumin. The method described here may provide insight into the in vivo potency of protease inhibitors and be useful for the preclinical evaluation and selection of new protease inhibitors for clinical studies.


Subject(s)
Blood Proteins/metabolism , HIV Protease Inhibitors/metabolism , HIV-1 , Mutation , Pyrimidinones/metabolism , Cell Line, Transformed , HIV Protease Inhibitors/pharmacology , HIV-1/genetics , Humans , Lopinavir , Ritonavir/metabolism
19.
J Virol ; 72(9): 7532-41, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9696850

ABSTRACT

ABT-378, a new human immunodeficiency virus type 1 (HIV-1) protease inhibitor which is significantly more active than ritonavir in cell culture, is currently under investigation for the treatment of AIDS. Development of viral resistance to ABT-378 in vitro was studied by serial passage of HIV-1 (pNL4-3) in MT-4 cells. Selection of viral variants with increasing concentrations of ABT-378 revealed a sequential appearance of mutations in the protease gene: I84V-L10F-M46I-T91S-V32I-I47V. Further selection at a 3.0 microM inhibitor concentration resulted in an additional change at residue 47 (V47A), as well as reversion at residue 32 back to the wild-type sequence. The 50% effective concentration of ABT-378 against passaged virus containing these additional changes was 338-fold higher than that against wild-type virus. In addition to changes in the protease gene, sequence analysis of passaged virus revealed mutations in the p1/p6 (P1' residue Leu to Phe) and p7/p1 (P2 residue Ala to Val) gag proteolytic processing sites. The p1/p6 mutation appeared in several clones derived from early passages and was present in all clones obtained from passage P11 (0.42 microM ABT-378) onward. The p7/p1 mutation appeared very late during the selection process and was strongly associated with the emergence of the additional change at residue 47 (V47A) and the reversion at residue 32 back to the wild-type sequence. Furthermore, this p7/p1 mutation was present in all clones obtained from passage P17 (3.0 microM ABT-378) onward and always occurred in conjunction with the p1/p6 mutation. Full-length molecular clones containing protease mutations observed very late during the selection process were constructed and found to be viable only in the presence of both the p7/p1 and p1/p6 cleavage-site mutations. This suggests that mutation of these gag proteolytic cleavage sites is required for the growth of highly resistant HIV-1 selected by ABT-378 and supports recent work demonstrating that mutations in the p7/p1/p6 region play an important role in conferring resistance to protease inhibitors (L. Doyon et al., J. Virol. 70:3763-3769, 1996; Y. M. Zhang et al., J. Virol. 71:6662-6670, 1997).


Subject(s)
Anti-HIV Agents/pharmacology , Genetic Variation , HIV Protease/drug effects , HIV-1/drug effects , Pyrimidinones/pharmacology , Amino Acid Sequence , Animals , Anti-HIV Agents/chemistry , Binding Sites , COS Cells , Cell Line, Transformed , Drug Resistance, Microbial , HIV Protease/genetics , HIV Protease/metabolism , HIV-1/enzymology , HIV-1/genetics , Humans , Lopinavir , Molecular Sequence Data , Molecular Structure , Mutation , Pyrimidinones/chemistry , Ritonavir/pharmacology , Saquinavir/pharmacology , Tumor Cells, Cultured
20.
J Med Chem ; 41(4): 602-17, 1998 Feb 12.
Article in English | MEDLINE | ID: mdl-9484509

ABSTRACT

The structure-activity studies leading to the potent and clinically efficacious HIV protease inhibitor ritonavir are described. Beginning with the moderately potent and orally bioavailable inhibitor A-80987, systematic investigation of peripheral (P3 and P2') heterocyclic groups designed to decrease the rate of hepatic metabolism provided analogues with improved pharmacokinetic properties after oral dosing in rats. Replacement of pyridyl groups with thiazoles provided increased chemical stability toward oxidation while maintaining sufficient aqueous solubility for oral absorption. Optimization of hydrophobic interactions with the HIV protease active site produced ritonavir, with excellent in vitro potency (EC50 = 0.02 microM) and high and sustained plasma concentrations after oral administration in four species. Details of the discovery and preclinical development of ritonavir are described.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease/metabolism , Ritonavir/analogs & derivatives , Ritonavir/chemistry , Administration, Oral , Animals , Biological Availability , Crystallography, X-Ray , HIV Protease/chemistry , HIV Protease Inhibitors/pharmacokinetics , HIV Protease Inhibitors/pharmacology , Metabolic Clearance Rate , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Conformation , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Ritonavir/pharmacokinetics , Ritonavir/pharmacology , Solubility , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...