Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
AJNR Am J Neuroradiol ; 36(12): 2354-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26381560

ABSTRACT

BACKGROUND AND PURPOSE: Current nonhuman primate stroke models are limited by either stroke variability or survivability. A new nonhuman primate stroke model was developed by using endovascular trapping techniques to limit collateral vessels with serial MR imaging and neurologic assessments. MATERIALS AND METHODS: Eight adult rhesus monkeys (female, 7-13 years of age) underwent MR imaging and Spetzler neurologic assessment followed by endovascular stroke induction consisting of superselective endovascular placement of surgical silk sutures into the right MCA by using a trapping technique. Two initial subjects were euthanized immediately following postocclusion MR imaging. The subsequent 6 subjects recovered and underwent follow-up MR imaging and Spetzler neurologic assessments at 48 hours, with 4 being followed to 96 hours. Stroke infarct volumes were measured, and the longitudinal Spetzler clinical neurologic scores were assessed. The brain tissues were harvested and prepared with hematoxylin-eosin staining. RESULTS: Focal permanent cerebral ischemia was induced in the targeted right MCA territory in all subjects. The volumes of the ischemic lesions at 6, 48, and 96 hours were 3.18 ± 1.007 mL (standard error of the mean) (n = 8), 6.70 ± 1.666 mL (standard error of the mean) (n = 6), and 7.23 ± 1.371 mL (standard error of the mean) (n = 4). For the survival animals, the immediate postsurgical Spetzler grading score improved from 60.7 at 24 hours to 68.7 at 48 hours. CONCLUSIONS: We report a trapping modification to an established endovascular suture stroke model that yielded reproducible ischemia and clinically quantifiable neurologic deficits with no strokes in nontarget areas. This technique may be useful in evaluating translational stroke and penumbral imaging research in addition to preclinical testing of neuroprotective therapies.


Subject(s)
Disease Models, Animal , Infarction, Middle Cerebral Artery/pathology , Animals , Endovascular Procedures , Female , Macaca mulatta , Magnetic Resonance Imaging
2.
J Virol ; 75(16): 7462-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11462018

ABSTRACT

The association of genotypic changes in human immunodeficiency virus (HIV) protease with reduced in vitro susceptibility to the new protease inhibitor lopinavir (previously ABT-378) was explored using a panel of viral isolates from subjects failing therapy with other protease inhibitors. Two statistical tests showed that specific mutations at 11 amino acid positions in protease (L10F/I/R/V, K20M/R, L24I, M46I/L, F53L, I54L/T/V, L63P, A71I/L/T/V, V82A/F/T, I84V, and L90M) were associated with reduced susceptibility. Mutations at positions 82, 54, 10, 63, 71, and 84 were most closely associated with relatively modest (4- and 10-fold) changes in phenotype, while the K20M/R and F53L mutations, in conjunction with multiple other mutations, were associated with >20- and >40-fold-reduced susceptibility, respectively. The median 50% inhibitory concentrations (IC(50)) of lopinavir against isolates with 0 to 3, 4 or 5, 6 or 7, and 8 to 10 of the above 11 mutations were 0.8-, 2.7-, 13.5-, and 44.0-fold higher, respectively, than the IC(50) against wild-type HIV. On average, the IC(50) of lopinavir increased by 1.74-fold per mutation in isolates containing three or more mutations. Each of the 16 viruses that displayed a >20-fold change in susceptibility contained mutations at residues 10, 54, 63, and 82 and/or 84, along with a median of three mutations at residues 20, 24, 46, 53, 71, and 90. The number of protease mutations from the 11 identified in these analyses (the lopinavir mutation score) may be useful for the interpretation of HIV genotypic resistance testing with respect to lopinavir-ritonavir (Kaletra) regimens and may provide insight into the genetic barrier to resistance to lopinavir-ritonavir in both antiretroviral therapy-naive and protease inhibitor-experienced patients.


Subject(s)
HIV Infections/virology , HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , HIV-1/drug effects , HIV-1/genetics , Pyrimidinones/pharmacology , Drug Resistance/genetics , Genome, Viral , HIV Infections/drug therapy , Humans , Lopinavir , Pyrimidinones/therapeutic use
4.
AIDS ; 13(2): 213-24, 1999 Feb 04.
Article in English | MEDLINE | ID: mdl-10202827

ABSTRACT

OBJECTIVE: To evaluate the safety and antiretroviral activity of ritonavir (Norvir) and saquinavir (Invirase) combination therapy in patients with HIV infection. DESIGN: A multicenter, randomized, open-label clinical trial. SETTING: Seven HIV research units in the USA and Canada. PATIENTS: A group of 141 adults with HIV infection, CD4 T lymphocyte counts of 100-500 x 10(6) cells/l, whether treated previously or not with reverse transcriptase inhibitor therapy, but without previous HIV protease inhibitor drug therapy. INTERVENTIONS: After discontinuation of prior therapy for 2 weeks, group I patients were randomized to receive either combination (A) ritonavir 400 mg and saquinavir 400 mg twice daily or (B) ritonavir 600 mg and saquinavir 400 mg twice daily. After an initial safety assessment of group I patients, group II patients were randomized to receive either (C) ritonavir 400 mg and saquinavir 400 mg three times daily or (D) ritonavir 600 mg and saquinavir 600 mg twice daily. Investigators were allowed to add up to two reverse transcriptase inhibitors (including at least one with which the patient had not been previously treated) to a patient's regimen after week 12 for failure to achieve or maintain an HIV RNA level < or = 200 copies/ml documented on two consecutive occasions. MEASUREMENTS: Plasma HIV RNA levels and CD4+ T-lymphocyte counts were measured at baseline, every 2 weeks for 2 months, and monthly thereafter. Safety was assessed through the reporting of adverse events, physical examinations, and the monitoring of routine laboratory tests. RESULTS: The 48 weeks of study treatment was completed by 75% (106/141) of the patients. Over 80% of the patients on treatment at week 48 had an HIV RNA level < or = 200 copies/ml. In addition, intent-to-treat and on-treatment analyses revealed comparable results. Suppression of plasma HIV RNA levels was similar for all treatment arms (mean areas under the curve minus baseline through 48 weeks were-1.9, -2.0, -1.6, -1.8 log10 copies/ml in ritonavir-saquinavir 400-400 mg twice daily, 600-400 mg twice daily, 400-400 mg three times daily, and 600-600 mg twice daily, respectively). Median CD4 T-lymphocyte count rose by 128 x 10(6) cells/l from baseline, with an interquartile range (IQR) of 82-221 x 10(6) cells/l. The most common adverse events were diarrhea, circumoral paresthesia, asthenia, and nausea. Reversible elevation of serum transaminases (> 5 x upper limit of normal) occurred in 10% (14/141) of the patients enrolled in this study and was associated with baseline abnormalities in liver function tests, baseline hepatitis B surface antigen positivity, or hepatitis C antibody positivity (relative risk, 5.0; 95% confidence interval 1.5-16.9). Most moderate or severe elevations in liver function tests occurred in patients treated with ritonavir-saquinavir 600-600 mg twice daily. CONCLUSIONS: Ritonavir 400 mg combined with saquinavir 400 mg twice daily with the selective addition of reverse transcriptase inhibitors was the best-tolerated regimen of four dose-ranging regimens and was equally as active as the higher dose combinations in HIV-positive patients without previous protease inhibitor treatment.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Protease Inhibitors/therapeutic use , HIV-1 , Ritonavir/therapeutic use , Saquinavir/therapeutic use , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Consumer Product Safety , Drug Therapy, Combination , Female , HIV Infections/cerebrospinal fluid , HIV Infections/mortality , HIV Infections/virology , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/pharmacokinetics , HIV-1/genetics , Humans , Male , Reverse Transcriptase Inhibitors/therapeutic use , Ritonavir/adverse effects , Ritonavir/pharmacokinetics , Saquinavir/adverse effects , Saquinavir/pharmacokinetics
5.
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
6.
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
7.
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
9.
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
10.
AIDS ; 12(5): F9-14, 1998 Mar 26.
Article in English | MEDLINE | ID: mdl-9543434

ABSTRACT

OBJECTIVE: To determine markers that are associated with the durability of virologic response to therapy with HIV protease inhibitors in HIV-infected individuals. DESIGN: This study encompassed two retrospective analyses of the duration of virologic response to protease inhibitor therapy. The first analysis included 29 patients receiving either monotherapy or combination therapy with the protease inhibitor ritonavir whose plasma HIV RNA levels rebounded from the point of greatest decline with mutations associated with resistance to ritonavir. The second analysis included a cohort of 102 patients who initially responded to randomized treatment with either monotherapy with ritonavir or combination therapy with ritonavir and zidovudine. METHODS: Durability of response was defined as the time from the initiation of therapy to the point at which plasma HIV RNA displayed a sustained increase of at least 0.6 log10 copies/ml from the nadir value. In the first analysis, durability of response was analyzed with respect to baseline HIV RNA, HIV RNA at the nadir, and the drop in HIV RNA from baseline to the nadir. In the second analysis, time to rebound was examined using Kaplan-Meier analysis, stratifying by either baseline HIV RNA or HIV RNA at the nadir. RESULTS: In both analyses, the durability of response was not highly associated with either baseline RNA or the magnitude of RNA decline from baseline. Instead, a strong relationship was observed between the durability of response and the nadir plasma HIV-1 RNA value (P < 0.01). The nadir in viral load was generally reached after 12 weeks of randomized therapy. CONCLUSIONS: Viral RNA determinations at intermediate timepoints may be prognostic of impending virologic failure of protease inhibitor therapy. Therapeutic strategies that allow intensification of initial antiretroviral regimens in the subset of patients with incomplete virological response before the emergence of high level resistance should be investigated.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/therapeutic use , HIV-1/drug effects , Predictive Value of Tests , RNA, Viral/blood , Drug Therapy, Combination , HIV-1/genetics , HIV-1/physiology , Humans , Mutation , Retrospective Studies , Ritonavir/therapeutic use , Treatment Outcome , Viral Load , Zidovudine/therapeutic use
11.
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
12.
Bioorg Med Chem Lett ; 8(24): 3531-6, 1998 Dec 15.
Article in English | MEDLINE | ID: mdl-9934466

ABSTRACT

The 2-isopropyl thiazolyl group is a highly optimized P3 ligand for C2 symmetry-based HIV protease inhibitors, as exemplified in the drug ritonavir. Here we report that incorporation of this P3 ligand into a piperazine hydroxyethylamine series also yielded novel, highly potent inhibitors. In tissue culture assays, the presence of human serum was less deleterious to the activity of these inhibitors than to that of ritonavir. Furthermore, potent activity against ritonavir resistant HIV was observed.


Subject(s)
HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacology , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Blood , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme Inhibitors , HIV/drug effects , HIV Protease Inhibitors/chemical synthesis , Humans , Ligands , Microbial Sensitivity Tests , Microsomes, Liver/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Piperazines/chemical synthesis , Piperazines/chemistry , Piperazines/pharmacology
13.
J Med Chem ; 40(20): 3144-50, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9379433

ABSTRACT

The E2 protein is required for the replication of human papillomaviruses (HPVs), which are responsible for anogenital warts and cervical carcinomas. Using an NMR-based screen, we tested compounds for binding to the DNA-binding domain of the HPV-E2 protein. Three classes of compounds were identified which bound to two distinct sites on the protein. Biphenyl and biphenyl ether compounds containing a carboxylic acid bind to a site near the DNA recognition helix and inhibit the binding of E2 to DNA. Benzophenone-containing compounds which lack a carboxylic acid group bind to the beta-barrel formed by the dimer interface and exhibit negligible effects on the binding of E2 to DNA. Structure-activity relationships from the biphenyl and biphenyl ether compounds were combined to produce a compound [5-(3'-(3",5"-dichlorophenoxy)-phenyl)-2,4-pentadienoic acid] with an IC50 value of approximately 10 microM. This compound represents a useful lead for the development of antiviral agents that interfere with HPV replication and further illustrates the usefulness of the SAR by NMR method in the drug discovery process.


Subject(s)
Antiviral Agents/chemistry , DNA-Binding Proteins/antagonists & inhibitors , DNA/metabolism , Drug Design , Repressor Proteins/metabolism , Trans-Activators/metabolism , Viral Proteins/antagonists & inhibitors , Antiviral Agents/pharmacology , Binding Sites , Biphenyl Compounds/pharmacology , Bovine papillomavirus 1 , Crystallography, X-Ray , DNA-Binding Proteins/metabolism , Humans , Ligands , Magnetic Resonance Spectroscopy , Models, Chemical , Models, Molecular , Papillomaviridae , Protein Conformation , Structure-Activity Relationship , Viral Proteins/metabolism
14.
Drug Metab Dispos ; 25(4): 489-501, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9107549

ABSTRACT

The metabolism and disposition of [14C]ritonavir (ABT-538, NOR-VIR), a potent, orally active HIV-1 protease inhibitor, were investigated in male and female Sprague-Dawley rats, beagle dogs, and HIV-negative male human volunteers. Rats and dogs received a 5 mg/kg iv, 20 mg/kg oral or 20 mg/kg intraduodenal dose, whereas humans received a single 600-mg liquid oral dose. Ritonavir was cleared primarily via hepatobiliary elimination in all three species. After iv or oral dosing in either rats or dogs, > 92% of the dose was recovered in rat and dog feces and < or = 4% was recovered in the urine. Humans excreted 86.3% of the oral dose in feces and 11.3% in urine over 6 days. Bile-exteriorized rats and dogs excreted 85.5% and 39.8%, respectively, of the iv dose in bile, with < 3% recovered in urine. Radio-HPLC analysis of bile, feces, and urine from all three species indicated extensive metabolism of ritonavir to a number of oxidative metabolites, some being species-specific, and all involving metabolism at the terminal functional groups of the molecule. Glucuronide metabolites were observed in dog only. Plasma radioactivity consisted predominantly of unchanged parent drug in all three species. M-2, the product of hydroxylation at the methine carbon of the terminal isopropyl moiety of ritonavir, was the only metabolite present in human plasma and made up 30.4% of the total dose recovered in human excreta over 6 days. Tissue distribution of ritonavir in rat was widespread, with good distribution into lymphatic tissue but low CNS penetration. Plasma protein binding of ritonavir was high (96-99.5%) in all species and was nonsaturable in humans at concentrations up to 30 micrograms/ml. Partitioning into the formed elements of whole blood was minimal.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Ritonavir/pharmacokinetics , Adult , Animals , Bile/metabolism , Blood Proteins/metabolism , Chromatography, High Pressure Liquid , Dogs , Feces/chemistry , Female , HIV Protease Inhibitors/blood , Humans , Male , Mass Spectrometry , Protein Binding , Rats , Rats, Sprague-Dawley , Ritonavir/blood , Ritonavir/urine , Tissue Distribution
15.
Antimicrob Agents Chemother ; 41(3): 654-60, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9056009

ABSTRACT

Coadministration with the human immunodeficiency virus (HIV) protease inhibitor ritonavir was investigated as a method for enhancing the levels of other peptidomimetic HIV protease inhibitors in plasma. In rat and human liver microsomes, ritonavir potently inhibited the cytochrome P450 (CYP)-mediated metabolism of saquinavir, indinavir, nelfinavir, and VX-478. The structural features of ritonavir responsible for CYP binding and inhibition were examined. Coadministration of other protease inhibitors with ritonavir in rats and dogs produced elevated and sustained plasma drug levels 8 to 12 h after a single dose. Drug exposure in rats was elevated by 8- to 46-fold. A > 50-fold enhancement of the concentrations of saquinavir in plasma was observed in humans following a single codose of ritonavir (600 mg) and saquinavir (200 mg). These results indicate that ritonavir can favorably alter the pharmacokinetic profiles of other protease inhibitors. Combination regimens of ritonavir and other protease inhibitors may thus play a role in the treatment of HIV infection. Because of potentially substantial drug level increases, however, such combinations require further investigation to establish safe regimens for clinical use.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Protease Inhibitors/pharmacokinetics , Ritonavir/pharmacology , Animals , Area Under Curve , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Dogs , Drug Interactions , Female , HIV Protease Inhibitors/pharmacology , Humans , Male , Rats , Rats, Sprague-Dawley
16.
Nat Med ; 2(7): 760-6, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8673921

ABSTRACT

Analysis of the HIV protease gene from the plasma of HIV-infected patients revealed substitutions at nine different codons selected in response to monotherapy with the protease inhibitor ritonavir. Mutants at valine-82, although insufficient to confer resistance, appeared first in most patients. Significant phenotypic resistance required multiple mutations in HIV protease, which emerged subsequently in an ordered, stepwise fashion. The appearance of resistance mutations was delayed in patients with higher plasma levels of ritonavir. Early mutants retained susceptibility to structurally diverse protease inhibitors, suggesting that dual protease inhibitor therapy might increase the duration of viral suppression.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV Protease/genetics , HIV/drug effects , Mutation , Thiazoles/pharmacology , Valine/analogs & derivatives , Codon , Genotype , HIV/enzymology , HIV/genetics , HIV Infections/blood , Humans , Phenotype , Ritonavir , Valine/genetics , Valine/pharmacology
17.
Antimicrob Agents Chemother ; 40(6): 1491-7, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8726025

ABSTRACT

The therapeutic utility of a human immunodeficiency virus type 1 (HIV-1) protease inhibitor may depend on its intracellular concentration, which is a property of its uptake, metabolism, and/or efflux. Previous studies in our laboratory indicated that the addition of alpha 1 acid glycoprotein (alpha 1 AGP) to the medium markedly increased the amount of the drug required to limit infection in vitro. In this study, physiologically relevant concentrations of alpha 1 AGP and a radiolabeled inhibitor, A-80987, were used to determine both the uptake and activity of the agent in HIV-1-infected human peripheral blood mononuclear cells and cell lines. Both the uptake and efflux of 14C-labeled A-80987 were rapid (t1/2, < 5 min). Uptake of the drug was linearly dependent on the concentration but insensitive to the metabolic inhibitors KF, sodium cyanide, or CCCP (carbonyl cyanide m-chlorophenyl hydrazone). The amount of A-80987 which entered the cells was inversely proportional to the concentration of alpha 1 AGP (r2, 0.99) and directly proportional to the amount of extracellular non-protein-bound drug (r2, 0.99). Most importantly, the antiviral activity of the drug in HIV-1-infected peripheral blood mononuclear cells and MT-2 cells was directly related to the amount of intracellular A-80987. This study demonstrates that A-80987 binds to alpha 1 AGP, resulting in a free fraction below 10%. Cellular uptake of A-80987 is proportionally decreased in the presence of alpha 1 AGP, which results in less-than-expected antiviral activity. Importantly, we demonstrate for the first time that the inhibition of HIV protease is highly correlated with the amount of intracellular inhibitor.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , HIV-1/drug effects , Orosomucoid/pharmacology , Pyridines/pharmacokinetics , Cell Line , HIV Protease Inhibitors/metabolism , HIV-1/metabolism , Humans , Orosomucoid/metabolism , Polymerase Chain Reaction , Protein Binding , Pyridines/metabolism , RNA, Viral/drug effects
18.
J Med Chem ; 39(2): 392-7, 1996 Jan 19.
Article in English | MEDLINE | ID: mdl-8558507

ABSTRACT

The design, synthesis, and molecular modeling studies of a novel series of azacyclic ureas, which are inhibitors of human immunodeficiency virus type 1 (HIV-1) protease that incorporate different ligands for the S1', S2, and S2' substrate-binding sites of HIV-1 protease are described. The synthesis of this series is highly flexible in the sense that the P1', P2, and P2' residues of the inhibitors can be changed independently. Molecular modeling studies on the phenyl ring of the P2 and P2' ligand suggested incorporation of hydrogen-bonding donor/acceptor groups at the 3' and 4-positions of the phenyl ring should increase binding potency. This led to the discovery of compound 7f (A-98881), which possesses high potency in the HIV-1 protease inhibition assay and the in vitro MT-4 cell culture assay (Ki = approximately 5 pM and EC50 = 0.002 microM). This compares well with the symmetrical cyclic urea 1 pioneered at DuPont Merck.


Subject(s)
HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/pharmacology , HIV-1/enzymology , Binding Sites , Drug Resistance, Microbial , HIV Protease/metabolism , HIV-1/drug effects , Models, Molecular
19.
J Med Chem ; 38(16): 2995-3002, 1995 Aug 04.
Article in English | MEDLINE | ID: mdl-7636862

ABSTRACT

Solid phase synthesis of non-oligomeric organic compounds has been pursued for high-efficiency generation of large numbers of structurally diverse compounds for drug screening. Known as chemical diversity libraries or combinatorial libraries (when the synthesis is carried out in a combinatorial fashion), these compounds can be used for de novo discovery of drug leads or for expedient structure--activity relationship (SAR) studies. To expand the scope of solid phase synthesis beyond the capability of the traditional method of solid phase synthesis for peptides, a strategy was developed for bi-directional solid phase synthesis starting with diamino alcohol or diamino diol core structures. The strategy relies on using bifunctional linkers to modify the core structures, simultaneously protecting the hydroxyl group or the diol moiety of the core and providing a carboxyl group for attachment of the modified cores to a solid support. The two NH2 groups of the modified cores attached to the solid support were then deprotected and reacted with a wide variety of amine-reactive reagents (carboxylic acids, sulfonyl chlorides, isocyanates, chloroformates, etc.) to extend the molecule in both directions. This strategy was successfully applied to automated parallel synthesis of a library of C2 symmetric inhibitors of HIV protease containing the known symmetry-based diamino diol and diamino alcohol core structures, thus enabling expedient access of large numbers of analogs in this series. A library of over 300 discrete compounds was synthesized using this methodology in order to identify potent (IC50 < 100 nM) HIV protease inhibitors with reduced size. This paper describes the technical aspects of this technology.


Subject(s)
HIV Protease Inhibitors/chemical synthesis , Alcohols/chemistry , Alcohols/pharmacology , Feasibility Studies , HIV Protease Inhibitors/chemistry , Humans , Methylurea Compounds/chemistry , Pyridines/chemistry , Structure-Activity Relationship , Valine/analogs & derivatives
20.
Proc Natl Acad Sci U S A ; 92(7): 2484-8, 1995 Mar 28.
Article in English | MEDLINE | ID: mdl-7708670

ABSTRACT

Examination of the structural basis for antiviral activity, oral pharmacokinetics, and hepatic metabolism among a series of symmetry-based inhibitors of the human immunodeficiency virus (HIV) protease led to the discovery of ABT-538, a promising experimental drug for the therapeutic intervention in acquired immunodeficiency syndrome (AIDS). ABT-538 exhibited potent in vitro activity against laboratory and clinical strains of HIV-1 [50% effective concentration (EC50) = 0.022-0.13 microM] and HIV-2 (EC50 = 0.16 microM). Following a single 10-mg/kg oral dose, plasma concentrations in rat, dog, and monkey exceeded the in vitro antiviral EC50 for > 12 h. In human trials, a single 400-mg dose of ABT-538 displayed a prolonged absorption profile and achieved a peak plasma concentration in excess of 5 micrograms/ml. These findings demonstrate that high oral bioavailability can be achieved in humans with peptidomimetic inhibitors of HIV protease.


Subject(s)
Antiviral Agents/pharmacokinetics , HIV Protease Inhibitors/pharmacokinetics , Thiazoles/pharmacokinetics , Valine/analogs & derivatives , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Bile/metabolism , Bile Ducts/physiology , Binding Sites , Biological Availability , Capsules , Female , HIV Protease/chemistry , HIV Protease Inhibitors/administration & dosage , HIV-1/drug effects , HIV-2/drug effects , Humans , Injections, Intravenous , Macaca fascicularis , Male , Metabolic Clearance Rate , Models, Molecular , Molecular Structure , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Ritonavir , Tablets , Thiazoles/administration & dosage , Thiazoles/pharmacology , Tissue Distribution , Valine/administration & dosage , Valine/pharmacokinetics , Valine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...