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.
J Pharmacol Exp Ther ; 290(1): 188-95, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381775

ABSTRACT

The anti-inflammatory properties of a novel pyrrolopyrimidine, PNU-142731A, in a murine model of antigen-induced eosinophilic lung inflammation are described. PNU-142731A, when given orally, demonstrated a dose-related inhibition of eosinophil- and lymphocyte-rich accumulation in the airways of ovalbumin (OA)-sensitized and challenged (OA/OA) C57BL/6 mice. The magnitude of the suppression of lung inflammation was also dependent on length of treatment. Reductions in the levels of interleukin (IL)-5, IL-6, and IgA in the bronchoalveolar lavage fluid of treated OA/OA mice, when compared with vehicle-sensitized control mice (V/OA), were observed. Plasma concentrations of IL-5, total IgE, and OA-specific IgG1 were also lowered in OA/OA mice by treatment. Histological assessment of formalin-fixed lung tissue sections confirmed that the compound blocked the accumulation of eosinophils in the airway tissue. Furthermore, significantly less mucus glycoproteins were seen in the lungs of PNU-142731A-treated OA/OA mice. Reverse transcription-polymerase chain reaction of lung tissue from PNU-142731A-dosed OA/OA mice demonstrated that mRNA for Th2 cytokines was less than that in vehicle-treated OA/OA controls. OA-elicited production of IL-4 by disaggregated lung tissue cells from PNU-142371A-treated OA/OA mice was also less than that of controls. In contrast, the release of Th1 cytokines (IL-2 and interferon-gamma) were elevated. Similarly, the OA-stimulated release of Th2 cytokines (IL-5 and IL-10) by splenocytes from PNU-142731A-treated OA/OA mice were inhibited. Combined therapy of OA/OA mice with PNU-142731A and suboptimal doses of dexamethasone revealed that PNU-142731A had steroid-sparing effects. These characteristics of PNU-142731A in a murine model of allergic tissue inflammation support its clinical development as a potential treatment for asthma.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Indoles/pharmacology , Pyrrolidines/pharmacology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Immunoglobulin A/metabolism , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , Immunoglobulins/biosynthesis , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/immunology , Leukocytes/metabolism , Lung/cytology , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Mucus/metabolism , Ovalbumin/immunology , RNA, Messenger/biosynthesis , Respiratory System/drug effects , Respiratory System/pathology , Reverse Transcriptase Polymerase Chain Reaction , Serine Proteinase Inhibitors/pharmacology , Spleen/cytology , Spleen/drug effects , Spleen/metabolism
2.
J Med Chem ; 40(7): 1149-64, 1997 Mar 28.
Article in English | MEDLINE | ID: mdl-9089336

ABSTRACT

Recently, cyclooctylpyranone derivatives with m-carboxamide substituents (e.g. 2c) were identified as potent, nonpeptidic HIV protease inhibitors, but these compounds lacked significant antiviral activity in cell culture. Substitution of a sulfonamide group at the meta position, however, produces compounds with excellent HIV protease binding affinity and antiviral activity. Guided by an iterative structure-based drug design process, we have prepared and evaluated a number of these derivatives, which are readily available via a seven-step synthesis. A few of the most potent compounds were further evaluated for such characteristics as pharmacokinetics and toxicity in rats and dogs. From this work, the p-cyanophenyl sulfonamide derivative 35k emerged as a promising inhibitor, was selected for further development, and entered phase I clinical trials.


Subject(s)
HIV Protease Inhibitors/chemical synthesis , Pyrones/chemical synthesis , Animals , Cell Line , Crystallography, X-Ray , Dogs , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/pharmacokinetics , Humans , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Models, Molecular , Pyrones/chemistry , Pyrones/pharmacokinetics , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemistry
3.
Chem Res Toxicol ; 9(8): 1230-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8951224

ABSTRACT

U-89843 is a novel pyrrolo[2,3-d]pyrimidine antioxidant with prophylactic activity in animal models of lung inflammation. During preclinical safety evaluation, U-89843 was found to give a positive response in the in vitro unscheduled DNA synthesis (UDS) assay, an assay which measures DNA repair following chemically-induced DNA damage in metabolically competent rat hepatocytes. Incubation of [14C]U-89843 with liver microsomes resulted in covalent binding of radioactive material to macromolecules by a process that was NADPH-dependent. U-89843 has been shown to undergo C-6 methylhydroxylation to give U-97924, in rat both in vivo and in vitro, in a reaction catalyzed by cytochrome P450 2C11. Synthetical U-97924 is chemically reactive and undergoes dimerization in aqueous solution. The dimerization of U-97924 was significantly inhibited by addition of nucleophiles such as methanol, glutathione, and N-acetylcysteine. Characterization of the corresponding methanol, glutathione, and N-acetylcysteine adducts of U-97924 supported the hypothesis of a reaction pathway involving reactive iminium species formed via dehydration of U-97924. The metabolism-dependent irreversible covalent binding of radioactive material to liver microsomal protein and DNA also is dramatically reduced in the presence of reduced glutathione (GSH). A trifluoromethyl analog of U-89843 was prepared in an effort to block the corresponding metabolic hydroxylation pathway. This new compound (U-107634) was found to be negative in the in vitro UDS assay, and its metabolic susceptibility toward hydroxylation at the C-6 methyl group was eliminated. These observations suggest that the positive in vitro UDS results of U-89843 are mediated by the bioactivation of U-89843, leading to reactive electrophilic intermediates derived from the (hydroxymethyl)pyrrole metabolite U-97924.


Subject(s)
Free Radical Scavengers/toxicity , Mutagens/toxicity , Pyrimidines/toxicity , Pyrroles/toxicity , Animals , Biotransformation , Chromatography, High Pressure Liquid , DNA Repair , Dogs , Free Radical Scavengers/chemistry , Free Radical Scavengers/metabolism , Gas Chromatography-Mass Spectrometry , Haplorhini , Humans , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Mutagens/chemistry , Mutagens/metabolism , Protein Binding , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Rats , Rats, Inbred F344 , Spectrometry, Mass, Fast Atom Bombardment , Spectrophotometry, Ultraviolet
4.
Drug Metab Dispos ; 24(2): 187-98, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8742230

ABSTRACT

The biotransformation of 6,7-dimethyl-2,4-di-1-pyrrolidinyl-7H-pyrrolo[2,3-d]pyrimidine (U-89843) has been studied in rat both in vitro and in vivo. Major metabolites observed by HPLC analysis of rat plasma, liver cytosol, and microsomal incubations were characterized by UV, LC/MS, and comparison with synthetic standards. The structures of the metabolites were shown to be the C-6 hydroxymethyl (U-97924), C-6 formyl (U-97865), and C-6 carboxyl analogs of U-89843. In the male rat, formation of U-97924 is mediated by cytochrome P4502C11. Kinetic analysis of U-97924 formation indicated that it was a high-affinity/high-capacity process (KM = 4.2 +/- 0.5 microM; Vmax = 21.2 +/- 0.8 nmol/mg/min). Formation of U-97865 via enzymatic oxidation from the primary metabolite U-97924 was catalyzed by both the microsomal subcellular fraction in a NADPH-dependent process (presumably via cytochrome P450) and in cytosol by NAD(+)-dependent alcohol dehydrogenase. Upon incubation with cytosolic fractions, U-97865 was found to undergo NAD(+)-dependent oxidation, mediated by aldehyde dehydrogenase, to the corresponding carboxylic acid. Although significant levels of U-89843, U-97924, and U-97865 were observed in vivo in rat plasma, only a minor amount of the carboxylic acid together with larger amounts of unidentified polar metabolites were excreted in urine and feces.


Subject(s)
Antioxidants/pharmacokinetics , Microsomes, Liver/metabolism , Pyrimidines/pharmacokinetics , Pyrroles/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet
5.
Acta Neurochir Suppl ; 66: 107-13, 1996.
Article in English | MEDLINE | ID: mdl-8780807

ABSTRACT

The 21-aminosteroid (lazaroid) tirilazad mesylate has been demonstrated to be a potent inhibitor of lipid peroxidation and to reduce traumatic and ischemic damage in a number of experimental models. Currently, tirilazad is being actively investigated in phase III clinical trials in head and spinal cord injury, ischemic stroke and subarachnoid hemorrhage. This compound acts in large part to protect the microvascular endothelium and consequently to maintain normal blood-brain barrier (BBB) permeability and cerebral blood flow autoregulatory mechanisms. However, due to its limited penetration into brain parenchyma, tirilazad has generally failed to affect delayed neuronal damage to the selectively vulnerable hippocampal CA1 and striatal regions. Recently, we have discovered a new group of antioxidant compounds, the pyrrolopyrimidines, which possess significantly improved ability to penetrate the BBB and gain direct access to neural tissue. Several compounds in the series, such as U-101033E, have demonstrated greater ability to protect the CA1 region in the gerbil transient forebrain ischemia model with a post-ischemic therapeutic window of at least four hours. In addition, U-101033E has been found to reduce infarct size in the mouse permanent middle cerebral artery occlusion model in contrast to tirilazad which is minimally effective. These results suggest that antioxidant compounds with improved brain parenchymal penetration are better able to limit certain types of ischemic brain damage compared to those which are localized in the cerebral microvasculature. On the other hand, microvascularly-localized agents like tirilazad appear to have better ability to limit BBB damage.


Subject(s)
Antioxidants/pharmacology , Blood-Brain Barrier/drug effects , Brain Damage, Chronic/pathology , Brain Injuries/pathology , Brain Ischemia/pathology , Neuroprotective Agents/pharmacology , Pregnatrienes/pharmacology , Animals , Blood-Brain Barrier/physiology , Brain/drug effects , Brain/pathology , Free Radical Scavengers/pharmacology , Gerbillinae , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Mice , Microcirculation/drug effects , Microcirculation/physiology , Rats , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...