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.
PLoS One ; 9(5): e97696, 2014.
Article in English | MEDLINE | ID: mdl-24846127

ABSTRACT

This study describes the development of aptamers as a therapy against influenza virus infection. Aptamers are oligonucleotides (like ssDNA or RNA) that are capable of binding to a variety of molecular targets with high affinity and specificity. We have studied the ssDNA aptamer BV02, which was designed to inhibit influenza infection by targeting the hemagglutinin viral protein, a protein that facilitates the first stage of the virus' infection. While testing other aptamers and during lead optimization, we realized that the dominant characteristics that determine the aptamer's binding to the influenza virus may not necessarily be sequence-specific, as with other known aptamers, but rather depend on general 2D structural motifs. We adopted QSAR (quantitative structure activity relationship) tool and developed computational algorithm that correlate six calculated structural and physicochemical properties to the aptamers' binding affinity to the virus. The QSAR study provided us with a predictive tool of the binding potential of an aptamer to the influenza virus. The correlation between the calculated and actual binding was R2 = 0.702 for the training set, and R2 = 0.66 for the independent test set. Moreover, in the test set the model's sensitivity was 89%, and the specificity was 87%, in selecting aptamers with enhanced viral binding. The most important properties that positively correlated with the aptamer's binding were the aptamer length, 2D-loops and repeating sequences of C nucleotides. Based on the structure-activity study, we have managed to produce aptamers having viral affinity that was more than 20 times higher than that of the original BV02 aptamer. Further testing of influenza infection in cell culture and animal models yielded aptamers with 10 to 15 times greater anti-viral activity than the BV02 aptamer. Our insights concerning the mechanism of action and the structural and physicochemical properties that govern the interaction with the influenza virus are discussed.


Subject(s)
Antiviral Agents , Aptamers, Nucleotide , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Orthomyxoviridae Infections/drug therapy , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Dogs , Drug Design , Madin Darby Canine Kidney Cells , Structure-Activity Relationship
2.
Neuropharmacology ; 53(4): 563-73, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17692343

ABSTRACT

Recent evidence suggests that 5-hydroxytryptamine (5-HT)(4) receptor activity enhances cognition and provides neuroprotection. Here we report the effects of VRX-03011, a novel partial 5-HT(4) agonist, that is both potent (K(i) approximately 30 nM) and highly selective (K(i) > 5 microM for all other 5-HT receptors tested). In separate experiments, rats received VRX-03011 (0.1-10 mg/kg i.p.) 30 min prior to spontaneous alternation testing in a no-delay or a 30-s delay condition. VRX-03011 (1, 5 and 10 mg/kg, but not 0.1 mg/kg) significantly enhanced delayed spontaneous alternation performance while none of the doses enhanced performance in the no-delay test. VRX-03011 (1 and 5 mg/kg) concomitantly enhanced hippocampal acetylcholine output and delayed spontaneous alternation scores compared to that of vehicle controls, but had no effect on hippocampal acetylcholine release under a resting condition. Moreover, suboptimal doses of VRX-03011 and the acetylcholinesterase inhibitor galanthamine combined to enhance memory. VRX-03011 also regulated amyloid precursor protein (APP) metabolism by inducing a concentration-dependent increase in the non-amyloidogenic soluble form of APP (sAPPalpha) with an EC(50) approximately 1--10 nM. VRX-03011 had no effect on contractile properties in guinea pig ileum or colon preparations with an EC(50) > 10 microM and did not alter rat intestinal transit at doses up to 10 mg/kg. These findings suggest that VRX-03011 may represent a novel treatment for Alzheimer's disease that reduces cognitive impairments and provides neuroprotection without gastrointestinal side effects.


Subject(s)
Acetylcholine/physiology , Hippocampus/physiology , Memory/physiology , Pyridones/pharmacology , Receptors, Serotonin, 5-HT4/physiology , Serotonin Receptor Agonists/pharmacology , Thiophenes/pharmacology , Animals , Cognition/drug effects , Cognition/physiology , Hippocampus/cytology , Hippocampus/drug effects , Ligands , Memory/drug effects , Models, Animal , Rats , Receptors, Serotonin, 5-HT4/drug effects , Receptors, Serotonin, 5-HT4/genetics , Transfection
3.
J Med Chem ; 49(11): 3116-35, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16722631

ABSTRACT

We report the discovery of a novel, potent, and selective amidosulfonamide nonazapirone 5-HT1A agonist for the treatment of anxiety and depression, which is now in Phase III clinical trials for generalized anxiety disorder (GAD). The discovery of 20m (PRX-00023), N-{3-[4-(4-cyclohexylmethanesulfonylaminobutyl)piperazin-1-yl]phenyl}acetamide, and its backup compounds, followed a new paradigm, driving the entire discovery process with in silico methods and seamlessly integrating computational chemistry with medicinal chemistry, which led to a very rapid discovery timeline. The program reached clinical trials within less than 2 years from initiation, spending less than 6 months in lead optimization with only 31 compounds synthesized. In this paper we detail the entire discovery process, which started with modeling the 3D structure of 5-HT1A using the PREDICT methodology, and then performing in silico screening on that structure leading to the discovery of a 1 nM lead compound (8). The lead compound was optimized following a strategy devised based on in silico 3D models and realized through an in silico-driven optimization process, rapidly overcoming selectivity issues (affinity to 5-HT1A vs alpha1-adrenergic receptor) and potential cardiovascular issues (hERG binding), leading to a clinical compound. Finally we report key in vivo preclinical and Phase I clinical data for 20m tolerability, pharmacokinetics, and pharmacodynamics and show that these favorable results are a direct outcome of the properties that were ascribed to the compound during the rational structure-based discovery process. We believe that this is one of the first examples for a Phase III drug candidate that was discovered and optimized, from start to finish, using in silico model-based methods as the primary tool.


Subject(s)
Anti-Anxiety Agents/chemistry , Antidepressive Agents/chemistry , Models, Molecular , Piperazines/chemical synthesis , Serotonin 5-HT1 Receptor Agonists , Sulfonamides/chemistry , Animals , Anti-Anxiety Agents/chemical synthesis , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/chemical synthesis , Antidepressive Agents/pharmacology , Binding, Competitive , Biological Availability , Cell Line , Clinical Trials, Phase I as Topic , Dogs , Drug Design , Half-Life , Humans , In Vitro Techniques , Male , Mice , Microsomes, Liver/metabolism , Patch-Clamp Techniques , Piperazines/chemistry , Piperazines/pharmacology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
4.
Proteins ; 57(1): 51-86, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15326594

ABSTRACT

G-protein coupled receptors (GPCRs) are a major group of drug targets for which only one x-ray structure is known (the nondrugable rhodopsin), limiting the application of structure-based drug discovery to GPCRs. In this paper we present the details of PREDICT, a new algorithmic approach for modeling the 3D structure of GPCRs without relying on homology to rhodopsin. PREDICT, which focuses on the transmembrane domain of GPCRs, starts from the primary sequence of the receptor, simultaneously optimizing multiple 'decoy' conformations of the protein in order to find its most stable structure, culminating in a virtual receptor-ligand complex. In this paper we present a comprehensive analysis of three PREDICT models for the dopamine D2, neurokinin NK1, and neuropeptide Y Y1 receptors. A shorter discussion of the CCR3 receptor model is also included. All models were found to be in good agreement with a large body of experimental data. The quality of the PREDICT models, at least for drug discovery purposes, was evaluated by their successful utilization in in-silico screening. Virtual screening using all three PREDICT models yielded enrichment factors 9-fold to 44-fold better than random screening. Namely, the PREDICT models can be used to identify active small-molecule ligands embedded in large compound libraries with an efficiency comparable to that obtained using crystal structures for non-GPCR targets.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Algorithms , Amino Acid Sequence , Animals , Binding Sites , Combinatorial Chemistry Techniques , Computer Simulation , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Chemical , Models, Molecular , Monte Carlo Method , Protein Conformation , Protein Structure, Secondary , Receptors, Dopamine D2/chemistry , Receptors, Neurokinin-1/chemistry , Receptors, Neuropeptide Y/chemistry , Rhodopsin/chemistry , Stereoisomerism , Thermodynamics
5.
Proc Natl Acad Sci U S A ; 101(31): 11304-9, 2004 Aug 03.
Article in English | MEDLINE | ID: mdl-15277683

ABSTRACT

The application of structure-based in silico methods to drug discovery is still considered a major challenge, especially when the x-ray structure of the target protein is unknown. Such is the case with human G protein-coupled receptors (GPCRs), one of the most important families of drug targets, where in the absence of x-ray structures, one has to rely on in silico 3D models. We report repeated success in using ab initio in silico GPCR models, generated by the predict method, for blind in silico screening when applied to a set of five different GPCR drug targets. More than 100,000 compounds were typically screened in silico for each target, leading to a selection of <100 "virtual hit" compounds to be tested in the lab. In vitro binding assays of the selected compounds confirm high hit rates, of 12-21% (full dose-response curves, Ki < 5 microM). In most cases, the best hit was a novel compound (New Chemical Entity) in the 1- to 100-nM range, with very promising pharmacological properties, as measured by a variety of in vitro and in vivo assays. These assays validated the quality of the hits as lead compounds for drug discovery. The results demonstrate the usefulness and robustness of ab initio in silico 3D models and of in silico screening for GPCR drug discovery.


Subject(s)
Algorithms , Drug Design , Models, Chemical , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Binding Sites , Combinatorial Chemistry Techniques , Humans , In Vitro Techniques , Protein Structure, Quaternary , Receptor, Serotonin, 5-HT1A/chemistry , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, CCR3 , Receptors, Chemokine/chemistry , Receptors, Chemokine/metabolism , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Receptors, Neurokinin-1/chemistry , Receptors, Neurokinin-1/metabolism , Receptors, Serotonin, 5-HT4/chemistry , Receptors, Serotonin, 5-HT4/metabolism
6.
Curr Opin Drug Discov Devel ; 6(3): 353-61, 2003 May.
Article in English | MEDLINE | ID: mdl-12833668

ABSTRACT

G protein-coupled receptors (GPCRs) are membrane-embedded proteins responsible for signal transduction; these receptors are, therefore, among the most important pharmaceutical drug targets. In the absence of X-ray structures, there have been numerous attempts to model the three-dimensional (3D) structure of GPCRs. In this review, the current status of GPCR modeling is evaluated, highlighting recent progress made in rhodopsin-based homology modeling and de novo modeling technology. Assessment of recent rhodopsin-based homology modeling studies indicates that, despite significant progress, these models do not yield hit rates that are sufficiently high for in silico screening (10 to 40% when screening for known binders). In contrast, the PREDICT modeling algorithm, which is independent of the rhodopsin structure, has now been fully validated in the context of drug discovery. PREDICT models are successfully used for drug discovery, yielding excellent hit rates (85 to 100% when screening for known binders), leading to the discovery of nanomolar-range new chemical entities for a variety of GPCR targets. Thus, 3D models of GPCRs should now allow the use of productive structure-based approaches for drug discovery.


Subject(s)
Drug Design , GTP-Binding Proteins/chemistry , Models, Molecular , Receptors, Cell Surface/chemistry , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends , Animals , Humans , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...