Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 685: 241-52, 2011.
Article in English | MEDLINE | ID: mdl-20981527

ABSTRACT

Fragment-based drug design (FBDD), which is comprised of both fragment screening and the use of fragment hits to design leads, began more than 15 years ago and has been steadily gaining in popularity and utility. Its origin lies on the fact that the coverage of chemical space and the binding efficiency of hits are directly related to the size of the compounds screened. Nevertheless, FBDD still faces challenges, among them developing fragment screening libraries that ensure optimal coverage of chemical space, physical properties and chemical tractability. Fragment screening also requires sensitive assays, often biophysical in nature, to detect weak binders. In this chapter we will introduce the technologies used to address these challenges and outline the experimental advantages that make FBDD one of the most popular new hit-to-lead process.


Subject(s)
Drug Design , Crystallography, X-Ray , Drug Evaluation, Preclinical , Magnetic Resonance Spectroscopy , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
2.
J Med Chem ; 53(16): 6003-17, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20718494

ABSTRACT

Previously, we reported the discovery of PSI-697 (1a), a C-2 benzyl substituted quinoline salicylic acid-based P-selectin inhibitor. It is active in a variety of animal models of cardiovascular disease. Compound 1a has also been shown to be well tolerated and safe in healthy volunteers at doses of up to 1200 mg in a phase 1 single ascending dose study. However, its oral bioavailability was low. Our goal was to identify a back up compound with equal potency, increased solubility, and increased exposure. We expanded our structure-activity studies in this series by branching at the alpha position of the C-2 benzyl side chain and through modification of substituents on the carboxylic A-ring of the quinoline. This resulted in discovery of PSI-421 with marked improvement in aqueous solubility and pharmacokinetic properties. This compound has shown oral efficacy in animal models of arterial and venous injury and was selected as a preclinical development compound for potential treatment of such diseases as atherosclerosis and deep vein thrombosis.


Subject(s)
Carotid Artery Injuries/drug therapy , Hydroxyquinolines/chemical synthesis , P-Selectin/antagonists & inhibitors , Salicylates/chemical synthesis , Venous Thrombosis/drug therapy , Administration, Oral , Animals , Caco-2 Cells , Cell Membrane Permeability , Dogs , Drug Stability , Humans , Hydroxyquinolines/pharmacokinetics , Hydroxyquinolines/pharmacology , Leukocyte Rolling/drug effects , Macaca fascicularis , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Models, Molecular , Papio , Rats , Rats, Sprague-Dawley , Salicylates/chemistry , Salicylates/pharmacology , Solubility , Structure-Activity Relationship
3.
J Med Chem ; 53(16): 6122-8, 2010 Aug 26.
Article in English | MEDLINE | ID: mdl-20666458

ABSTRACT

Acidic mammalian chitinase (AMCase) is a member of the glycosyl hydrolase 18 family (EC 3.2.1.14) that has been implicated in the pathophysiology of allergic airway disease such as asthma. Small molecule inhibitors of AMCase were identified using a combination of high-throughput screening, fragment screening, and virtual screening techniques and characterized by enzyme inhibition and NMR and Biacore binding experiments. X-ray structures of the inhibitors in complex with AMCase revealed that the larger more potent HTS hits, e.g. 5-(4-(2-(4-bromophenoxy)ethyl)piperazine-1-yl)-1H-1,2,4-triazol-3-amine 1, spanned from the active site pocket to a hydrophobic pocket. Smaller fragments identified by FBS occupy both these pockets independently and suggest potential strategies for linking fragments. Compound 1 is a 200 nM AMCase inhibitor which reduced AMCase enzymatic activity in the bronchoalveolar lavage fluid in allergen-challenged mice after oral dosing.


Subject(s)
Chitinases/antagonists & inhibitors , Models, Molecular , Piperazines/chemical synthesis , Triazoles/chemical synthesis , Allergens/immunology , Animals , Bronchoalveolar Lavage Fluid , Catalytic Domain , Crystallography, X-Ray , Female , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred C57BL , Piperazines/chemistry , Piperazines/pharmacology , Protein Binding , Respiratory Hypersensitivity/drug therapy , Respiratory Hypersensitivity/enzymology , Respiratory Hypersensitivity/immunology , Structure-Activity Relationship , Surface Plasmon Resonance , Triazoles/chemistry , Triazoles/pharmacology
4.
Biochim Biophys Acta ; 1798(2): 87-93, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19835839

ABSTRACT

Neisseria meningitidis is a major cause of meningitis. Although protective vaccination is available against some pathogenic serogroups, serogroup B meningococci have been a challenge for vaccinologists. A family of outer membrane lipoproteins, LP2086 (or factor H binding proteins, fHbp), has been shown to elicit bactericidal antibodies and is currently part of a cocktail vaccine candidate. The NMR structure of the variant LP2086-B01 in micellar solution provided insights on the topology of this family of proteins on the biological membrane. Based on flow cytometry experiments on whole meningococcal cells, binding experiments with monoclonal antibodies, and the NMR structure in micellar solution, we previously proposed that LP2086-B01 anchors the outer bacterial membrane through its lipidated N-terminal cysteine, while a flexible 20 residue linker positions the protein above the layer of lipo-oligosaccharides that surrounds the bacteria. This topology was suggested to increase the antigen exposure to the immune system. In the present work, using micellar solution as a membrane mimicking system, we characterized the backbone dynamics of the variant LP2086-B01 in both its lipidated and unlipidated forms. In addition, binding experiments with a Fab fragment derived from the monoclonal MN86-1042-2 were also performed. Our data suggests that due to the length and flexibility of the N-terminal linker, the antigen is not in contact with the micelle, thus making both N- and C-domains highly available to the host immune system. This dynamic model, combined with the binding data obtained with MN86-1042-2, supports our previously proposed arrangement that LP2086-B01 exposes one face to the extracellular space. Binding of MN86-1042-2 antibody shows that the N-domain is the primary target of this monoclonal, providing further indication that this domain is immunologically important for this family of proteins.


Subject(s)
Antibodies, Bacterial/chemistry , Antibodies, Monoclonal/chemistry , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Lipopolysaccharides/chemistry , Models, Molecular , Neisseria meningitidis/chemistry , Animals , Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Humans , Lipopolysaccharides/immunology , Mice , Micelles , Neisseria meningitidis/immunology , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Tertiary/physiology
5.
J Med Chem ; 53(3): 1238-49, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20038108

ABSTRACT

To aid in the pursuit of selective kinase inhibitors, we have developed a unique ATP site binder tool for the detection of binders outside the ATP site by nuclear magnetic resonance (NMR). We report here the novel synthesis that led to this paramagnetic spin-labeled pyrazolopyrimidine probe (1), which exhibits nanomolar inhibitory activity against multiple kinases. We demonstrate the application of this probe by performing NMR binding experiments with Lck and Src kinases and utilize it to detect the binding of two compounds proximal to the ATP site. The complex structure of the probe with Lck is also presented, revealing how the probe fits in the ATP site and the specific interactions it has with the protein. We believe that this spin-labeled probe is a valuable tool that holds broad applicability in a screen for non-ATP site binders.


Subject(s)
Adenosine Triphosphate/metabolism , Magnetic Resonance Spectroscopy , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/chemistry , Protein Kinases/metabolism , Spin Labels/chemical synthesis , Binding Sites , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Humans , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
6.
J Biol Chem ; 284(13): 8738-46, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19103601

ABSTRACT

LP2086 is a family of outer membrane lipoproteins from Neisseria meningitidis, which elicits bactericidal antibodies and are currently undergoing human clinical trials in a bivalent formulation where each antigen represents one of the two known LP2086 subfamilies. Here we report the NMR structure of the recombinant LP2086 variant B01, a representative of the LP2086 subfamily B. The structure reveals a novel fold composed of two domains: a "taco-shaped" N-terminal beta-sheet and a C-terminal beta-barrel connected by a linker. The structure in micellar solution is consistent with a model of LP2086 anchored to the outer membrane bilayer through its lipidated N terminus. A long flexible chain connects the folded part of the protein to the lipid anchor and acts as spacer, making both domains accessible to the host immune system. Antibodies broadly reactive against members from both subfamilies have been mapped to the N terminus. A surface of subfamily-defining residues was identified on one face of the protein, offering an explanation for the induction of subfamily-specific bactericidal antibodies.


Subject(s)
Antibodies, Bacterial/chemistry , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Lipid Bilayers/chemistry , Meningococcal Vaccines/chemistry , Micelles , Neisseria meningitidis/chemistry , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Base Sequence , Humans , Lipid Bilayers/immunology , Meningococcal Vaccines/genetics , Meningococcal Vaccines/immunology , Mice , Molecular Sequence Data , Neisseria meningitidis/genetics , Neisseria meningitidis/immunology , Nuclear Magnetic Resonance, Biomolecular/methods , Peptide Mapping/methods , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
7.
J Med Chem ; 51(19): 5958-63, 2008 Oct 09.
Article in English | MEDLINE | ID: mdl-18783200

ABSTRACT

The protein kinase C (PKC) family of serine/threonine kinases is implicated in a wide variety of cellular processes. The PKC theta (PKCtheta) isoform is involved in TCR signal transduction and T cell activation and regulates T cell mediated diseases, including lung inflammation and airway hyperresponsiveness. Thus inhibition of PKCtheta enzyme activity by a small molecule represents an attractive strategy for the treatment of asthma. A PKCtheta high-throughput screening (HTS) campaign led to the identification of 4-(3-bromophenylamino)-5-(3,4-dimethoxyphenyl)-3-pyridinecarbonitrile 4a, a low microM ATP competitive PKCtheta inhibitor. Structure based hit-to-lead optimization led to the identification of 5-(3,4-dimethoxyphenyl)-4-(1H-indol-5-ylamino)-3-pyridinecarbonitrile 4p, a 70 nM PKCtheta inhibitor. Compound 4p was selective for inhibition of novel PKC isoforms over a panel of 21 serine/threonine, tyrosine, and phosphoinositol kinases, in addition to the conventional and atypical PKCs, PKCbeta, and PKCzeta, respectively. Compound 4p also inhibited IL-2 production in antiCD3/anti-CD28 activated T cells enriched from splenocytes.


Subject(s)
Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Nitriles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Indoles/chemical synthesis , Indoles/chemistry , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Isoenzymes/deficiency , Isoenzymes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Protein Kinase C/deficiency , Protein Kinase C/drug effects , Protein Kinase C-theta , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Stereoisomerism , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
10.
J Med Chem ; 50(1): 21-39, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17201408

ABSTRACT

Leukocyte recruitment of sites of inflammation and tissue injury involves leukocyte rolling along the endothelial wall, followed by firm adherence of the leukocyte, and finally transmigration of the leukocyte across cell junctions into the underlying tissue. The initial rolling step is mediated by the interaction of leukocyte glycoproteins containing active moieties such as sialyl Lewisx (sLex) with P-selectin expressed on endothelial cells. Consequently, inhibition of this interaction by means of a small molecule P-selectin antagonist is an attractive strategy for the treatment of inflammatory diseases such as arthritis. High-throughput screening of the Wyeth chemical library identified the quinoline salicylic acid class of compounds (1) as antagonists of P-selectin, with potency in in vitro and cell-based assays far superior to that of sLex. Through iterative medicinal chemistry, we identified analogues with improved P-selectin activity, decreased inhibition of dihydrooratate dehydrogenase, and acceptable CYP profiles. Lead compound 36 was efficacious in the rat AIA model of rheumatoid arthritis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Hydroxyquinolines/chemical synthesis , P-Selectin/metabolism , Quinolines/chemical synthesis , Salicylates/chemical synthesis , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Biological Availability , Cytochrome P-450 Enzyme Inhibitors , Databases, Factual , Edema/drug therapy , Humans , Hydroxyquinolines/pharmacokinetics , Hydroxyquinolines/pharmacology , In Vitro Techniques , Leukocyte Rolling/drug effects , Male , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Salicylates/pharmacokinetics , Salicylates/pharmacology , Structure-Activity Relationship
11.
J Med Chem ; 50(1): 40-64, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17201409

ABSTRACT

P-selectin-PSGL-1 interaction causes rolling of leukocytes on the endothelial cell surface, which subsequently leads to firm adherence and leukocyte transmigration through the vessel wall into the surrounding tissues. P-selectin is upregulated on the surface of both platelets and endothelium in a variety of atherosclerosis-associated conditions. Consequently, inhibition of this interaction by means of a small molecule P-selectin antagonist is an attractive strategy for the treatment of atherosclerosis. High-throughput screening and subsequent analoging had led to the identification of compound 1 as the lead candidate. Herein, we report the continuation of this work and the discovery of a second-generation series, the tetrahydrobenzoquinoline salicylic acids. These compounds have improved pharmacokinetic properties, and a number of them have shown oral efficacy in mouse and rat models of atherogenesis and vascular injury. The lead 31 (PSI-697), is currently in clinical development for the treatment of atherothrombotic vascular events.


Subject(s)
Atherosclerosis/prevention & control , Fibrinolytic Agents/chemical synthesis , Hydroxyquinolines/chemical synthesis , P-Selectin/metabolism , Quinolines/chemical synthesis , Salicylates/chemical synthesis , Administration, Oral , Animals , Apolipoproteins E/genetics , Carotid Stenosis/prevention & control , Dogs , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Hydroxyquinolines/pharmacokinetics , Hydroxyquinolines/pharmacology , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Leukocyte Rolling/drug effects , Male , Mice , Mice, Knockout , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Salicylates/pharmacokinetics , Salicylates/pharmacology , Structure-Activity Relationship
12.
Bioorg Med Chem ; 14(23): 7953-61, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16919463

ABSTRACT

ZipA is a membrane anchored protein in Escherichia coli that interacts with FtsZ, a homolog of eukaryotic tubulins, forming a septal ring structure that mediates bacterial cell division. Thus, the ZipA/FtsZ protein-protein interaction is a potential target for an antibacterial agent. We report here an NMR-based fragment screening approach which identified several hits that bind to the C-terminal region of ZipA. The screen was performed by 1H-15N HSQC experiments on a library of 825 fragments that are small, lead-like, and highly soluble. Seven hits were identified, and the binding mode of the best one was revealed in the X-ray crystal structure. Similar to the ZipA/FtsZ contacts, the driving force in the binding of the small molecule ligands to ZipA is achieved through hydrophobic interactions. Analogs of this hit were also evaluated by NMR and X-ray crystal structures of these analogs with ZipA were obtained, providing structural information to help guide the medicinal chemistry efforts.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Carrier Proteins/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Escherichia coli Proteins/antagonists & inhibitors , Magnetic Resonance Spectroscopy , Multiprotein Complexes/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Drug Design , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Ligands , Peptide Fragments/metabolism , Protein Binding , Structure-Activity Relationship
13.
J Med Chem ; 48(13): 4346-57, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-15974587

ABSTRACT

A search for noncarbohydrate sLe(x) mimics led to the development of quinic acid derivatives as selectin inhibitors. At Wyeth we solved the first cocrystal structure of a small molecule, quinic acid, with E-selectin. In the cocomplex two hydroxyls of quinic acid mimic the calcium-bound fucose of the tetrasaccharide sLe(x). The X-ray structure, together with structure based computational methods, was used to design quinic acid based libraries that were synthesized and evaluated for their ability to block the interaction of sLex with P-selectin. A large number of analogues were prepared using solution-phase parallel synthesis. Selected compounds showed decrease in leukocyte rolling in the IVM mouse model. Compound 2 inhibited neutrophil influx in the murine TIP model and demonstrated good plasma exposure.


Subject(s)
E-Selectin/metabolism , Oligosaccharides/chemistry , Quinic Acid/analogs & derivatives , Quinic Acid/pharmacology , Animals , Binding Sites , Crystallography, X-Ray , Drug Design , Fucose , Jugular Veins/drug effects , Jugular Veins/physiology , Kinetics , Lewis Blood Group Antigens , Magnetic Resonance Spectroscopy , Male , Models, Molecular , Molecular Conformation , Molecular Structure , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Oligosaccharides/chemical synthesis , Oligosaccharides/pharmacology , Rats , Rats, Sprague-Dawley , Sialyl Lewis X Antigen , Surface Plasmon Resonance
16.
Bioorg Med Chem Lett ; 14(6): 1427-31, 2004 Mar 22.
Article in English | MEDLINE | ID: mdl-15006376

ABSTRACT

The binding of FtsZ to ZipA is a potential target for antibacterial therapy. Based on a small molecule inhibitor of the ZipA-FtsZ interaction, a parallel synthesis of small molecules was initiated which targeted a key region of ZipA involved in FtsZ binding. The X-ray crystal structure of one of these molecules complexed with ZipA was solved. The structure revealed an unexpected binding mode, facilitated by desolvation of a loosely bound surface water.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Drug Design , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/metabolism , Indoles/chemical synthesis , Quinazolines/chemical synthesis , Amino Acid Sequence , Indoles/chemistry , Indoles/metabolism , Molecular Sequence Data , Protein Binding/physiology , Quinazolines/chemistry , Quinazolines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...