Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(22): 13546-13560, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32910646

ABSTRACT

Increased fructose consumption and its subsequent metabolism have been implicated in metabolic disorders such as nonalcoholic fatty liver disease and steatohepatitis (NAFLD/NASH) and insulin resistance. Ketohexokinase (KHK) converts fructose to fructose-1-phosphate (F1P) in the first step of the metabolic cascade. Herein we report the discovery of a first-in-class KHK inhibitor, PF-06835919 (8), currently in phase 2 clinical trials. The discovery of 8 was built upon our originally reported, fragment-derived lead 1 and the recognition of an alternative, rotated binding mode upon changing the ribose-pocket binding moiety from a pyrrolidinyl to an azetidinyl ring system. This new binding mode enabled efficient exploration of the vector directed at the Arg-108 residue, leading to the identification of highly potent 3-azabicyclo[3.1.0]hexane acetic acid-based KHK inhibitors by combined use of parallel medicinal chemistry and structure-based drug design.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/chemistry , Fructokinases/antagonists & inhibitors , Fructokinases/metabolism , Fructose/adverse effects , Metabolic Diseases/enzymology , Animals , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Fructose/administration & dosage , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Insulin Resistance/physiology , Male , Metabolic Diseases/chemically induced , Metabolic Diseases/drug therapy , Protein Structure, Secondary , Rats , Rats, Wistar
2.
J Med Chem ; 61(10): 4635-4640, 2018 05 24.
Article in English | MEDLINE | ID: mdl-29718668

ABSTRACT

We disclose the discovery and X-ray cocrystal data of potent, selective quinazoline inhibitors of PDE1. Inhibitor ( S)-3 readily attains free plasma concentrations above PDE1 IC50 values and has restricted brain access. The racemic compound 3 inhibits >75% of PDE hydrolytic activity in soluble samples of human myocardium, consistent with heightened PDE1 activity in this tissue. These compounds represent promising new tools to probe the value of PDE1 inhibition in the treatment of cardiovascular disease.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 1/antagonists & inhibitors , Drug Discovery , Myocardium/enzymology , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Quinazolines/chemistry , Cyclic AMP/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Conformation
4.
J Med Chem ; 61(7): 3008-3026, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29498843

ABSTRACT

Monoacylglycerol lipase (MAGL) inhibition provides a potential treatment approach to neuroinflammation through modulation of both the endocannabinoid pathway and arachidonoyl signaling in the central nervous system (CNS). Herein we report the discovery of compound 15 (PF-06795071), a potent and selective covalent MAGL inhibitor, featuring a novel trifluoromethyl glycol leaving group that confers significant physicochemical property improvements as compared with earlier inhibitor series with more lipophilic leaving groups. The design strategy focused on identifying an optimized leaving group that delivers MAGL potency, serine hydrolase selectivity, and CNS exposure while simultaneously reducing log  D, improving solubility, and minimizing chemical lability. Compound 15 achieves excellent CNS exposure, extended 2-AG elevation effect in vivo, and decreased brain inflammatory markers in response to an inflammatory challenge.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carbamates/chemical synthesis , Carbamates/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Neuritis/drug therapy , Amidohydrolases/antagonists & inhibitors , Animals , Arachidonic Acids/metabolism , Biomarkers , Brain Chemistry/drug effects , Dogs , Drug Design , Drug Discovery , Endocannabinoids/metabolism , Glycerides/metabolism , Humans , Macaca mulatta , Models, Molecular , Rats , Rats, Wistar , Structure-Activity Relationship
5.
J Med Chem ; 61(3): 1001-1018, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29293004

ABSTRACT

Computational modeling was used to direct the synthesis of analogs of previously reported phosphodiesterase 2A (PDE2A) inhibitor 1 with an imidazotriazine core to yield compounds of significantly enhanced potency. The analog PF-05180999 (30) was subsequently identified as a preclinical candidate targeting cognitive impairment associated with schizophrenia. Compound 30 demonstrated potent binding to PDE2A in brain tissue, dose responsive mouse brain cGMP increases, and reversal of N-methyl-d-aspartate (NMDA) antagonist-induced (MK-801, ketamine) effects in electrophysiology and working memory models in rats. Preclinical pharmacokinetics revealed unbound brain/unbound plasma levels approaching unity and good oral bioavailability resulting in an average concentration at steady state (Cav,ss) predicted human dose of 30 mg once daily (q.d.). Modeling of a modified release formulation suggested that 25 mg twice daily (b.i.d.) could maintain plasma levels of 30 at or above targeted efficacious plasma levels for 24 h, which became part of the human clinical plan.


Subject(s)
Brain/drug effects , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Animals , Biological Availability , Brain/physiology , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Inhibitory Concentration 50 , Memory, Short-Term/drug effects , Molecular Docking Simulation , Protein Conformation
6.
J Med Chem ; 60(23): 9860-9873, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29148769

ABSTRACT

Monoacylglycerol lipase (MAGL) is the main enzyme responsible for degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG) in the CNS. MAGL catalyzes the conversion of 2-AG to arachidonic acid (AA), a precursor to the proinflammatory eicosannoids such as prostaglandins. Herein we describe highly efficient MAGL inhibitors, identified through a parallel medicinal chemistry approach that highlighted the improved efficiency of azetidine and piperidine-derived carbamates. The discovery and optimization of 3-substituted azetidine carbamate irreversible inhibitors of MAGL were aided by the generation of inhibitor-bound MAGL crystal structures. Compound 6, a highly efficient and selective MAGL inhibitor against recombinant enzyme and in a cellular context, was tested in vivo and shown to elevate central 2-AG levels at a 10 mg/kg dose.


Subject(s)
Azetidines/pharmacology , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Piperidines/pharmacology , Animals , Azetidines/chemistry , Azetidines/pharmacokinetics , Carbamates/chemistry , Carbamates/pharmacokinetics , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Mice, Inbred C57BL , Models, Molecular , Monoacylglycerol Lipases/metabolism , Piperidines/chemistry , Piperidines/pharmacokinetics , Recombinant Proteins/metabolism
7.
J Med Chem ; 60(18): 7835-7849, 2017 09 28.
Article in English | MEDLINE | ID: mdl-28853885

ABSTRACT

Increased fructose consumption and its subsequent metabolism have been implicated in hepatic steatosis, dyslipidemia, obesity, and insulin resistance in humans. Since ketohexokinase (KHK) is the principal enzyme responsible for fructose metabolism, identification of a selective KHK inhibitor may help to further elucidate the effect of KHK inhibition on these metabolic disorders. Until now, studies on KHK inhibition with small molecules have been limited due to the lack of viable in vivo pharmacological tools. Herein we report the discovery of 12, a selective KHK inhibitor with potency and properties suitable for evaluating KHK inhibition in rat models. Key structural features interacting with KHK were discovered through fragment-based screening and subsequent optimization using structure-based drug design, and parallel medicinal chemistry led to the identification of pyridine 12.


Subject(s)
Drug Design , Fructokinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Animals , Crystallography, X-Ray , Fructokinases/chemistry , Fructokinases/metabolism , Humans , Male , Molecular Docking Simulation , Pyridines/chemistry , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
8.
J Med Chem ; 60(13): 5673-5698, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28574706

ABSTRACT

Phosphodiesterase 2A (PDE2A) inhibitors have been reported to demonstrate in vivo activity in preclinical models of cognition. To more fully explore the biology of PDE2A inhibition, we sought to identify potent PDE2A inhibitors with improved brain penetration as compared to current literature compounds. Applying estimated human dose calculations while simultaneously leveraging synthetically enabled chemistry and structure-based drug design has resulted in a highly potent, selective, brain penetrant compound 71 (PF-05085727) that effects in vivo biochemical changes commensurate with PDE2A inhibition along with behavioral and electrophysiological reversal of the effects of NMDA antagonists in rodents. This data supports the ability of PDE2A inhibitors to potentiate NMDA signaling and their further development for clinical cognition indications.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Design , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dogs , Haplorhini , Humans , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/pharmacokinetics , Rats
9.
ACS Med Chem Lett ; 7(11): 999-1004, 2016 Nov 10.
Article in English | MEDLINE | ID: mdl-27882198

ABSTRACT

The tubulysin class of natural products has attracted much attention from the medicinal chemistry community due to its potent cytotoxicity against a wide range of human cancer cell lines, including significant activity in multidrug-resistant carcinoma models. As a result of their potency, the tubulysins have become an important tool for use in targeted therapy, being widely pursued as payloads in the development of novel small molecule drug conjugates (SMDCs) and antibody-drug conjugates (ADCs). A structure-based and parallel medicinal chemistry approach was applied to the synthesis of novel tubulysin analogues. These efforts led to the discovery of a number of novel and potent cytotoxic tubulysin analogues, providing a framework for our simultaneous report, which highlights the discovery of tubulysin-based ADCs, including use of site-specific conjugation to address in vivo stability of the C-11 acetate functionality.

10.
ACS Chem Biol ; 11(9): 2529-40, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27391855

ABSTRACT

Lysophospholipase-like 1 (LYPLAL1) is an uncharacterized metabolic serine hydrolase. Human genome-wide association studies link variants of the gene encoding this enzyme to fat distribution, waist-to-hip ratio, and nonalcoholic fatty liver disease. We describe the discovery of potent and selective covalent small-molecule inhibitors of LYPLAL1 and their use to investigate its role in hepatic metabolism. In hepatocytes, selective inhibition of LYPLAL1 increased glucose production supporting the inference that LYPLAL1 is a significant actor in hepatic metabolism. The results provide an example of how a selective chemical tool can contribute to evaluating a hypothetical target for therapeutic intervention, even in the absence of complete biochemical characterization.


Subject(s)
Hydrolases/metabolism , Lysophospholipase/antagonists & inhibitors , Serine/metabolism , Animals , Crystallization , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Lysophospholipase/chemistry
11.
Mol Pharmacol ; 89(5): 541-51, 2016 May.
Article in English | MEDLINE | ID: mdl-26912815

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are glutamate-gated ion channels that play key roles in brain physiology and pathology. Because numerous pathologic conditions involve NMDAR overactivation, subunit-selective antagonists hold strong therapeutic potential, although clinical successes remain limited. Among the most promising NMDAR-targeting drugs are allosteric inhibitors of GluN2B-containing receptors. Since the discovery of ifenprodil, a range of GluN2B-selective compounds with strikingly different structural motifs have been identified. This molecular diversity raises the possibility of distinct binding sites, although supporting data are lacking. Using X-ray crystallography, we show that EVT-101, a GluN2B antagonist structurally unrelated to the classic phenylethanolamine pharmacophore, binds at the same GluN1/GluN2B dimer interface as ifenprodil but adopts a remarkably different binding mode involving a distinct subcavity and receptor interactions. Mutagenesis experiments demonstrate that this novel binding site is physiologically relevant. Moreover, in silico docking unveils that GluN2B-selective antagonists broadly divide into two distinct classes according to binding pose. These data widen the allosteric and pharmacological landscape of NMDARs and offer a renewed structural framework for designing next-generation GluN2B antagonists with therapeutic value for brain disorders.


Subject(s)
Adrenergic alpha-Antagonists/metabolism , Drugs, Investigational/metabolism , Membrane Transport Modulators/metabolism , Models, Molecular , Receptors, N-Methyl-D-Aspartate/metabolism , Adrenergic alpha-Antagonists/chemistry , Adrenergic alpha-Antagonists/pharmacology , Allosteric Regulation/drug effects , Amino Acid Substitution , Animals , Binding Sites , Computational Biology , Drugs, Investigational/chemistry , Drugs, Investigational/pharmacology , Expert Systems , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacology , Ligands , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/pharmacology , Molecular Conformation , Molecular Docking Simulation , Mutation , Oxadiazoles/chemistry , Oxadiazoles/metabolism , Oxadiazoles/pharmacology , Piperidines/chemistry , Piperidines/metabolism , Piperidines/pharmacology , Protein Interaction Domains and Motifs , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/pharmacology , Pyrimidines/chemistry , Pyrimidines/metabolism , Pyrimidines/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
12.
J Med Chem ; 58(10): 4291-308, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25905800

ABSTRACT

A unique tetrahydrofuran ether class of highly potent α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor potentiators has been identified using rational and structure-based drug design. An acyclic lead compound, containing an ether-linked isopropylsulfonamide and biphenyl group, was pharmacologically augmented by converting it to a conformationally constrained tetrahydrofuran to improve key interactions with the human GluA2 ligand-binding domain. Subsequent replacement of the distal phenyl motif with 2-cyanothiophene to enhance its potency, selectivity, and metabolic stability afforded N-{(3S,4S)-4-[4-(5-cyano-2-thienyl)phenoxy]tetrahydrofuran-3-yl}propane-2-sulfonamide (PF-04958242, 3), whose preclinical characterization suggests an adequate therapeutic index, aided by low projected human oral pharmacokinetic variability, for clinical studies exploring its ability to attenuate cognitive deficits in patients with schizophrenia.


Subject(s)
Drug Evaluation, Preclinical/methods , Receptors, AMPA/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology , Administration, Oral , Adolescent , Adult , Aged , Animals , Binding Sites , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Discovery , Drug Stability , Female , Humans , Male , Memory, Short-Term/drug effects , Mice, Inbred C57BL , Middle Aged , Protein Conformation , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Structure-Activity Relationship , Sulfonamides/chemistry , Thiophenes/chemistry , Young Adult
13.
J Med Chem ; 58(7): 3223-52, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25781223

ABSTRACT

In recent years, the first generation of ß-secretase (BACE1) inhibitors advanced into clinical development for the treatment of Alzheimer's disease (AD). However, the alignment of drug-like properties and selectivity remains a major challenge. Herein, we describe the discovery of a novel class of potent, low clearance, CNS penetrant BACE1 inhibitors represented by thioamidine 5. Further profiling suggested that a high fraction of the metabolism (>95%) was due to CYP2D6, increasing the potential risk for victim-based drug-drug interactions (DDI) and variable exposure in the clinic due to the polymorphic nature of this enzyme. To guide future design, we solved crystal structures of CYP2D6 complexes with substrate 5 and its corresponding metabolic product pyrazole 6, which provided insight into the binding mode and movements between substrate/inhibitor complexes. Guided by the BACE1 and CYP2D6 crystal structures, we designed and synthesized analogues with reduced risk for DDI, central efficacy, and improved hERG therapeutic margins.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/chemistry , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , Cytochrome P-450 CYP2D6/chemistry , Drug Interactions , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Amino Acid Sequence , Amyloidogenic Proteins/metabolism , Animals , Crystallography, X-Ray , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Drug Design , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Inhibitory Concentration 50 , Male , Mice, Inbred Strains , Models, Molecular , Molecular Sequence Data , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacokinetics , Pyrazoles/chemistry , Structure-Activity Relationship
14.
Proc Natl Acad Sci U S A ; 112(12): E1414-22, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25775568

ABSTRACT

Phosphodiesterase 4 (PDE4) is an essential contributor to intracellular signaling and an important drug target. The four members of this enzyme family (PDE4A to -D) are functional dimers in which each subunit contains two upstream conserved regions (UCR), UCR1 and -2, which precede the C-terminal catalytic domain. Alternative promoters, transcriptional start sites, and mRNA splicing lead to the existence of over 25 variants of PDE4, broadly classified as long, short, and supershort forms. We report the X-ray crystal structure of long form PDE4B containing UCR1, UCR2, and the catalytic domain, crystallized as a dimer in which a disulfide bond cross-links cysteines engineered into UCR2 and the catalytic domain. Biochemical and mass spectrometric analyses showed that the UCR2-catalytic domain interaction occurs in trans, and established that this interaction regulates the catalytic activity of PDE4. By elucidating the key structural determinants of dimerization, we show that only long forms of PDE4 can be regulated by this mechanism. The results also provide a structural basis for the long-standing observation of high- and low-affinity binding sites for the prototypic inhibitor rolipram.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Protein Engineering/methods , Alternative Splicing , Catalytic Domain , Chromatography, Liquid , Codon , Crystallography, X-Ray , Cyclic AMP/metabolism , Dysostoses/enzymology , Gene Expression Regulation, Enzymologic , Genetic Variation , Humans , Intellectual Disability/enzymology , Mass Spectrometry , Models, Molecular , Mutation , Osteochondrodysplasias/enzymology , Phosphorylation , Protein Conformation , Protein Multimerization , Rolipram/chemistry , Signal Transduction , X-Ray Diffraction
15.
J Med Chem ; 57(24): 10527-43, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25431858

ABSTRACT

Auristatins, synthetic analogues of the antineoplastic natural product Dolastatin 10, are ultrapotent cytotoxic microtubule inhibitors that are clinically used as payloads in antibody-drug conjugates (ADCs). The design and synthesis of several new auristatin analogues with N-terminal modifications that include amino acids with α,α-disubstituted carbon atoms are described, including the discovery of our lead auristatin, PF-06380101. This modification of the peptide structure is unprecedented and led to analogues with excellent potencies in tumor cell proliferation assays and differential ADME properties when compared to other synthetic auristatin analogues that are used in the preparation of ADCs. In addition, auristatin cocrystal structures with tubulin are being presented that allow for the detailed examination of their binding modes. A surprising finding is that all analyzed analogues have a cis-configuration at the Val-Dil amide bond in their functionally relevant tubulin bound state, whereas in solution this bond is exclusively in the trans-configuration. This remarkable observation shines light onto the preferred binding mode of auristatins and serves as a valuable tool for structure-based drug design.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Depsipeptides/chemistry , Depsipeptides/pharmacology , Drug Discovery , Neoplasms/drug therapy , Animals , Area Under Curve , Cells, Cultured , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Models, Molecular , Molecular Structure , Protein Conformation , Rats , Rats, Wistar , Structure-Activity Relationship , Tandem Mass Spectrometry , Tubulin/metabolism
16.
J Med Chem ; 56(22): 9180-91, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24215237

ABSTRACT

Positive allosteric modulators ("potentiators") of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPAR) enhance excitatory neurotransmission and may improve the cognitive deficits associated with various neurological disorders. The dihydroisoxazole (DHI) series of AMPAR potentiators described herein originated from the identification of 7 by a high-throughput functional activity screen using mouse embryonic stem (mES) cell-derived neuronal precursors. Subsequent structure-based drug design using X-ray crystal structures of the ligand-binding domain of human GluA2 led to the discovery of both PF-04725379 (11), which in tritiated form became a novel ligand for characterizing the binding affinities of subsequent AMPAR potentiators in rat brain homogenate, and PF-04701475 (8a), a prototype used to explore AMPAR-mediated pharmacology in vivo. Lead series optimization provided 16a, a functionally potent compound lacking the potentially bioactivatable aniline within 8a, but retaining desirable in vitro ADME properties.


Subject(s)
Drug Discovery , Isoxazoles/chemistry , Isoxazoles/pharmacology , Receptors, AMPA/metabolism , Absorption , Allosteric Regulation/drug effects , Animals , High-Throughput Screening Assays , Humans , Isoxazoles/metabolism , Isoxazoles/pharmacokinetics , Male , Mice , Models, Molecular , Protein Structure, Tertiary , Rats , Receptors, AMPA/chemistry , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 23(11): 3443-7, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23597790

ABSTRACT

Selective phosphodiesterase 2 (PDE2) inhibitors are shown to have efficacy in a rat model of osteoarthritis (OA) pain. We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of phosphodiesterase 4 (PDE4) inhibitors, while minimizing PDE4 inhibitory activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like binding mode orthogonal to the cAMP-like binding mode found in PDE4. Extensive structure activity relationship studies ultimately led to identification of pyrazolodiazepinone, 22, which was >1000-fold selective for PDE2 over recombinant, full length PDEs 1B, 3A, 3B, 4A, 4B, 4C, 7A, 7B, 8A, 8B, 9, 10 and 11. Compound 22 also retained excellent PDE2 selectivity (241-fold to 419-fold) over the remaining recombinant, full length PDEs, 1A, 4D, 5, and 6. Compound 22 exhibited good pharmacokinetic properties and excellent oral bioavailability (F=78%, rat). In an in vivo rat model of OA pain, compound 22 had significant analgesic activity 1 and 3h after a single, 10 mg/kg, subcutaneous dose.


Subject(s)
Azepines/chemistry , Azirines/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Dihydropyridines/chemistry , Phosphodiesterase Inhibitors/chemistry , Pyrazoles/chemistry , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/therapeutic use , Animals , Azepines/pharmacokinetics , Azepines/therapeutic use , Azirines/pharmacokinetics , Azirines/therapeutic use , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dihydropyridines/pharmacokinetics , Dihydropyridines/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Half-Life , Osteoarthritis/drug therapy , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphodiesterase Inhibitors/therapeutic use , Protein Binding , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Rats , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 23(11): 3438-42, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23582272

ABSTRACT

We identified potent, selective PDE2 inhibitors by optimizing residual PDE2 activity in a series of PDE4 inhibitors, while simultaneously minimizing PDE4 activity. These newly designed PDE2 inhibitors bind to the PDE2 enzyme in a cGMP-like mode in contrast to the cAMP-like binding mode found in PDE4. Structure activity relationship studies coupled with an inhibitor bound crystal structure in the active site of the catalytic domain of PDE2 identified structural features required to minimize PDE4 inhibition while simultaneously maximizing PDE2 inhibition.


Subject(s)
Azirines/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Dihydropyridines/chemistry , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase Inhibitors/chemistry , Animals , Azirines/metabolism , Azirines/therapeutic use , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dihydropyridines/metabolism , Dihydropyridines/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical , Osteoarthritis/drug therapy , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/therapeutic use , Protein Binding , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 23(7): 1961-6, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23466229

ABSTRACT

The structure-based design, synthesis, and biological evaluation of a new pyrazole series of irreversible KAT II inhibitors are described herein. The modification of the inhibitor scaffold of 1 and 2 from a dihydroquinolinone core to a tetrahydropyrazolopyridinone core led to discovery of a new series of potent KAT II inhibitors with excellent physicochemical properties. Compound 20 is the most potent and lipophilically efficient of these new pyrazole analogs, with a k(inact)/K(i) value of 112,000 M(-1)s(-1) and lipophilic efficiency (LipE) of 8.53. The X-ray crystal structure of 20 with KAT II demonstrates key features that contribute to this remarkable potency and binding efficiency.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Pyrazoles/pharmacology , Transaminases/antagonists & inhibitors , Catalytic Domain/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Models, Molecular , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Transaminases/metabolism
20.
Curr Top Med Chem ; 13(1): 26-54, 2013.
Article in English | MEDLINE | ID: mdl-23409764

ABSTRACT

Schizophrenia is a highly debilitating mental disorder which afflicts approximately 1% of the global population. Cognitive and negative deficits account for the lifelong disability associated with schizophrenia, whose symptoms are not effectively addressed by current treatments. New medicines are needed to treat these aspects of the disease. Neurodevelopmental, neuropathological, genetic, and behavioral pharmacological data indicate that schizophrenia stems from a dysfunction of glutamate synaptic transmission, particularly in frontal cortical networks. A number of novel pre- and postsynaptic mechanisms affecting glutamatergic synaptic transmission have emerged as viable targets for schizophrenia. While developing orthosteric glutamatergic agents for these targets has proven extremely difficult, targeting allosteric sites of these targets has emerged as a promising alternative. From a medicinal chemistry perspective, allosteric sites provide an opportunity of finding agents with better drug-like properties and greater target specificity. Furthermore, allosteric modulators are better suited to maintaining the highly precise temporal and spatial aspects of glutamatergic synaptic transmission. Herein, we review neuropathological and genomic/genetic evidence underscoring the importance of glutamate synaptic dysfunction in the etiology of schizophrenia and make a case for allosteric targets for therapeutic intervention. We review progress in identifying allosteric modulators of AMPA receptors, NMDA receptors, and metabotropic glutamate receptors, all with the aim of restoring physiological glutamatergic synaptic transmission. Challenges remain given the complexity of schizophrenia and the difficulty in studying cognition in animals and humans. Nonetheless, important compounds have emerged from these efforts and promising preclinical and variable clinical validation has been achieved.


Subject(s)
Antipsychotic Agents/pharmacology , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/metabolism , Allosteric Regulation , Allosteric Site , Cognition/drug effects , Drug Design , Excitatory Amino Acid Agents/pharmacology , Humans , Ligands , Models, Molecular , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/physiopathology , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...