Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 352(2): 327-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25502803

ABSTRACT

Phosphodiesterase 10A (PDE10A) inhibitors have therapeutic potential for the treatment of psychiatric and neurologic disorders, such as schizophrenia and Huntington's disease. One of the key requirements for successful central nervous system drug development is to demonstrate target coverage of therapeutic candidates in brain for lead optimization in the drug discovery phase and for assisting dose selection in clinical development. Therefore, we identified AMG 580 [1-(4-(3-(4-(1H-benzo[d]imidazole-2-carbonyl)phenoxy)pyrazin-2-yl)piperidin-1-yl)-2-fluoropropan-1-one], a novel, selective small-molecule antagonist with subnanomolar affinity for rat, primate, and human PDE10A. We showed that AMG 580 is suitable as a tracer for lead optimization to determine target coverage by novel PDE10A inhibitors using triple-stage quadrupole liquid chromatography-tandem mass spectrometry technology. [(3)H]AMG 580 bound with high affinity in a specific and saturable manner to both striatal homogenates and brain slices from rats, baboons, and human in vitro. Moreover, [(18)F]AMG 580 demonstrated prominent uptake by positron emission tomography in rats, suggesting that radiolabeled AMG 580 may be suitable for further development as a noninvasive radiotracer for target coverage measurements in clinical studies. These results indicate that AMG 580 is a potential imaging biomarker for mapping PDE10A distribution and ensuring target coverage by therapeutic PDE10A inhibitors in clinical studies.


Subject(s)
Benzimidazoles/pharmacology , Brain/enzymology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Positron-Emission Tomography/methods , Pyrazines/pharmacology , Animals , Benzimidazoles/pharmacokinetics , Brain/diagnostic imaging , Chromatography, Liquid , Female , Fluorine Radioisotopes , Humans , Male , Mass Spectrometry , Molecular Structure , Papio , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacokinetics , Protein Binding , Pyrazines/pharmacokinetics , Radioligand Assay , Rats, Sprague-Dawley , Species Specificity , Stereoisomerism , Surface Plasmon Resonance , Tissue Distribution
2.
Nucl Med Biol ; 41(4): 343-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24607437

ABSTRACT

INTRODUCTION: Phosphodiesterase 10A (PDE10A) is an intracellular enzyme responsible for the breakdown of cyclic nucleotides which are important secondary messengers in the central nervous system. Inhibition of PDE10A has been identified as a potential therapeutic target for treatment of various neuropsychiatric disorders. To assist the drug development program, we have identified a selective PDE10A PET tracer, [(11)C]AMG 7980, for imaging PDE10A distribution using positron emission tomography. METHODS: [(11)C]AMG 7980 was prepared in a one-pot, two-step reaction. Dynamic PET scans were performed in non-human primates following a bolus or bolus plus constant infusion tracer injection paradigm. Regions-of-interest were defined on individuals' MRIs and transferred to the co-registered PET images. Data were analyzed using Logan graphical analysis with metabolite-corrected input function, the simplified reference tissue model (SRTM) method and occupancy plots. A benchmark PDE10A inhibitor was used to demonstrate PDE10A-specific binding. RESULTS: [(11)C]AMG 7980 was prepared with a mean specific activity of 99 ± 74 GBq/µmol (n=10) and a synthesis time of 45 min. Specific binding of the tracer was localized to the striatum and globus pallidus (GP) and low in other brain regions. Thalamus was used as the reference tissue to derive binding potentials (BPND). The BPND for caudate, putamen, and GP were 0.23, 0.65, 0.51, respectively by the graphical method, and 0.42, 0.76, and 0.75 from the SRTM method. A dose dependent decrease of BPND was observed with the pre-treatment of a PDE10A inhibitor. A bolus plus infusion injection paradigm yielded similar results. CONCLUSION: [(11)C]AMG 7980 has been successfully used for imaging PDE10A in non-human primate brain. Despite the fast brain kinetics it can be used to measure target occupancy of PDE10A inhibitors in non-human primates and potentially applicable to humans.


Subject(s)
Aminopyridines , Phosphoric Diester Hydrolases/metabolism , Positron-Emission Tomography/methods , Pyridazines , Aminopyridines/chemical synthesis , Aminopyridines/chemistry , Aminopyridines/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Feasibility Studies , Kinetics , Male , Papio , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridazines/metabolism , Radiochemistry
3.
Bioorg Med Chem Lett ; 23(23): 6447-54, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24139583

ABSTRACT

γ-Secretase modulators (GSMs) are potentially disease-modifying treatments for Alzheimer's disease. They selectively lower pathogenic Aß42 levels by shifting the enzyme cleavage sites without inhibiting γ-secretase activity, possibly avoiding known adverse effects observed with complete inhibition of the enzyme complex. A cell-based HTS effort identified the sulfonamide 1 as a GSM lead. Lead optimization studies identified compound 25 with improved cell potency, PKDM properties, and it lowered Aß42 levels in the cerebrospinal fluid (CSF) of Sprague-Dawley rats following oral administration. Further optimization of 25 to improve cellular potency is described.


Subject(s)
Alzheimer Disease/drug therapy , Amides/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Picolines/pharmacology , Alzheimer Disease/enzymology , Amides/chemistry , Animals , HEK293 Cells , Humans , Picolines/chemistry , Rats , Rats, Sprague-Dawley
4.
J Med Chem ; 56(21): 8781-92, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24102193

ABSTRACT

Our development of PDE10A inhibitors began with an HTS screening hit (1) that exhibited both high p-glycoprotein (P-gp) efflux ratios in rat and human and poor metabolic stability. On the basis of cocrystal structure of 1 in human PDE10A enzyme, we designed a novel keto-benzimidazole 26 with comparable PDE10A potency devoid of efflux liabilities. On target in vivo coverage of PDE10A in rat brain was assessed using our previously reported LC-MS/MS receptor occupancy (RO) technology. Compound 26 achieved 55% RO of PDE10A at 30 mg/kg po and covered PDE10A receptors in rat brain in a dose-dependent manner. Cocrystal structure of 26 in PDE10A confirmed the binding mode of the novel scaffold. Further optimization resulted in the identification of keto-benzimidazole 34, which showed an increased in vivo efficacy of 57% RO in rats at 10 mg/kg po and an improved in vivo rat clearance and oral bioavailability.


Subject(s)
Benzimidazoles/pharmacology , Drug Design , Ketones/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/chemical synthesis , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Ketones/administration & dosage , Ketones/chemical synthesis , Male , Models, Molecular , Molecular Structure , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/chemical synthesis , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Swine
6.
Bioorg Med Chem Lett ; 22(22): 6938-42, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23044369

ABSTRACT

We report our successful effort to increase the PDE3 selectivity of PDE10A inhibitor pyridyl cinnoline 1 using a combination of computational modeling and structural-activity relationship investigations. An analysis of the PDE3 catalytic domain compared to the co-crystal structure of cinnoline analog 1 in PDE10A revealed two areas of structural differences in the active sites and suggested areas on the scaffold that could be modified to exploit those unique structural features. Once SAR established the cinnoline as the optimal scaffold, modifications on the methoxy groups of the cinnoline and the methyl group on the pyridine led to the discovery of compounds 33 and 36. Both compounds achieved significant improvement in selectivity against PDE3 while maintaining their PDE10A inhibitory activity and in vivo metabolic stability comparable to 1.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry , Heterocyclic Compounds, 2-Ring/chemistry , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Pyridines/chemistry , Animals , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Drug Design , Half-Life , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/pharmacokinetics , Phosphoric Diester Hydrolases/metabolism , Pyridines/chemical synthesis , Pyridines/pharmacokinetics , Rats , Structure-Activity Relationship
7.
J Med Chem ; 55(10): 4776-87, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22548439

ABSTRACT

A radiolabeled tracer for imaging therapeutic targets in the brain is a valuable tool for lead optimization in CNS drug discovery and for dose selection in clinical development. We report the rapid identification of a novel phosphodiesterase 10A (PDE10A) tracer candidate using a LC-MS/MS technology. This structurally distinct PDE10A tracer, AMG-7980 (5), has been shown to have good uptake in the striatum (1.2% ID/g tissue), high specificity (striatum/thalamus ratio of 10), and saturable binding in vivo. The PDE10A affinity (K(D)) and PDE10A target density (B(max)) were determined to be 0.94 nM and 2.3 pmol/mg protein, respectively, using [(3)H]5 on rat striatum homogenate. Autoradiography on rat brain sections indicated that the tracer signal was consistent with known PDE10A expression pattern. The specific binding of [(3)H]5 to rat brain was blocked by another structurally distinct, published PDE10A inhibitor, MP-10. Lastly, our tracer was used to measure in vivo PDE10A target occupancy of a PDE10A inhibitor in rats using LC-MS/MS technology.


Subject(s)
Aminopyridines/chemical synthesis , Phosphoric Diester Hydrolases/metabolism , Pyridazines/chemical synthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aminopyridines/chemistry , Aminopyridines/pharmacokinetics , Animals , Brain/diagnostic imaging , Brain/enzymology , Cell Line , Chromatography, Liquid , Dogs , Humans , In Vitro Techniques , Male , Permeability , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacokinetics , Protein Binding , Pyrazoles/pharmacokinetics , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Quinolines/pharmacokinetics , Radionuclide Imaging , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Stereoisomerism , Surface Plasmon Resonance , Tandem Mass Spectrometry , Tissue Distribution , Tritium
8.
Bioorg Med Chem Lett ; 22(6): 2262-5, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22365755

ABSTRACT

We report the discovery of 6,7-dimethoxy-4-(pyridin-3-yl)cinnolines as novel inhibitors of phosphodiesterase 10A (PDE10A). Systematic examination and analyses of structure-activity-relationships resulted in single digit nM potency against PDE10A. X-ray co-crystal structure revealed the mode of binding in the enzyme's catalytic domain and the source of selectivity against other PDEs. High in vivo clearance in rats was addressed with the help of metabolite identification (ID) studies. These findings combined resulted in compound 39, a promising potent inhibitor of PDE10A with good in vivo metabolic stability in rats and efficacy in a rodent behavioral model.


Subject(s)
Coumarins/chemical synthesis , Phosphodiesterase Inhibitors/chemical synthesis , Phosphoric Diester Hydrolases/metabolism , Psychotropic Drugs/chemical synthesis , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Binding Sites , Coumarins/pharmacology , Drug Discovery , Humans , Models, Molecular , Phosphodiesterase Inhibitors/administration & dosage , Protein Binding , Psychotropic Drugs/administration & dosage , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 18(18): 5115-7, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18723346

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Kit is associated with an increasing number of human diseases, including certain cancers and mast cell diseases. Interference of c-Kit signaling with multi-kinase inhibitors has been shown clinically to successfully treat gastrointestinal stromal tumors and mastocytosis. Targeted therapy of c-Kit activity may provide therapeutic advantages against off-target effects for non-oncology applications. A new structural class of c-Kit inhibitors is described, including in vitro c-Kit potency, kinase selectivity, and the observed binding mode.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Isoxazoles/chemical synthesis , Isoxazoles/pharmacology , Proto-Oncogene Proteins c-kit/drug effects , Proto-Oncogene Proteins c-kit/metabolism , Amides/chemistry , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Humans , Isoxazoles/chemistry , Molecular Conformation , Molecular Structure , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...