Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Publication year range
1.
Bioorg Med Chem Lett ; 25(17): 3488-94, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26212776

ABSTRACT

Synthesis and SAR studies of novel triazolobenzazepinones as gamma secretase modulators (GSMs) are presented in this communication. Starting from our azepinone leads, optimization studies toward improving central lowering of Aß42 led to the discovery of novel benzo-fused azepinones. Several benzazepinones were profiled in vivo and found to lower brain Aß42 levels in Sprague Dawley rats and transgenic APP-YAC mice in a dose-dependent manner after a single oral dose. Compound 34 was further progressed into a pilot study in our cisterna-magna-ported rhesus monkey model, where we observed robust lowering of CSF Aß42 levels.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Animals , Drug Discovery , Macaca mulatta , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley
2.
Bioorg Med Chem Lett ; 25(21): 4812-4819, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26195137

ABSTRACT

The IC50 of a beta-secretase (BACE-1) lead compound was improved ∼200-fold from 11 µM to 55 nM through the addition of a single methyl group. Computational chemistry, small molecule NMR, and protein crystallography capabilities were used to compare the solution conformation of the ligand under varying pH conditions to its conformation when bound in the active site. Chemical modification then explored available binding pockets adjacent to the ligand. A strategically placed methyl group not only maintained the required pKa of the piperidine nitrogen and filled a small hydrophobic pocket, but more importantly, stabilized the conformation best suited for optimized binding to the receptor.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Hydantoins/chemistry , Hydantoins/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Hydantoins/chemical synthesis , Methylation , Models, Molecular , Molecular Structure , Structure-Activity Relationship
3.
Neuroimage ; 68: 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23238431

ABSTRACT

Antagonism of the central opioid receptor like-1 receptor (ORL1) has been implicated in cognition, and has been a focus of drug discovery efforts to ameliorate the cognitive deficits that remain during the stable treatment of schizophrenia with current antipsychotics. In order to facilitate dose selection for phase II clinical testing an ORL1-specific PET tracer was developed to determine drug plasma concentration versus occupancy relationships in order to ensure that the doses selected and the degree of target engagement were sufficient to ensure adequate proof of concept testing. MK-0911 is a selective, high affinity antagonist for the ORL1 receptor radiolabeled with high specific activity (18)F for positron emission tomography (PET) studies. Evaluation of [(18)F]MK-0911 in rhesus monkey PET studies showed a pattern of brain uptake which was consistent with the known distribution of ORL1. In vitro autoradiography with [(18)F]MK-0911 in rhesus monkey and human brain tissue slices showed a regional distribution that was consistent with in vivo imaging results in monkey. Pre-treatment of rhesus monkeys with high doses of structurally diverse ORL1 antagonists MK-0584, MK-0337, or MK-5757 achieved blockade of [(18)F]MK-0911 in all gray matter regions. Baseline PET studies with [(18)F]MK-0911 in healthy human subjects showed tracer distribution and kinetics similar to that observed in rhesus monkey. Quantification of [(18)F]MK-0911 uptake in repeat human baseline PET studies showed a test-retest variability in volume of distribution (V(T)) averaging 3% across brain regions. Humans dosed orally with MK-5757 showed reduced [(18)F]MK-0911 tracer concentration in brain proportional with MK-5757 dose and plasma level. [(18)F]MK-0911 was useful for determining MK-5757-induced receptor occupancy of ORL1 to guide MK-5757 dose-selection for clinical proof-of-concept studies. Additionally, [(18)F]MK-0911 may be a useful tool for studying the pharmacology of ORL1 in various human populations and disease states.


Subject(s)
Benzimidazoles/pharmacokinetics , Brain/diagnostic imaging , Fluorine Radioisotopes/pharmacokinetics , Piperidines/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, Opioid/metabolism , Adult , Animals , Benzimidazoles/chemistry , Brain/metabolism , Fluorine Radioisotopes/chemistry , Humans , Macaca mulatta , Male , Middle Aged , Piperidines/chemistry , Radiopharmaceuticals/chemistry , Tissue Distribution , Young Adult , Nociceptin Receptor
4.
J Pharmacol Exp Ther ; 347(2): 478-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23975906

ABSTRACT

Calcitonin gene-related peptide (CGRP) is a potent neuropeptide whose agonist interaction with the CGRP receptor (CGRP-R) in the periphery promotes vasodilation, neurogenic inflammation and trigeminovascular sensory activation. This process is implicated in the cause of migraine headaches, and CGRP-R antagonists in clinical development have proven effective in treating migraine-related pain in humans. CGRP-R is expressed on blood vessel smooth muscle and sensory trigeminal neurons and fibers in the periphery as well as in the central nervous system. However, it is not clear what role the inhibition of central CGRP-R plays in migraine pain relief. To this end, the CGRP-R positron emission tomography (PET) tracer [(11)C]MK-4232 (2-[(8R)-8-(3,5-difluorophenyl)-6,8-[6-(11)C]dimethyl-10-oxo-6,9-diazaspiro[4.5]decan-9-yl]-N-[(2R)-2'-oxospiro[1,3-dihydroindene-2,3'-1H-pyrrolo[2,3-b]pyridine]-5-yl]acetamide) was discovered and developed for use in clinical PET studies. In rhesus monkeys and humans, [(11)C]MK-4232 displayed rapid brain uptake and a regional brain distribution consistent with the known distribution of CGRP-R. Monkey PET studies with [(11)C]MK-4232 after intravenous dosing with CGRP-R antagonists validated the ability of [(11)C]MK-4232 to detect changes in CGRP-R occupancy in proportion to drug plasma concentration. Application of [(11)C]MK-4232 in human PET studies revealed that telcagepant achieved only low receptor occupancy at an efficacious dose (140 mg PO). Therefore, it is unlikely that antagonism of central CGRP-R is required for migraine efficacy. However, it is not known whether high central CGRP-R antagonism may provide additional therapeutic benefit.


Subject(s)
Acetanilides/pharmacokinetics , Analgesics/pharmacokinetics , Azepines/pharmacokinetics , Brain/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists , Imidazoles/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Spiro Compounds/pharmacokinetics , Acetanilides/chemistry , Adult , Analgesics/therapeutic use , Animals , Azepines/therapeutic use , Brain/diagnostic imaging , Carbon Radioisotopes , Female , Humans , Imidazoles/therapeutic use , Macaca mulatta , Male , Middle Aged , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Molecular Structure , Protein Binding , Radiopharmaceuticals/chemistry , Species Specificity , Spiro Compounds/chemistry , Tissue Distribution , Young Adult
5.
Neuroimage ; 59(3): 2589-99, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-21930214

ABSTRACT

BACKGROUND: Glycine transporter 1 (GlyT1) inhibitors have emerged as potential treatments for schizophrenia due to their potentiation of NMDA receptor activity by modulating the local concentrations of the NMDA co-agonist glycine. [18F]MK-6577 is a potent and selective GlyT1 inhibitor PET tracer. Although differences in ligand kinetics can be expected between non-human primates and humans, the tracer pre-clinical evaluation can provide valuable information supporting protocol design and quantification in the clinical space. The main objective of this work was to evaluate the in vivo kinetics of [18F]MK-6577 in rhesus monkey brain. Additionally, a method for estimating the tracer input function from the tracer brain tissue kinetics and venous sampling was validated. This technique was applied for determination of the dose-occupancy relationship of a GlyT1 inhibitor in monkey brain. METHODS: Compartmental and Logan graphical analysis were utilized for quantification of the [18F]MK-6577 binding using the measured tracer arterial input function. The stability of the tracer volume of distribution relative to scan length was assessed. The proposed model-based input function method takes advantage of the agreement between the tracer concentration in arterial and venous plasma from ~5 min. The approach estimates the initial peak of the input curve by adding a gamma like function term to the measured venous curve. The parameters of the model function were estimated by simultaneously fitting several brain time activity curves to a compartmental model. RESULTS: Good agreement was found between the model-based and the measured arterial plasma curve and the corresponding distribution volumes. The Logan analysis was the preferred method of analysis providing reliable and stable volume of distribution and occupancy results using a 90 and possibly 60 min scan length. CONCLUSION: The model-based input function method and Logan analysis are well suited for quantification of [18F]MK-6577 binding and GlyT1 occupancy in monkey brain.


Subject(s)
Brain Chemistry/physiology , Brain/diagnostic imaging , Excitatory Amino Acid Agonists , Glycine Plasma Membrane Transport Proteins/metabolism , Algorithms , Animals , Corpus Striatum/diagnostic imaging , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacokinetics , Fluorine Radioisotopes , Glycine Plasma Membrane Transport Proteins/analysis , Image Processing, Computer-Assisted , Macaca mulatta , Models, Neurological , Models, Statistical , Positron-Emission Tomography , Receptors, N-Methyl-D-Aspartate/physiology , Thalamus/diagnostic imaging
6.
J Neurosci Res ; 90(12): 2247-58, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22987781

ABSTRACT

Reduction in cerebrospinal fluid (CSF) amyloid ß42 (Aß42) and elevation in total tau and phospho-thr181 tau consistently differentiate between Alzheimer's disease (AD) and age-matched control subjects. In contrast, CSF ß-site APP-cleaving enzyme activity (BACE1) and soluble amyloid precursor proteins α and ß (sAPPα and sAPPß) are without consistent patterns in AD subjects. Plasma sampling is much easier, with fewer side effects, and is readily applied in primary care centers, so we have developed and validated novel plasma BACE activity, sAPPß, and sAPPα assays and investigated their ability to distinguish AD from age-matched controls. Plasma BACE activity assay was sensitive and specific, with signal being immunodepleted with a specific BACE1 antibody and inhibited with a BACE1-specific inhibitor. Plasma sAPPß and sAPPα assays were specific, with signal diluting linearly, immunodepleted with specific antibodies, and at background levels in APP knockout mice. In rhesus monkeys, BACE1 but not γ-secretase inhibitor led to significant lowering of plasma sAPPß with concurrent elevation of plasma sAPPα. AD subjects showed a significant increase in plasma BACE1 activity, sAPPß, sAPPα, and Aß42 (P < 0.001) compared with age-matched controls. In conclusion, plasma BACE activity and sAPP endpoints provide novel investigative biomarkers for AD diagnosis and potential pharmacodynamic biomarkers for secretase inhibitor studies.


Subject(s)
Alzheimer Disease/blood , Amyloid Precursor Protein Secretases/blood , Amyloid beta-Peptides/blood , Amyloid beta-Protein Precursor/blood , Aspartic Acid Endopeptidases/blood , Peptide Fragments/blood , Aged , Alzheimer Disease/diagnosis , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/deficiency , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/deficiency , Biomarkers , Female , Humans , Immunohistochemistry/methods , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Sensitivity and Specificity , Sulfonamides/pharmacology
7.
J Neurosci ; 30(19): 6743-50, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463236

ABSTRACT

The accumulation of amyloid beta (Abeta) in Alzheimer's disease is caused by an imbalance of production and clearance, which leads to increased soluble Abeta species and extracellular plaque formation in the brain. Multiple Abeta-lowering therapies are currently in development: an important goal is to characterize the molecular mechanisms of action and effects on physiological processing of Abeta, as well as other amyloid precursor protein (APP) metabolites, in models which approximate human Abeta physiology. To this end, we report the translation of the human in vivo stable-isotope-labeling kinetics (SILK) method to a rhesus monkey cisterna magna ported (CMP) nonhuman primate model, and use the model to test the mechanisms of action of a gamma-secretase inhibitor (GSI). A major concern of inhibiting the enzymes which produce Abeta (beta- and gamma-secretase) is that precursors of Abeta may accumulate and cause a rapid increase in Abeta production when enzyme inhibition discontinues. In this study, the GSI MK-0752 was administered to conscious CMP rhesus monkeys in conjunction with in vivo stable-isotope-labeling, and dose-dependently reduced newly generated CNS Abeta. In contrast to systemic Abeta metabolism, CNS Abeta production was not increased after the GSI was cleared. These results indicate that most of the CNS APP was metabolized to products other than Abeta, including C-terminal truncated forms of Abeta: 1-14, 1-15 and 1-16; this demonstrates an alternative degradation pathway for CNS amyloid precursor protein during gamma-secretase inhibition.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Spinal Cord/metabolism , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/cerebrospinal fluid , Animals , Brain/enzymology , Carbon Radioisotopes , Cross-Over Studies , Humans , Isotope Labeling/methods , Kinetics , Macaca mulatta , Male , Models, Animal , Species Specificity , Spinal Cord/enzymology , Time Factors
8.
Neuroimage ; 54(4): 2635-42, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21078401

ABSTRACT

Neuropeptide Y receptor subtype 1 (NPY Y1) has been implicated in appetite regulation, and antagonists of NPY Y1 are being explored as potential therapeutics for obesity. An NPY Y1 PET tracer is useful for determining the level of target engagement by NPY Y1 antagonists in preclinical and clinical studies. Here we report the synthesis and evaluation of [(18)F]Y1-973, a novel PET tracer for NPY Y1. [(18)F]Y1-973 was radiolabeled by reaction of a primary chloride with [(18)F]KF/K2.2.2 followed by deprotection with HCl. [(18)F]Y1-973 was produced with high radiochemical purity (>98%) and high specific activity (>1000 Ci/mmol). PET studies in rhesus monkey brain showed that the distribution of [(18)F]Y1-973 was consistent with the known NPY Y1 distribution; uptake was highest in the striatum and cortical regions and lowest in the pons, cerebellum nuclei, and brain stem. Blockade of [(18)F]Y1-973 uptake with NPY Y1 antagonist Y1-718 revealed a specific signal that was dose-dependently reduced in all regions of grey matter to a similarly low level of tracer uptake, indicative of an NPY Y1 specific signal. In vitro autoradiographic studies with [(18)F]Y1-973 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the in vivo PET studies. Highest binding density was observed in the dentate gyrus, caudate-putamen, and cortical regions; moderate binding density in the hypothalamus and thalamus; and lowest binding density in the globus pallidus and cerebellum. In vitro saturation binding studies in rhesus monkey and human caudate-putamen homogenates confirmed a similarly high B(max)/K(d) ratio for [(18)F]Y1-973, suggesting the tracer may provide a specific signal in human brain of similar magnitude to that observed in rhesus monkey. [(18)F]Y1-973 is a suitable PET tracer for imaging NPY Y1 in rhesus monkey with potential for translation to human PET studies.


Subject(s)
Brain/diagnostic imaging , Fluorine Radioisotopes/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Receptors, Neuropeptide Y/biosynthesis , Animals , Autoradiography , Humans , Macaca mulatta , Positron-Emission Tomography , Radioactive Tracers
9.
J Pharmacol Exp Ther ; 338(2): 568-78, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21531794

ABSTRACT

Genetic studies have linked both osteoporotic and high bone mass phenotypes to low-density lipoprotein receptor-related proteins (LRP4, LRP5, and LRP6). LRPs are receptors for inhibitory Dickkopf-1 (DKK1) protein, and treatment modalities that modulate LRP/DKK1 binding therefore may act as stimulators of bone mass accrual. Here, we report that RH2-18, a fully human monoclonal anti-DKK1 antibody elicits systemic pharmacologic bone efficacy and new bone formation at endosteal bone surfaces in vivo in a mouse model of estrogen-deficiency-induced osteopenia. This was paralleled by partial-to-complete resolution of osteopenia (bone mineral density) at all of the skeletal sites investigated in femur and lumbar-vertebral bodies and the restoration of trabecular bone microarchitecture. More importantly, testing of RH2-18 in adult, osteopenic rhesus macaques demonstrated a rate-limiting role of DKK1 at multiple skeletal sites and responsiveness to treatment. In conclusion, this study provides pharmacologic evidence for the modulation of DKK1 bioactivity in the adult osteopenic skeleton as a viable approach to resolve osteopenia in animal models. Thus, data described here suggest that targeting DKK1 through means such as a fully human anti-DKK1-antibody provides a potential bone-anabolic treatment for postmenopausal osteoporosis.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Intercellular Signaling Peptides and Proteins/physiology , Osteogenesis/immunology , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/therapy , Animals , Bone Density/immunology , CHO Cells , Cricetinae , Cricetulus , Female , HEK293 Cells , Humans , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Osteoporosis, Postmenopausal/pathology
10.
Synapse ; 65(2): 125-35, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20524178

ABSTRACT

Two moderately lipophilic, high affinity ligands for metabotropic glutamate receptor subtype 1 (mGluR1) were radiolabeled with a positron-emitting radioisotope and evaluated in rhesus monkey as potential PET tracers. Both ligands were radiolabeled with fluorine-18 via nucleophilic displacement of the corresponding 2-chloropyridine precursor with [¹8F]potassium fluoride. [¹8F]MK-1312 was found to have a suitable signal for quantification of mGluR1 receptors in nonhuman primates and was more thoroughly characterized. In vitro autoradiographic studies with [¹8F]MK-1312 in rhesus monkey and human brain tissue slices revealed an uptake distribution consistent with the known distribution of mGluR1, with the highest uptake in the cerebellum, moderate uptake in the hippocampus, thalamus, and cortical regions, and lowest uptake in the caudate and putamen. In vitro saturation binding studies in rhesus monkey and human cerebellum homogenates confirmed that [¹8F]MK-1312 binds to a single site with a B(max) /K(d) ratio of 132 and 98, respectively. PET studies in rhesus monkey with [¹8F]MK-1312 showed high brain uptake and a regional distribution consistent with in vitro autoradiography results. Blockade of [¹8F]MK-1312 uptake with mGluR1 allosteric antagonist MK-5435 dose-dependently reduced tracer uptake in all regions of gray matter to a similarly low level of tracer uptake. This revealed a large specific signal useful for determination of mGluR1 receptor occupancy in rhesus monkey. Taken together, these results are promising for clinical PET studies with [¹8F]MK-1312 to determine mGluR1 occupancy of MK-5435.


Subject(s)
Brain/drug effects , Brain/diagnostic imaging , Excitatory Amino Acid Agents , Positron-Emission Tomography , Receptors, Metabotropic Glutamate/metabolism , Animals , Autoradiography/methods , Binding Sites/drug effects , Brain/metabolism , Brain Mapping , Dose-Response Relationship, Drug , Excitatory Amino Acid Agents/chemical synthesis , Excitatory Amino Acid Agents/chemistry , Excitatory Amino Acid Agents/pharmacokinetics , Fluorine Radioisotopes/chemistry , Fluorine Radioisotopes/pharmacokinetics , Humans , Indoles/chemical synthesis , Indoles/pharmacokinetics , Inhibitory Concentration 50 , Ligands , Macaca mulatta , Tissue Distribution , Triazoles/chemical synthesis , Triazoles/pharmacokinetics
11.
Synapse ; 65(4): 261-70, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20687108

ABSTRACT

Two positron emission tomography radiotracers for the glycine transporter 1 (GlyT1) are reported here. Each radiotracer is a propylsulfonamide-containing benzamide and was labeled with either carbon-11 or fluorine-18. [¹¹C]CMPyPB was synthesized by the alkylation of a 3-hydroxypyridine precursor using [¹¹C]MeI, and [¹8F]MK-6577 was synthesized by a nucleophilic aromatic substitution reaction using a 2-chloropyridine precursor. Each tracer shows good uptake into rhesus monkey brain with the expected distribution of highest uptake in the pons, thalamus, and cerebellum and lower uptake in the striatum and gray matter of the frontal cortex. In vivo blockade and chase studies of [¹8F]MK-6577 showed a large specific signal and reversible binding. In vitro autoradiographic studies with [¹8F]MK-6577 showed a large specific signal in both rhesus monkey and human brain slices and a distribution consistent with the in vivo results and those reported in the literature. In vivo metabolism studies in rhesus monkeys demonstrated that only more-polar metabolites are formed for each tracer. Of these two tracers, [¹8F]MK-6577 was more extensively characterized and is a promising clinical positron emission tomography tracer for imaging GlyT1 and for measuring GlyT1 occupancy of therapeutic compounds.


Subject(s)
Benzamides/chemical synthesis , Carbon Radioisotopes , Fluorine Radioisotopes , Glycine Plasma Membrane Transport Proteins/blood , Positron-Emission Tomography/methods , Pyridines/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Benzamides/blood , Carbon Radioisotopes/blood , Cell Line , Drug Evaluation, Preclinical/methods , Fluorine Radioisotopes/blood , Glycine Plasma Membrane Transport Proteins/metabolism , Humans , Macaca mulatta , Pyridines/blood , Sulfonamides/blood
12.
Bioorg Med Chem Lett ; 21(5): 1532-5, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21295466

ABSTRACT

Although the S3 pocket of the thrombin active site is lined with lipophilic amino acid residues, the accommodation of polarity within the lipophilic P3 moiety of small molecule inhibitors is possible provided that the polar functionality is capable of pointing away from the binding pocket outwards toward solvent while simultaneously allowing the lipophilic portion of the P3 ligand to interact with the S3 amino acid residues. Manipulation of this motif provided the means to effect optimization of functional potency, in vivo antithrombotic efficacy and oral bioavailability in a series of 3-aminopyrazinone thrombin inhibitors which contained non-charged groups at the P1 position.


Subject(s)
Anticoagulants/chemical synthesis , Blood Coagulation Disorders/drug therapy , Drug Design , Pyrazines/chemical synthesis , Thrombin/antagonists & inhibitors , Administration, Oral , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacology , Binding Sites , Biological Availability , Dogs , Molecular Structure , Pyrazines/chemistry , Pyrazines/pharmacology , Rats , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 21(5): 1536-40, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21295467

ABSTRACT

A novel 1,3,5-trisubstituted benzamide thrombin inhibitor template was designed via hybridization of a known aminopyridinoneacetamide and a known 1,3,5-trisubstituted phenyl ether. Optimization of this lead afforded a novel potent series of biaryl 1,3,5-trisubstituted benzenes with excellent functional anticoagulant potency.


Subject(s)
Antithrombins/chemical synthesis , Benzene/chemical synthesis , Drug Design , Thrombin/antagonists & inhibitors , Antithrombins/chemistry , Antithrombins/pharmacology , Benzene/chemistry , Benzene/pharmacology , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship
14.
Bull Math Biol ; 73(1): 230-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20411345

ABSTRACT

Aggregation of the small peptide amyloid beta (Aß) into oligomers and fibrils in the brain is believed to be a precursor to Alzheimer's disease. Aß is produced via multiple proteolytic cleavages of amyloid precursor protein (APP), mediated by the enzymes ß- and γ-secretase. In this study, we examine the temporal dynamics of soluble (unaggregated) Aß in the plasma and cerebral-spinal fluid (CSF) of rhesus monkeys treated with different oral doses of a γ-secretase inhibitor. A dose-dependent reduction of Aß concentration was observed within hours of drug ingestion, for all doses tested. Aß concentration in the CSF returned to its predrug level over the monitoring period. In contrast, Aß concentration in the plasma exhibited an unexpected overshoot to as high as 200% of the predrug concentration, and this overshoot persisted as late as 72 hours post-drug ingestion. To account for these observations, we proposed and analyzed a minimal physiological model for Aß dynamics that could fit the data. Our analysis suggests that the overshoot arises from the attenuation of an Aß clearance mechanism, possibly due to the inhibitor. Our model predicts that the efficacy of Aß clearance recovers to its basal (pretreatment) value with a characteristic time of >48 hours, matching the time-scale of the overshoot. These results point to the need for a more detailed investigation of soluble Aß clearance mechanisms and their interaction with Aß-reducing drugs.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Models, Biological , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Macaca mulatta , Mathematical Concepts , Models, Animal , Solubility
15.
J Proteome Res ; 9(3): 1392-401, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20095649

ABSTRACT

The rapid identification of protein biomarkers in biofluids is important to drug discovery and development. Here, we describe a general proteomic approach for the discovery and identification of proteins that exhibit a statistically significant difference in abundance in cerebrospinal fluid (CSF) before and after pharmacological intervention. This approach, differential mass spectrometry (dMS), is based on the analysis of full scan mass spectrometry data. The dMS workflow does not require complex mixing and pooling strategies, or isotope labeling techniques. Accordingly, clinical samples can be analyzed individually, allowing the use of longitudinal designs and within-subject data analysis in which each subject acts as its own control. As a proof of concept, we performed multifactorial dMS analyses on CSF samples drawn at 6 time points from n = 6 cisterna magna ported (CMP) rhesus monkeys treated with 2 potent gamma secretase inhibitors (GSI) or comparable vehicle in a 3-way crossover study that included a total of 108 individual CSF samples. Using analysis of variance and statistical filtering on the aligned and normalized LC-MS data sets, we detected 26 features that were significantly altered in CSF by drug treatment. Of those 26 features, which belong to 10 distinct isotopic distributions, 20 were identified by MS/MS as 7 peptides from CD99, a cell surface protein. Six features from the remaining 3 isotopic distributions were not identified. A subsequent analysis showed that the relative abundance of these 26 features showed the same temporal profile as the ELISA measured levels of CSF A beta 42 peptide, a known pharmacodynamic marker for gamma-secretase inhibition. These data demonstrate that dMS is a promising approach for the discovery, quantification, and identification of candidate target engagement biomarkers in CSF.


Subject(s)
Cerebrospinal Fluid Proteins/analysis , Mass Spectrometry/methods , Proteomics/methods , Algorithms , Amino Acid Sequence , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Area Under Curve , Biomarkers/cerebrospinal fluid , Cerebrospinal Fluid Proteins/metabolism , Macaca mulatta , Molecular Sequence Data , Oligopeptides/pharmacokinetics , Peptide Fragments/analysis , Peptide Fragments/metabolism
16.
J Pharmacol Exp Ther ; 333(1): 152-60, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20065019

ABSTRACT

Calcitonin gene-related peptide (CGRP) has long been hypothesized to play a key role in migraine pathophysiology, and the advent of small-molecule antagonists has clearly demonstrated a clinical link between blocking the CGRP receptor and migraine efficacy. 2-[(8R)-8-(3,5-Difluorophenyl)-10-oxo-6,9-diazaspiro[4.5]dec-9-yl]-N-[(2R)-2'-oxo-1,1',2',3-tetrahydrospiro[indene-2,3'-pyrrolo[2,3-b]pyridin]-5-yl]acetamide (MK-3207) represents the third CGRP receptor antagonist to display clinical efficacy in migraine trials. Here, we report the pharmacological characterization of MK-3207, a potent and orally bioavailable CGRP receptor antagonist. In vitro, MK-3207 is a potent antagonist of the human and rhesus monkey CGRP receptors (K(i) = 0.024 nM). In common with other CGRP receptor antagonists, MK-3207 displays lower affinity for CGRP receptors from other species, including canine and rodent. As a consequence of species selectivity, the in vivo potency was assessed in a rhesus monkey pharmacodynamic assay measuring capsaicin-induced changes in forearm dermal blood flow via laser Doppler imaging. MK-3207 produced a concentration-dependent inhibition of dermal vasodilation, with plasma concentrations of 0.8 and 7 nM required to block 50 and 90% of the blood flow increase, respectively. The tritiated analog [3H]MK-3207 was used to study the binding characteristics on the human CGRP receptor. [3H]MK-3207 displayed reversible and saturable binding (K(D) = 0.06 nM), and the off-rate was determined to be 0.012 min(-1), with a t(1/2) value of 59 min. In vitro autoradiography studies on rhesus monkey brain slices identified the highest level of binding in the cerebellum, brainstem, and meninges. Finally, as an index of central nervous system penetrability, the in vivo cerebrospinal fluid/plasma ratio was determined to be 2 to 3% in cisterna magna-ported rhesus monkeys.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Spiro Compounds/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Autoradiography , Binding, Competitive , Biological Transport , Brain/metabolism , Bridged Bicyclo Compounds, Heterocyclic/blood , Bridged Bicyclo Compounds, Heterocyclic/cerebrospinal fluid , Cell Line , Chlorocebus aethiops , Female , Humans , Kinetics , Macaca mulatta , Male , Mice , Radioligand Assay , Receptors, Adrenomedullin , Receptors, Calcitonin/metabolism , Receptors, Calcitonin Gene-Related Peptide/physiology , Receptors, Islet Amyloid Polypeptide , Receptors, Peptide/metabolism , Spiro Compounds/blood , Spiro Compounds/cerebrospinal fluid , Vasodilation/drug effects
17.
Bioorg Med Chem Lett ; 20(5): 1779-82, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20122828

ABSTRACT

This Letter describes the one pot synthesis of tertiary carbinamine 3 and related analogs of brain penetrant BACE-1 inhibitors via the alkylation of the Schiff base intermediate 2. The methodology developed for this study provided a convenient and rapid means to explore the P1 region of these types of inhibitors, where the P1 group is installed in the final step using a one-pot two-step protocol. Further SAR studies led to the identification of 10 which is twofold more potent in vitro as compared to the lead compound. This inhibitor was characterized in a cisterna magna ported rhesus monkey model, where significant lowering of CSF Abeta40 was observed.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/enzymology , Enzyme Inhibitors/chemistry , Oxadiazoles/chemistry , Sulfonamides/chemistry , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Humans , Macaca mulatta , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacokinetics , Peptide Fragments/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacokinetics
18.
J Pharmacol Exp Ther ; 328(1): 131-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18854490

ABSTRACT

beta-Site amyloid precursor protein (APP)-cleaving enzyme (BACE) 1 cleavage of amyloid precursor protein is an essential step in the generation of the potentially neurotoxic and amyloidogenic A beta 42 peptides in Alzheimer's disease. Although previous mouse studies have shown brain A beta lowering after BACE1 inhibition, extension of such studies to nonhuman primates or man was precluded by poor potency, brain penetration, and pharmacokinetics of available inhibitors. In this study, a novel tertiary carbinamine BACE1 inhibitor, tertiary carbinamine (TC)-1, was assessed in a unique cisterna magna ported rhesus monkey model, where the temporal dynamics of A beta in cerebrospinal fluid (CSF) and plasma could be evaluated. TC-1, a potent inhibitor (IC(50) approximately 0.4 nM), has excellent passive membrane permeability, low susceptibility to P-glycoprotein transport, and lowered brain A beta levels in a mouse model. Intravenous infusion of TC-1 led to a significant but transient lowering of CSF and plasma A beta levels in conscious rhesus monkeys because it underwent CYP3A4-mediated metabolism. Oral codosing of TC-1 with ritonavir, a potent CYP3A4 inhibitor, twice daily over 3.5 days in rhesus monkeys led to sustained plasma TC-1 exposure and a significant and sustained reduction in CSF sAPP beta, A beta 40, A beta 42, and plasma A beta 40 levels. CSF A beta 42 lowering showed an EC(50) of approximately 20 nM with respect to the CSF [TC-1] levels, demonstrating excellent concordance with its potency in a cell-based assay. These results demonstrate the first in vivo proof of concept of CSF A beta lowering after oral administration of a BACE1 inhibitor in a nonhuman primate.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/antagonists & inhibitors , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Humans , Infusions, Intravenous , Macaca mulatta , Mice , Mice, Transgenic , Transfection
19.
Bioorg Med Chem Lett ; 18(2): 716-20, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18061443

ABSTRACT

Antagonism of the bradykinin B(1) receptor represents a potential treatment for chronic pain and inflammation. Novel antagonists incorporating alpha-hydroxy amides were designed that display low-nanomolar affinity for the human bradykinin B(1) receptor and good bioavailability in the rat and dog. In addition, these functionally active compounds show high passive permeability and low susceptibility to phosphoglycoprotein mediated efflux, predictive of good CNS exposure.


Subject(s)
Amides/pharmacology , Bradykinin B1 Receptor Antagonists , Amides/chemistry , Amides/pharmacokinetics , Animals , Biological Availability , Blood-Brain Barrier , Cytochrome P-450 Enzyme Inhibitors , Dogs , Half-Life , Humans , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
20.
J Med Chem ; 50(2): 272-82, 2007 Jan 25.
Article in English | MEDLINE | ID: mdl-17228869

ABSTRACT

A series of biphenylaminocyclopropane carboxamide based bradykinin B1 receptor antagonists has been developed that possesses good pharmacokinetic properties and is CNS penetrant. Discovery that the replacement of the trifluoropropionamide in the lead structure with polyhaloacetamides, particularly a trifluoroacetamide, significantly reduced P-glycoprotein mediated efflux for the series proved essential. One of these novel bradykinin B1 antagonists (13b) also exhibited suitable pharmacokinetic properties and efficient ex vivo receptor occupancy for further development as a novel approach for the treatment of pain and inflammation.


Subject(s)
Acetamides/chemical synthesis , Amides/chemical synthesis , Aminobiphenyl Compounds/chemical synthesis , Benzoates/chemical synthesis , Bradykinin B1 Receptor Antagonists , Brain/metabolism , Cyclopropanes/chemical synthesis , Spinal Cord/metabolism , Acetamides/pharmacokinetics , Acetamides/pharmacology , Administration, Oral , Amides/pharmacokinetics , Amides/pharmacology , Aminobiphenyl Compounds/pharmacokinetics , Aminobiphenyl Compounds/pharmacology , Analgesics/chemical synthesis , Analgesics/chemistry , Analgesics/pharmacology , Animals , Animals, Genetically Modified , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoates/pharmacokinetics , Benzoates/pharmacology , Biological Availability , Blood-Brain Barrier/metabolism , CHO Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Cyclopropanes/pharmacokinetics , Cyclopropanes/pharmacology , Female , Humans , Macaca mulatta , Male , Mice , Rabbits , Radioligand Assay , Rats , Species Specificity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL