Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Ann Nucl Med ; 38(4): 247-263, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38145430

ABSTRACT

INTRODUCTION: Functions of existing automatic module systems for synthesis of radiopharmaceuticals mainly focus on the radiolabeling of small molecules. There are few modules which have achieved full-automatic radiolabeling of non-metallic and metallic nuclides on small molecules, peptides, and antibody drugs. This study aimed to develop and test a full-automatic multifunctional module system for the safe, stable, and efficient production of radiopharmaceuticals. METHODS: According to characteristics of labeling process of radioactive drugs, using UG and Solidworks softwares, full-automatic cassette-based synthesis module system Mortenon M1 for synthesis of radiopharmaceuticals with various radionuclides, was designed and tested. Mortenon M1 has at least three significant highlights: the cassettes are disposable, and there is no need of manual cleaning; the synthesis method program is flexible and can be edited freely by users according to special needs; this module system is suitable for radiolabeling of both small-molecule and macromolecular drugs, with potentially various radionuclides including 18F, 64Cu, 68Ga, 89Zr, 177Lu, etc. By program control methods for certain drugs, Mortenon M1 was used for radiolabeling of both small-molecule drugs such as [68Ga]-FAPI-46 and macromolecular drugs such as [89Zr]-TROP2 antibody. Quality control assays for product purity were performed with radio-iTLC and radio-HPLC, and the radiotracers were confirmed for application in microPET imaging in xenograft tumor-bearing mouse models. RESULTS: Functional tests for Mortenon M1 module system were conducted, with [68Ga]-FAPI-46 and [89Zr]-TROP2 antibody as goal synthetic products, and it displayed that with the cassette modules, the preset goals could be achieved successfully. The radiolabeling synthesis yield was good ([68Ga]-FAPI-46, 70.63% ± 2.85%, n = 10; [89Zr]-TROP2, 82.31% ± 3.92%, n = 10), and the radiochemical purity via radio-iTLC assay of the radiolabeled products was above 99% after purification. MicroPET imaging results showed that the radiolabeled tracers had reasonable radioactive distribution in MDA-MB-231 and SNU-620 xenograft tumor-bearing mice, and the tumor targeted radiouptake was satisfactory for diagnosis. CONCLUSION: This study demonstrated that the full-automatic module system Mortenon M1 is efficient for radiolabeling synthesis of both small-molecule and macromolecular substrates. It may be helpful to reduce radiation exposure for safety, provide qualified radiolabeled products and reliable PET diagnosis, and ensure stable production and supply of radiopharmaceuticals.


Subject(s)
Neoplasms , Radiopharmaceuticals , Humans , Animals , Mice , Radiopharmaceuticals/chemistry , Gallium Radioisotopes/chemistry , Radioisotopes/chemistry , Peptides , Antibodies
2.
Mol Imaging Biol ; 18(4): 579-87, 2016 08.
Article in English | MEDLINE | ID: mdl-26596571

ABSTRACT

PURPOSE: A positron emission tomography (PET) tracer for the enzyme phosphodiesterase 10A (PDE10A) is desirable to guide the discovery and development of PDE10A inhibitors as potential therapeutics. The preclinical characterization of the PDE10A PET tracer [(11)C]MK-8193 is described. PROCEDURES: In vitro binding studies with [(3)H]MK-8193 were conducted in rat, monkey, and human brain tissue. PET studies with [(11)C]MK-8193 were conducted in rats and rhesus monkeys at baseline and following administration of a PDE10A inhibitor. RESULTS: [(3)H]MK-8193 is a high-affinity, selective PDE10A radioligand in rat, monkey, and human brain tissue. In vivo, [(11)C]MK-8193 displays rapid kinetics, low test-retest variability, and a large specific signal that is displaced by a structurally diverse PDE10A inhibitor, enabling the determination of pharmacokinetic/enzyme occupancy relationships. CONCLUSIONS: [(11)C]MK-8193 is a useful PET tracer for the preclinical characterization of PDE10A therapeutic candidates in rat and monkey. Further evaluation of [(11)C]MK-8193 in humans is warranted.


Subject(s)
Heterocyclic Compounds, 2-Ring/chemistry , Phosphoric Diester Hydrolases/metabolism , Positron-Emission Tomography/methods , Animals , Brain/diagnostic imaging , Carbon Radioisotopes , Female , Heterocyclic Compounds, 2-Ring/blood , Heterocyclic Compounds, 2-Ring/chemical synthesis , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Humans , Macaca mulatta , Male , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Rats , Time Factors
3.
Bioorg Med Chem Lett ; 25(21): 4893-4898, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26077491

ABSTRACT

Phosphodiesterase 10A (PDE10A) inhibition has recently been identified as a potential mechanism to treat multiple symptoms that manifest in schizophrenia. In order to facilitate preclinical development and support key proof-of-concept clinical trials of novel PDE10A inhibitors, it is critical to discover positron emission tomography (PET) tracers that enable plasma concentration/PDE10A occupancy relationships to be established across species with structurally diverse PDE10A inhibitors. In this Letter, we describe how a high-throughput screening hit was optimized to provide [(11)C]MK-8193 (8j), a PET tracer that supports the determination of plasma concentration/PDE10A occupancy relationships for structurally diverse series of PDE10A inhibitors in both rat and rhesus monkey.


Subject(s)
Drug Discovery , Heterocyclic Compounds, 2-Ring/chemistry , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Positron-Emission Tomography , Animals , Brain/metabolism , Carbon Radioisotopes , Crystallography, X-Ray , Dose-Response Relationship, Drug , Heterocyclic Compounds, 2-Ring/chemical synthesis , Macaca mulatta , Models, Molecular , Molecular Structure , Phosphodiesterase Inhibitors/chemical synthesis , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/blood , Rats , Structure-Activity Relationship
4.
J Med Chem ; 56(14): 5940-8, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23808489

ABSTRACT

Hydroisoindoline 2 has been previously identified as a potent, brain-penetrant NK1 receptor antagonist with a long duration of action and improved profile of CYP3A4 inhibition and induction compared to aprepitant. However, compound 2 is predicted, based on data in preclinical species, to have a human half-life longer than 40 h and likely to have drug-drug-interactions (DDI), as 2 is a victim of CYP3A4 inhibition caused by its exclusive clearance pathway via CYP3A4 oxidation in humans. We now report 2-[(3aR,4R,5S,7aS)-5-{(1S)-1-[3,5-bis(trifluoromethyl)phenyl]-2-hydroxyethoxy}-4-(2-methylphenyl)octahydro-2H-isoindol-2-yl]-1,3-oxazol-4(5H)-one (3) as a next generation NK1 antagonist that possesses an additional clearance pathway through glucuronidation in addition to that via CYP3A4 oxidation. Compound 3 has a much lower propensity for drug-drug interactions and a reduced estimated human half-life consistent with once daily dosing. In preclinical species, compound 3 has demonstrated potency, brain penetration, and a safety profile similar to 2, as well as excellent pharmacokinetics.


Subject(s)
Isoindoles/chemical synthesis , Neurokinin-1 Receptor Antagonists/chemical synthesis , Oxazoles/chemical synthesis , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Drug Interactions , Glucuronides/metabolism , Humans , Isoindoles/chemistry , Isoindoles/pharmacokinetics , Isoindoles/pharmacology , Metabolic Clearance Rate , Neurokinin-1 Receptor Antagonists/chemistry , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Neurokinin-1 Receptor Antagonists/pharmacology , Oxazoles/chemistry , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Peptide Fragments/pharmacology , Substance P/analogs & derivatives , Substance P/pharmacology
5.
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
6.
J Psychopharmacol ; 25(3): 329-44, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20156926

ABSTRACT

In the accompanying paper we describe how MRK-409 unexpectedly produced sedation in man at relatively low levels of GABA(A) receptor occupancy (∼10%). Since it was not clear whether this sedation was mediated via the α2/α3 or α1 GABA(A) subtype(s), we characterized the properties of TPA023B, a high-affinity imidazotriazine which, like MRK-409, has partial agonist efficacy at the α2 and α3 subtype but is an antagonist at the α1 subtype, at which MRK-409 has weak partial agonism. TPA023B gave dose- and time-dependent occupancy of rat brain GABA(A) receptors as measured using an in vivo [(3)H]flumazenil binding assay, with 50% occupancy corresponding to a respective dose and plasma drug concentration of 0.09 mg/kg and 19 ng/mL, the latter of which was similar to that observed in mice (25 ng/mL) and comparable to values obtained in baboon and man using [(11)C]flumazenil PET (10 and 5.8 ng/mL, respectively). TPA023B was anxiolytic in rodent and primate (squirrel monkey) models of anxiety (elevated plus maze, fear-potentiated startle, conditioned suppression of drinking, conditioned emotional response) yet had no significant effects in rodent or primate assays of ataxia and/or myorelaxation (rotarod, chain-pulling, lever pressing), up to doses (10 mg/kg) corresponding to occupancy of greater than 99%. In man, TPA023B was well tolerated at a dose (1.5 mg) that produced occupancy of >50%, suggesting that the sedation previously seen with MRK-409 is due to the partial agonist efficacy of that compound at the α1 subtype, and highlighting the importance of antagonist efficacy at this particular GABA(A) receptor population for avoiding sedation in man.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , GABA-A Receptor Agonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hydrocarbons, Fluorinated/pharmacology , Adolescent , Adult , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/adverse effects , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/adverse effects , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/adverse effects , Male , Mice , Middle Aged , Protein Subunits , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Saimiri , Species Specificity , Time Factors , Young Adult
7.
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
8.
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
9.
ACS Med Chem Lett ; 1(7): 350-4, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-24900218

ABSTRACT

Amalgamation of the structure-activity relationship of two series of GlyT1 inhibitors developed at Merck led to the discovery of a clinical candidate, compound 16 (DCCCyB), which demonstrated excellent in vivo occupancy of GlyT1 transporters in rhesus monkey as determined by displacement of a PET tracer ligand.

10.
J Pharmacol Exp Ther ; 332(1): 17-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19779131

ABSTRACT

The GABA(A) receptor alpha2/alpha3 subtype-selective compound 7-(1,1-dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine (TPA023; also known as MK-0777) is a triazolopyridazine that has similar, subnanomolar affinity for the benzodiazepine binding site of alpha1-, alpha2-, alpha3-, and alpha5-containing GABA(A) receptors and has partial agonist efficacy at the alpha2 and alpha3 but not the alpha1 or alpha5 subtypes. The purpose of the present study was to define the relationship between plasma TPA023 concentrations and benzodiazepine binding site occupancy across species measured using various methods. Thus, occupancy was measured using either in vivo [(3)H]flumazenil binding or [(11)C]flumazenil small-animal positron emission tomography (microPET) in rats, [(123)I]iomazenil gamma-scintigraphy in rhesus monkeys, and [(11)C]flumazenil PET in baboons and humans. For each study, plasma-occupancy curves were derived, and the plasma concentration of TPA023 required to produce 50% occupancy (EC(50)) was calculated. The EC(50) values for rats, rhesus monkeys, and baboons were all similar and ranged from 19 to 30 ng/ml, although in humans, the EC(50) was slightly lower at 9 ng/ml. In humans, a single 2-mg dose of TPA023 produced in the region of 50 to 60% occupancy in the absence of overt sedative-like effects. Considering that nonselective full agonists are associated with sedation at occupancies of less than 30%, these data emphasize the relatively nonsedating nature of TPA023.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists , Pyridazines/pharmacology , Triazoles/pharmacology , Animals , Binding Sites , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Flumazenil/pharmacology , GABA Agonists/blood , GABA-A Receptor Antagonists , Humans , Macaca mulatta , Papio , Positron-Emission Tomography , Protein Binding , Protein Subunits , Pyridazines/blood , Radioligand Assay , Rats , Species Specificity , Tissue Distribution , Triazoles/blood
11.
Synapse ; 63(12): 1122-32, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19670309

ABSTRACT

Two histamine H3 receptor (H3R) inverse agonist PET tracers have been synthesized and characterized in preclinical studies. Each tracer has high affinity for the histamine H3 receptor, has suitable lipophilicity, and neither is a substrate for the P-glycoprotein efflux pump. A common phenolic precursor was used to synthesize each tracer with high specific activity and radiochemical purity by an alkylation reaction using either [(11)C]MeI or [(18)F]FCD(2)Br. Autoradiographic studies in rhesus monkey and human brain slices showed that each tracer had a widespread distribution with high binding densities in frontal cortex, globus pallidus and striatum, and lower uptake in cerebellum. The specificity of this expression pattern was demonstrated by the blockade of the autoradiographic signal by either the H3R agonist R-alpha-methylhistamine or a histamine H3R inverse agonist. In vivo PET imaging studies in rhesus monkey showed rapid uptake of each tracer into the brain with the same distribution seen in the autoradiographic studies. Each tracer could be blocked by pretreatment with a histamine H3R inverse agonist giving a good specific signal. Comparison of the in vitro metabolism of each compound showed slower metabolism in human liver microsomes than in rhesus monkey liver microsomes, with each compound having a similar clearance rate in humans. The in vivo metabolism of 1b in rhesus monkey showed that at 60 min, approximately 35% of the circulating counts were due to the parent. These tracers are very promising candidates as clinical PET tracers to both study the histamine H3R system and measure receptor occupancy of H3R therapeutic compounds.


Subject(s)
Benzofurans/pharmacology , Benzofurans/pharmacokinetics , Brain/metabolism , Histamine Agonists , Piperidines/pharmacology , Piperidines/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacology , Receptors, Histamine H3/metabolism , Animals , Autoradiography , Benzofurans/blood , Brain/drug effects , Carbon Radioisotopes , Drug Inverse Agonism , Fluorine Radioisotopes , Histamine Agonists/blood , Histamine Agonists/pharmacokinetics , Histamine Agonists/pharmacology , Humans , Isotope Labeling , Macaca mulatta , Magnetic Resonance Imaging , Methylhistamines/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Piperidines/blood , Radiopharmaceuticals/pharmacokinetics
12.
J Pharmacol Exp Ther ; 331(2): 470-84, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19704033

ABSTRACT

3-tert-Butyl-7-(5-methylisoxazol-3-yl)-2-(1-methyl-1H-1,2,4-triazol-5-ylmethoxy)-pyrazolo[1,5-d][1,2,4]triazine (MRK-016) is a pyrazolotriazine with an affinity of between 0.8 and 1.5 nM for the benzodiazepine binding site of native rat brain and recombinant human alpha1-, alpha2-, alpha3-, and alpha5-containing GABA(A) receptors. It has inverse agonist efficacy selective for the alpha5 subtype, and this alpha5 inverse agonism is greater than that of the prototypic alpha5-selective compound 3-(5-methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-hdyl)methyloxy]-1,2,4-triazolo[3,4-a]phthalazine (alpha5IA). Consistent with its greater alpha5 inverse agonism, MRK-016 increased long-term potentiation in mouse hippocampal slices to a greater extent than alpha5IA. MRK-016 gave good receptor occupancy after oral dosing in rats, with the dose required to produce 50% occupancy being 0.39 mg/kg and a corresponding rat plasma EC(50) value of 15 ng/ml that was similar to the rhesus monkey plasma EC(50) value of 21 ng/ml obtained using [(11)C]flumazenil positron emission tomography. In normal rats, MRK-016 enhanced cognitive performance in the delayed matching-to-position version of the Morris water maze but was not anxiogenic, and in mice it was not proconvulsant and did not produce kindling. MRK-016 had a short half-life in rat, dog, and rhesus monkey (0.3-0.5 h) but had a much lower rate of turnover in human compared with rat, dog, or rhesus monkey hepatocytes. Accordingly, in human, MRK-016 had a longer half-life than in preclinical species ( approximately 3.5 h). Although it was well tolerated in young males, with a maximal tolerated single dose of 5 mg corresponding to an estimated occupancy in the region of 75%, MRK-016 was poorly tolerated in elderly subjects, even at a dose of 0.5 mg, which, along with its variable human pharmacokinetics, precluded its further development.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists , Isoxazoles/pharmacology , Triazines/pharmacology , Animals , Anxiety/psychology , Behavior, Animal/drug effects , Convulsants/pharmacology , Dogs , Dose-Response Relationship, Drug , Electric Stimulation , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Fibroblasts , Flumazenil/metabolism , GABA Agonists/metabolism , GABA Agonists/pharmacokinetics , GABA Modulators/metabolism , Hepatocytes/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Macaca mulatta , Male , Maze Learning/drug effects , Mice , Patch-Clamp Techniques , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Young Adult
13.
Br J Pharmacol ; 157(5): 796-803, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19422390

ABSTRACT

BACKGROUND AND PURPOSE: alpha5IA (3-(5-methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3,4-a]phthalazine) is a triazolophthalazine with subnanomolar affinity for alpha1-, alpha2-, alpha3- and alpha5-containing GABA(A) receptors. Here we have evaluated the relationship between plasma alpha5IA concentrations and benzodiazepine binding site occupancy in rodents and primates (rhesus monkey). EXPERIMENTAL APPROACH: In awake rats, occupancy was measured at various times after oral dosing with alpha5IA (0.03-30 mgxkg(-1)) using an in vivo {[(3)H]flumazenil (8-fluoro 5,6-dihydro-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylic acid ethyl ester)} binding assay. In anaesthetized rhesus monkeys, occupancy was measured using {[(123)I]iomazenil (ethyl 5,6-dihydro-7-iodo-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylic acid ethyl ester)} gamma-scintigraphy and a bolus/infusion paradigm. In both rat and rhesus monkey, the plasma drug concentration corresponding to 50% occupancy (EC(50)) was calculated. KEY RESULTS: In rats, alpha5IA occupancy was dose- and time-dependent with maximum occupancy occurring within the first 2 h. However, rat plasma EC(50) was time-independent, ranging from 42 to 67 ngxmL(-1) over a 24 h time course with the average being 52 ngxmL(-1) (i.e. occupancy decreased as plasma drug concentrations fell). In rhesus monkeys, the EC(50) for alpha5IA displacing steady-state [(123)I]iomazenil binding was 57 ngxmL(-1). CONCLUSIONS AND IMPLICATIONS: Rat plasma EC(50) values did not vary as a function of time indicating that alpha5IA dissociates readily for the GABA(A) receptor in vivo. These data also suggest that despite the different assays used (terminal assays of [(3)H]flumazenil in vivo binding in rats and [(123)I]iomazenil gamma-scintigraphy in anaesthetized rhesus monkeys), these techniques produced similar plasma alpha5IA EC(50) values (52 and 57 ngxmL(-1) respectively) and that the plasma-occupancy relationship for alpha5IA translates across these two species.


Subject(s)
Benzodiazepines/metabolism , Brain/metabolism , Pyridines/metabolism , Receptors, GABA-A/metabolism , Administration, Oral , Animals , Benzodiazepines/administration & dosage , Benzodiazepines/blood , Benzodiazepines/pharmacokinetics , Binding Sites , Binding, Competitive , Brain/diagnostic imaging , Dose-Response Relationship, Drug , Flumazenil/administration & dosage , Flumazenil/analogs & derivatives , Flumazenil/metabolism , GABA-A Receptor Antagonists , Infusions, Intravenous , Injections, Intravenous , Iodine Radioisotopes , Ligands , Macaca mulatta , Male , Pyridines/administration & dosage , Pyridines/blood , Pyridines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tomography, Emission-Computed, Single-Photon
14.
J Med Chem ; 52(9): 3039-46, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19354254

ABSTRACT

3-[(3aR,4R,5S,7aS)-5-{(1R)-1-[3,5-bis(trifluoromethyl)phenyl]ethoxy}-4-(4-fluorophenyl)octahydro-2H-isoindol-2-yl]cyclopent-2-en-1-one (17) is a high affinity, brain-penetrant, hydroisoindoline-based neurokinin-1 (NK(1)) receptor antagonist with a long central duration of action in preclinical species and a minimal drug-drug interaction profile. Positron emission tomography (PET) studies in rhesus showed that this compound provides 90% NK(1) receptor blockade in rhesus brain at a plasma level of 67 nM, which is about 10-fold more potent than aprepitant, an NK(1) antagonist marketed for the prevention of chemotherapy-induced and postoperative nausea and vomiting (CINV and PONV). The synthesis of this enantiomerically pure compound containing five stereocenters includes a Diels-Alder condensation, one chiral separation of the cyclohexanol intermediate, an ether formation using a trichloroacetimidate intermediate, and bis-alkylation to form the cyclic amine.


Subject(s)
Brain/metabolism , Isoindoles/metabolism , Isoindoles/pharmacology , Neurokinin-1 Receptor Antagonists , Administration, Oral , Animals , Aprepitant , CHO Cells , Cricetinae , Cricetulus , Drug Interactions , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Isoindoles/chemical synthesis , Isoindoles/pharmacokinetics , Macaca mulatta , Morpholines/pharmacology , Stereoisomerism
15.
Mol Imaging Biol ; 11(4): 246-52, 2009.
Article in English | MEDLINE | ID: mdl-19130142

ABSTRACT

PURPOSE: The in vitro and in vivo evaluation of the selective, high affinity (human CB1 IC(50) 0.49 nM) inverse agonist CB1R tracer [(11)C]CB-119, a close analog of the previously disclosed [(18)F]MK-9470, was undertaken. PROCEDURES: [(11)C]CB-119 was synthesized with high specific activity by alkylation of a phenolic precursor with [(11)C]methyl iodide. In vitro autoradiographic studies using rhesus brain slices were carried out using [(3)H]CB-119, and in vivo imaging studies were carried out using [(11)C]CB-119 in rhesus monkeys under baseline and blocked conditions. RESULTS: Autoradiographic studies in rhesus brain showed the expected distribution pattern for CB1R with highest binding in the cerebral cortex, cerebellum, caudate/putamen, globus pallidus, substantia nigra, and hippocampus. Lower binding was seen in the posterior hypothalamus, ventral tegmental area, and periventricular gray area, and the lowest binding was in the thalamic nuclei. The binding of [(3)H]CB-119 was fully blocked by the addition of 10 microM CB-119. Rhesus positron emission tomography imaging studies showed very good brain uptake and a distribution pattern consistent with that seen in the autoradiographic studies. The kinetics of tracer uptake was slow. The brain uptake was blocked by pretreatment with taranabant, a CB1R inverse agonist. The specific signal (total/nonspecific) in rhesus putamen at 90 min was approximately 6:1. CONCLUSIONS: [(11)C]CB-119 is a suitable tracer for imaging central CB1 receptors.


Subject(s)
Amides/metabolism , Brain/diagnostic imaging , Brain/metabolism , Positron-Emission Tomography , Pyridines/metabolism , Radiopharmaceuticals/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Autoradiography , Carbon Radioisotopes , Ligands , Macaca mulatta , Protein Binding , Receptor, Cannabinoid, CB1/agonists , Tissue Distribution , Tritium/chemistry
16.
Cell Metab ; 7(1): 68-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18177726

ABSTRACT

Cannabinoid 1 receptor (CB1R) inverse agonists are emerging as a potential obesity therapy. However, the physiological mechanisms by which these agents modulate human energy balance are incompletely elucidated. Here, we describe a comprehensive clinical research study of taranabant, a structurally novel acyclic CB1R inverse agonist. Positron emission tomography imaging using the selective CB1R tracer [(18)F]MK-9470 confirmed central nervous system receptor occupancy levels ( approximately 10%-40%) associated with energy balance/weight-loss effects in animals. In a 12-week weight-loss study, taranabant induced statistically significant weight loss compared to placebo in obese subjects over the entire range of evaluated doses (0.5, 2, 4, and 6 mg once per day) (p < 0.001). Taranabant treatment was associated with dose-related increased incidence of clinical adverse events, including mild to moderate gastrointestinal and psychiatric effects. Mechanism-of-action studies suggest that engagement of the CB1R by taranabant leads to weight loss by reducing food intake and increasing energy expenditure and fat oxidation.


Subject(s)
Amides/pharmacology , Energy Intake/drug effects , Energy Metabolism/drug effects , Pyridines/pharmacology , Receptor, Cannabinoid, CB1/agonists , Weight Loss/drug effects , Adult , Aged , Amides/therapeutic use , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Fats/metabolism , Humans , Middle Aged , Positron-Emission Tomography , Pyridines/therapeutic use
17.
Bioorg Med Chem Lett ; 17(18): 5191-8, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17637506

ABSTRACT

SAR studies on amides, ureas, and vinylogous amides derived from pyrrolidine led to the discovery of several potent hNK(1) antagonists. One particular vinylogous amide (45b) had excellent potency, selectivity, pharmacokinetic profile, and functional activity in vivo. An in vivo rhesus macaque brain receptor occupancy PET study for compound 45b revealed an estimated Occ(90) approximately 300 ng/ml.


Subject(s)
Neurokinin-1 Receptor Antagonists , Pyrrolidines/pharmacology , Administration, Oral , Animals , Biological Availability , Brain/metabolism , Humans , Macaca mulatta , Pyrrolidines/pharmacokinetics , Species Specificity
18.
J Med Chem ; 50(15): 3427-30, 2007 Jul 26.
Article in English | MEDLINE | ID: mdl-17608398

ABSTRACT

The discovery of a structurally distinct cannabinoid-1 receptor (CB1R) positron emission tomography tracer is described. Starting from an acyclic amide CB1R inverse agonist (1) as the lead compound, an efficient route to introduce 18F to the molecule was developed. Further optimization focused on reducing the lipophilicity and increasing the CB1R affinity. These efforts led to the identification of [18F]-16 that exhibited good brain uptake and an excellent signal-to-noise ratio in rhesus monkeys.


Subject(s)
Amides/chemical synthesis , Fluorine Radioisotopes , Pyridines/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Receptor, Cannabinoid, CB1/metabolism , Amides/chemistry , Amides/pharmacokinetics , Animals , Brain/diagnostic imaging , Brain/metabolism , CHO Cells , Cricetinae , Cricetulus , Humans , Macaca mulatta , Positron-Emission Tomography , Pyridines/chemistry , Pyridines/pharmacokinetics , Radioligand Assay , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Recombinant Proteins/metabolism , Structure-Activity Relationship
19.
Proc Natl Acad Sci U S A ; 104(23): 9800-5, 2007 Jun 05.
Article in English | MEDLINE | ID: mdl-17535893

ABSTRACT

[(18)F]MK-9470 is a selective, high-affinity, inverse agonist (human IC(50), 0.7 nM) for the cannabinoid CB1 receptor (CB1R) that has been developed for use in human brain imaging. Autoradiographic studies in rhesus monkey brain showed that [(18)F]MK-9470 binding is aligned with the reported distribution of CB1 receptors with high specific binding in the cerebral cortex, cerebellum, caudate/putamen, globus pallidus, substantia nigra, and hippocampus. Positron emission tomography (PET) imaging studies in rhesus monkeys showed high brain uptake and a distribution pattern generally consistent with that seen in the autoradiographic studies. Uptake was blocked by pretreatment with a potent CB1 inverse agonist, MK-0364. The ratio of total to nonspecific binding in putamen was 4-5:1, indicative of a strong specific signal that was confirmed to be reversible via displacement studies with MK-0364. Baseline PET imaging studies in human research subject demonstrated behavior of [(18)F]MK-9470 very similar to that seen in monkeys, with very good test-retest variability (7%). Proof of concept studies in healthy young male human subjects showed that MK-0364, given orally, produced a dose-related reduction in [(18)F]MK-9470 binding reflecting CB1R receptor occupancy by the drug. Thus, [(18)F]MK-9470 has the potential to be a valuable, noninvasive research tool for the in vivo study of CB1R biology and pharmacology in a variety of neuropsychiatric disorders in humans. In addition, it allows demonstration of target engagement and noninvasive dose-occupancy studies to aid in dose selection for clinical trials of CB1R inverse agonists.


Subject(s)
Brain/anatomy & histology , Positron-Emission Tomography/methods , Radioactive Tracers , Receptor, Cannabinoid, CB1/ultrastructure , Amides/metabolism , Animals , Autoradiography , Brain/metabolism , Fluorine Radioisotopes , Humans , Image Processing, Computer-Assisted , Macaca mulatta , Male , Molecular Structure , Pyridines/metabolism , Receptor, Cannabinoid, CB1/metabolism
20.
Cell Metab ; 4(4): 275-82, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17011500

ABSTRACT

Neuropeptide Y (NPY) is a potent orexigenic neuropeptide, and antagonism of NPY Y1 and NPY Y5 receptors (NPYxR) is considered a potentially important anti-obesity drug target. We tested the hypothesis that blockade of the NPY5R will lead to weight loss in humans using MK-0557, a potent, highly selective, orally active NPY5R antagonist. The initial series of experiments reported herein, including a multiple-dose positron-emission tomography study and a 12 week proof-of concept/dose-ranging study, suggested an optimal MK-0557 dose of 1 mg/day. The hypothesis was then tested in a 52 week, multicenter, randomized, double-blind, placebo-controlled trial involving 1661 overweight and obese patients. Although statistically significant at 52 weeks, the magnitude of induced weight loss was not clinically meaningful. These observations provide the first clinical insight into the human NPY-energy homeostatic pathway and suggest that solely targeting the NPY5R in future drug development programs is unlikely to produce therapeutic efficacy.


Subject(s)
Anti-Obesity Agents/therapeutic use , Cyclohexanes/therapeutic use , Obesity/drug therapy , Pyrazoles/therapeutic use , Receptors, Neuropeptide Y/antagonists & inhibitors , Spiro Compounds/therapeutic use , Administration, Oral , Adolescent , Adult , Aged , Anti-Obesity Agents/administration & dosage , Body Weight , Cyclohexanes/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Middle Aged , Molecular Structure , Placebos , Positron-Emission Tomography/methods , Pyrazoles/administration & dosage , Receptors, Neuropeptide Y/metabolism , Sensitivity and Specificity , Spiro Compounds/administration & dosage , Structure-Activity Relationship , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...