Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Nucl Med ; 63(12): 1919-1924, 2022 12.
Article in English | MEDLINE | ID: mdl-35772961

ABSTRACT

Phosphodiesterase-4 (PDE4), which metabolizes the second messenger cyclic adenosine monophosphate (cAMP), has 4 isozymes: PDE4A, PDE4B, PDE4C, and PDE4D. PDE4B and PDE4D have the highest expression in the brain and may play a role in the pathophysiology and treatment of depression and dementia. This study evaluated the properties of the newly developed PDE4B-selective radioligand 18F-PF-06445974 in the brains of rodents, monkeys, and humans. Methods: Three monkeys and 5 healthy human volunteers underwent PET scans after intravenous injection of 18F-PF-06445974. Brain uptake was quantified as total distribution volume (V T) using the standard 2-tissue-compartment model and serial concentrations of parent radioligand in arterial plasma. Results: 18F-PF-06445974 readily distributed throughout monkey and human brain and had the highest binding in the thalamus. The value of V T was well identified by a 2-tissue-compartment model but increased by 10% during the terminal portions (40 and 60 min) of the monkey and human scans, respectively, consistent with radiometabolite accumulation in the brain. The average human V T values for the whole brain were 9.5 ± 2.4 mL ⋅ cm-3 Radiochromatographic analyses in knockout mice showed that 2 efflux transporters-permeability glycoprotein (P-gp) and breast cancer resistance protein (BCRP)-completely cleared the problematic radiometabolite but also partially cleared the parent radioligand from the brain. In vitro studies with the human transporters suggest that the parent radioligand was a partial substrate for BCRP and, to a lesser extent, for P-gp. Conclusion: 18F-PF-06445974 quantified PDE4B in the human brain with reasonable, but not complete, success. The gold standard compartmental method of analyzing brain and plasma data successfully identified the regional densities of PDE4B, which were widespread and highest in the thalamus, as expected. Because the radiometabolite-induced error was only about 10%, the radioligand is, in the opinion of the authors, suitable to extend to clinical studies.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Neoplasm Proteins , Animals , Mice , Humans , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Neoplasm Proteins/metabolism , Positron-Emission Tomography/methods , Brain/diagnostic imaging , Brain/metabolism , Haplorhini/metabolism , Radiopharmaceuticals/metabolism
2.
Front Neurosci ; 14: 600178, 2020.
Article in English | MEDLINE | ID: mdl-33551724

ABSTRACT

PDE10A, a phosphodiesterase that inactivates both cAMP and cGMP, is a unique signaling molecule in being highly and nearly exclusively expressed in striatal medium spiny neurons. These neurons dynamically integrate cortical information with dopamine-signaled value to mediate action selection among available behavioral options. Medium spiny neurons are components of either the direct or indirect striatal output pathways. Selective activation of indirect pathway medium spiny neurons by dopamine D2 receptor antagonists is putatively a key element in the mechanism of their antipsychotic efficacy. While PDE10A is expressed in all medium spiny neurons, studies in rodents indicated that PDE10A inhibition has behavioral effects in several key assays that phenocopy dopamine D2 receptor inhibition. This finding gave rise to the hypothesis that PDE10A inhibition also preferentially activates indirect pathway medium spiny neurons, a hypothesis that is consistent with electrophysiological, neurochemical, and molecular effects of PDE10A inhibitors. These data underwrote industry-wide efforts to investigate and develop PDE10A inhibitors as novel antipsychotics. Disappointingly, PDE10A inhibitors from 3 companies failed to evidence antipsychotic activity in patients with schizophrenia to the same extent as standard-of-care D2 antagonists. Given the notable similarities between PDE10A inhibitors and D2 antagonists, gaining an understanding of why only the latter class is antipsychotic affords a unique window into the basis for this therapeutic efficacy. With this in mind, we review the data on PDE10A inhibition as a step toward back-translating the limited antipsychotic efficacy of PDE10A inhibitors, hopefully to inform new efforts to develop better therapeutics to treat psychosis and schizophrenia.

3.
J Clin Psychopharmacol ; 39(6): 575-582, 2019.
Article in English | MEDLINE | ID: mdl-31688451

ABSTRACT

BACKGROUND: Current treatments for psychotic symptoms associated with schizophrenia often provide inadequate efficacy with unacceptable adverse effects. Improved therapeutics have long been a goal of research. Preclinical testing suggests that phosphodiesterase 10A (PDE10A) inhibitors may provide a novel approach to treating psychosis associated with schizophrenia. METHODS: The efficacy and safety of a highly selective PDE10A inhibitor, PF-02545920, was evaluated in a phase 2 multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Eligible patients (18-65 years) with an acute exacerbation of schizophrenia were randomized 2:2:1:2 to PF-02545920 (5 or 15 mg every 12 hours [Q12H] titrated), risperidone (3 mg Q12H), or placebo for 28 days (n = 74:74:37:74). The primary objectives were to evaluate the efficacy of PF-02545920 using the Positive and Negative Syndrome Scale (PANNS) and safety/tolerability. RESULTS: At day 28, PF-02545920 (either dose) was not significantly different from placebo for mean change from baseline in the PANNS total score (primary end point) or most other end points. Pharmacokinetics exposures seemed adequate for binding/inhibiting PDE10A enzyme. Risperidone was statistically different from placebo for the PANNS total score, demonstrating study sensitivity. Incidence rates for adverse events were similar among the groups. Both doses of PF-02545920 were generally well tolerated. Dystonia occurred in 1, 6, 0, and 3 patients in the PF-02545920 5 mg Q12H, PF-02545920 15 mg Q12H, risperidone, and placebo groups, respectively. CONCLUSIONS: Neither dose of PF-02545920 was superior to placebo for the primary and most secondary end points. This indicates that PDE10A inhibition does not produce an antipsychotic effect in patients with acute exacerbation of schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Dystonia/chemically induced , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/drug effects , Pyrazoles/pharmacology , Quinolines/pharmacology , Risperidone/pharmacology , Schizophrenia/drug therapy , Treatment Outcome , Acute Disease , Adult , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/adverse effects , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Quinolines/administration & dosage , Quinolines/adverse effects , Risperidone/administration & dosage , Risperidone/adverse effects , Severity of Illness Index
4.
Front Neurosci ; 13: 837, 2019.
Article in English | MEDLINE | ID: mdl-31507355

ABSTRACT

PDE9A is a cGMP-specific phosphodiesterase expressed in neurons throughout the brain that has attracted attention as a therapeutic target to treat cognitive disorders. Indeed, PDE9A inhibitors are under evaluation in clinical trials as a treatment for Alzheimer's disease and schizophrenia. However, little is known about the cGMP signaling cascades regulated by PDE9A. Canonical cGMP signaling in brain follows the activation of neuronal nitric oxide synthase (nNOS) and the generation of nitric oxide, which activates soluble guanylyl cyclase and cGMP synthesis. However, we show that in mice, PDE9A regulates a pool of cGMP that is independent of nNOS, specifically, and nitric oxide signaling in general. This PDE9A-regulated cGMP pool appears to be highly compartmentalized and independent of cGMP pools regulated by several PDEs. These findings provide a new foundation for study of the upstream and downstream signaling elements regulated by PDE9A and its potential as a therapeutic target for brain disease.

5.
J Clin Psychopharmacol ; 39(4): 318-328, 2019.
Article in English | MEDLINE | ID: mdl-31205187

ABSTRACT

BACKGROUND: Effective treatments for managing suboptimal clinical responses to current therapy for schizophrenia remain a critical unmet need. Phosphodiesterase 10A (PDE10A) inhibition represents a mechanistically novel approach to the treatment of schizophrenia, with preclinical studies suggesting improvements in partially responsive symptoms could be achieved via adjunctive use of the PDE10A inhibitor PF-02545920. Therefore, the adjunctive safety, tolerability, pharmacokinetics, and efficacy of multiple repeat doses of PF-02545920 were investigated in a phase 1b study and subsequent phase 2 study. METHODS: The phase 1b study randomized 37 adult patients with stable symptomatology and stable antipsychotic regimens within 3 cohorts. Study participants received ascending doses of PF-02545920 or placebo for 10 to 18 days. The phase 2 study randomized 240 outpatients with stable symptomatology but suboptimal response to current antipsychotic regimens 1:1:1 to PF-02545920 5 mg, PF-02545920 15 mg, or placebo every 12 hours for 12 weeks. The primary efficacy end point of the phase 2 study was change in the Positive and Negative Syndrome Scale total score from baseline to week 12, with changes in other clinical assessments as secondary end points. RESULTS: Treatment was well tolerated, and observed PF-02545920 exposures were within the range predicted to be adequate for demonstrating efficacy. However, no significant differences in the prespecified efficacy end points between the 2 PF-02545920 treatment arms and placebo were observed. CONCLUSIONS: Current data and results of a prior monotherapy study in which PF-02545920 failed to differentiate from placebo refute the hypothesis that PDE10A inhibitors have use as antipsychotic agents for schizophrenia.


Subject(s)
Phosphodiesterase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Quinolines/therapeutic use , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/therapeutic use , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Psychiatric Status Rating Scales
6.
Brain Res ; 1711: 140-145, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30664847

ABSTRACT

Highly specific and sensitive biomarkers for pathologies related to dysfunctions in the basal ganglia circuit are of great value to assess therapeutic efficacy not only clinically to establish an early diagnosis, but also in terms of monitoring the efficacy of therapeutic interventions and decelerated neurodegeneration. The phosphodiesterase 10A (PDE10A) enzyme plays a central role in striatal signaling and is implicated in several neuropsychiatric disorders involving striatal pathology, such as Huntingtons disease (HD) and schizophrenia. Inhibition of PDE10A activates the neurons in the striatum and consequently leads to alteration of behavioral aspects modulated by the striatal circuit. [18F]MNI-659, (2-(2-(3-(4-(2-[18F]fluoroethoxy)phenyl)-7-methyl-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione), is a newly developed PET radioligand that shows a high binding to PDE10A in the human brain in vivo. In the present study, we examined the in vitro binding of [18F]MNI-659 in human postmortem brain to gain a better understanding of the presence, density, disease-related alterations and therapy related to changes in PDE10A expression. The results show high specific binding of [18F]MNI-659 in the caudate nucleus, putamen and the hippocampal formation. Low specific [18F]MNI-659 binding was detected in nucleus accumbens in comparison to the caudate nucleus and putamen. In vitro binding studies with [18F]MNI-659 will facilitate in elucidating better understanding of the role of PDE10A activity in health and disease that may lead to new diagnostic opportunities in HD.


Subject(s)
Brain/enzymology , Phosphoric Diester Hydrolases/metabolism , Phthalimides , Positron-Emission Tomography/methods , Quinazolinones , Adult , Aged , Basal Ganglia/enzymology , Basal Ganglia/metabolism , Brain/diagnostic imaging , Cadaver , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Female , Fluorine Radioisotopes , Humans , Male , Middle Aged , Neostriatum/enzymology , Neostriatum/metabolism , Radiopharmaceuticals
8.
J Med Chem ; 61(3): 1001-1018, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29293004

ABSTRACT

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


Subject(s)
Brain/drug effects , Brain/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Animals , Biological Availability , Brain/physiology , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Inhibitory Concentration 50 , Memory, Short-Term/drug effects , Molecular Docking Simulation , Protein Conformation
9.
J Med Chem ; 60(13): 5673-5698, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28574706

ABSTRACT

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


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Drug Design , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Crystallography, X-Ray , Cyclic Nucleotide Phosphodiesterases, Type 2/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dogs , Haplorhini , Humans , Mice , Molecular Docking Simulation , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/pharmacokinetics , Rats
10.
Brain Behav Immun ; 64: 285-295, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28438557

ABSTRACT

Inhibitors of phosphodiesterase-4 (PDE4) have been approved for the treatment of inflammatory disorders, but are associated with dose-limiting nausea and vomiting. These side effects are hypothesized to be mediated by inhibition of the PDE4D isozyme. Here we demonstrate the anti-inflammatory effects of the novel brain penetrant PDE4D-sparing PDE4 inhibitor, ABI-4. ABI-4 was a potent (EC50∼14nM) inhibitor of lipopolysaccharide (LPS) induced TNF-α release from mouse microglia and human PBMCs. ABI-4 (0.32mg/kg) blocked LPS-induced release of pro-inflammatory cytokines (TNF-α, IL-1ß, IL-6) in blood and brain of mice. In a rat model of endotoxin induced uveitis, ABI-4 (0.03-0.3mg/kg) demonstrated steroid-like efficacy in preventing leucocyte infiltration of the aqueous humor when administered 4h after LPS. LPS (0.32mg/kg×5days) caused a 30% upregulation of translocator protein (TSPO) binding which was prevented by co-administration of ABI-4 (0.32mg/kg). In a paradigm to assess motivation, LPS (0.32mg/kg) reduced the number of rewards received, whereas the effect was significantly blunted in mice dosed with ABI-4 (P<0.05) or in PDE4B-/- mice. PDE4B was also shown to modulate brain and plasma levels of TNF-α and IL-1ß in aged mice. Aged mice dosed chronically with ABI-4 (0.32mg/kg) as well as aged PDE4B-/- mice, had significantly lower levels of TNF-α and IL-1ß in brain and plasma relative to vehicle treated or PDE4+/+ mice. Together these data demonstrate that the PDE4D sparing, PDE4 inhibitor, ABI-4 retains potency and efficacy in exerting anti-inflammatory effects. This mechanism warrants further investigation in human disorders involving neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Brain/drug effects , Encephalitis/drug therapy , Inflammation/drug therapy , Phosphodiesterase 4 Inhibitors/administration & dosage , Animals , Brain/metabolism , Encephalitis/chemically induced , Encephalitis/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation Mediators/metabolism , Isoenzymes/administration & dosage , Lipopolysaccharides , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Motivation/drug effects , Rats, Inbred Lew
11.
ACS Chem Neurosci ; 8(1): 165-177, 2017 01 18.
Article in English | MEDLINE | ID: mdl-27715007

ABSTRACT

Dopamine receptor antagonism is a compelling molecular target for the treatment of a range of psychiatric disorders, including substance use disorders. From our corporate compound file, we identified a structurally unique D3 receptor (D3R) antagonist scaffold, 1. Through a hybrid approach, we merged key pharmacophore elements from 1 and D3 agonist 2 to yield the novel D3R/D2R antagonist PF-4363467 (3). Compound 3 was designed to possess CNS drug-like properties as defined by its CNS MPO desirability score (≥4/6). In addition to good physicochemical properties, 3 exhibited low nanomolar affinity for the D3R (D3 Ki = 3.1 nM), good subtype selectivity over D2R (D2 Ki = 692 nM), and high selectivity for D3R versus other biogenic amine receptors. In vivo, 3 dose-dependently attenuated opioid self-administration and opioid drug-seeking behavior in a rat operant reinstatement model using animals trained to self-administer fentanyl. Further, traditional extrapyramidal symptoms (EPS), adverse side effects arising from D2R antagonism, were not observed despite high D2 receptor occupancy (RO) in rodents, suggesting that compound 3 has a unique in vivo profile. Collectively, our data support further investigation of dual D3R and D2R antagonists for the treatment of drug addiction.


Subject(s)
Analgesics, Opioid/adverse effects , Dopamine D2 Receptor Antagonists/chemistry , Dopamine D2 Receptor Antagonists/pharmacology , Drug-Seeking Behavior/drug effects , Receptors, Dopamine D3/antagonists & inhibitors , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Cell Line, Transformed , Conditioning, Operant/drug effects , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Fentanyl/adverse effects , Humans , Male , Neuroblastoma/pathology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Self Administration , Sulfonamides/chemistry , Sulfonamides/pharmacology
12.
Neuron ; 92(6): 1220-1237, 2016 Dec 21.
Article in English | MEDLINE | ID: mdl-27916455

ABSTRACT

Huntington's disease (HD) symptoms are driven to a large extent by dysfunction of the basal ganglia circuitry. HD patients exhibit reduced striatal phoshodiesterase 10 (PDE10) levels. Using HD mouse models that exhibit reduced PDE10, we demonstrate the benefit of pharmacologic PDE10 inhibition to acutely correct basal ganglia circuitry deficits. PDE10 inhibition restored corticostriatal input and boosted cortically driven indirect pathway activity. Cyclic nucleotide signaling is impaired in HD models, and PDE10 loss may represent a homeostatic adaptation to maintain signaling. Elevation of both cAMP and cGMP by PDE10 inhibition was required for rescue. Phosphoproteomic profiling of striatum in response to PDE10 inhibition highlighted plausible neural substrates responsible for the improvement. Early chronic PDE10 inhibition in Q175 mice showed improvements beyond those seen with acute administration after symptom onset, including partial reversal of striatal deregulated transcripts and the prevention of the emergence of HD neurophysiological deficits. VIDEO ABSTRACT.


Subject(s)
Cerebral Cortex/drug effects , Huntington Disease/physiopathology , Neostriatum/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Animals , Basal Ganglia/diagnostic imaging , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Basal Ganglia/physiopathology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Disease Models, Animal , Huntington Disease/metabolism , Mice , Neostriatum/diagnostic imaging , Neostriatum/metabolism , Neostriatum/physiopathology , Phosphoric Diester Hydrolases , Positron-Emission Tomography , Subthalamic Nucleus/diagnostic imaging , Subthalamic Nucleus/drug effects , Subthalamic Nucleus/metabolism , Subthalamic Nucleus/physiopathology , Tritium
13.
Am J Hum Genet ; 98(4): 735-43, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058446

ABSTRACT

Deficits in the basal ganglia pathways modulating cortical motor activity underlie both Parkinson disease (PD) and Huntington disease (HD). Phosphodiesterase 10A (PDE10A) is enriched in the striatum, and animal data suggest that it is a key regulator of this circuitry. Here, we report on germline PDE10A mutations in eight individuals from two families affected by a hyperkinetic movement disorder due to homozygous mutations c.320A>G (p.Tyr107Cys) and c.346G>C (p.Ala116Pro). Both mutations lead to a reduction in PDE10A levels in recombinant cellular systems, and critically, positron-emission-tomography (PET) studies with a specific PDE10A ligand confirmed that the p.Tyr107Cys variant also reduced striatal PDE10A levels in one of the affected individuals. A knock-in mouse model carrying the homologous p.Tyr97Cys variant had decreased striatal PDE10A and also displayed motor abnormalities. Striatal preparations from this animal had an impaired capacity to degrade cyclic adenosine monophosphate (cAMP) and a blunted pharmacological response to PDE10A inhibitors. These observations highlight the critical role of PDE10A in motor control across species.


Subject(s)
Corpus Striatum/pathology , Hyperkinesis/genetics , Mutation , Phosphoric Diester Hydrolases/genetics , Alleles , Amino Acid Sequence , Animals , Disease Models, Animal , Gene Expression Regulation , Genetic Variation , HEK293 Cells , Humans , Hyperkinesis/diagnosis , Hyperkinesis/pathology , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Pedigree , Phosphodiesterase Inhibitors/metabolism , Sequence Alignment
14.
J Med Chem ; 58(10): 4291-308, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25905800

ABSTRACT

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


Subject(s)
Drug Evaluation, Preclinical/methods , Receptors, AMPA/metabolism , Sulfonamides/pharmacology , Thiophenes/pharmacology , Administration, Oral , Adolescent , Adult , Aged , Animals , Binding Sites , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Discovery , Drug Stability , Female , Humans , Male , Memory, Short-Term/drug effects , Mice, Inbred C57BL , Middle Aged , Protein Conformation , Rats, Sprague-Dawley , Schizophrenia/drug therapy , Structure-Activity Relationship , Sulfonamides/chemistry , Thiophenes/chemistry , Young Adult
15.
J Neurosci ; 35(14): 5781-91, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855188

ABSTRACT

The striatum contains a rich variety of cyclic nucleotide phosphodiesterases (PDEs), which play a critical role in the regulation of cAMP and cGMP signaling. The dual-substrate enzyme PDE10A is the most highly expressed PDE in striatal medium-sized spiny neurons (MSNs) with low micromolar affinity for both cyclic nucleotides. Previously, we have shown that systemic and local administration of the selective PDE10A inhibitor TP-10 potently increased the responsiveness of MSNs to cortical stimulation. However, the signaling mechanisms underlying PDE10A inhibitor-induced changes in corticostriatal transmission are only partially understood. The current studies assessed the respective roles of cAMP and cGMP in the above effects using soluble guanylyl cyclase (sGC) or adenylate cyclase (AC) specific inhibitors. Cortically evoked spike activity was monitored in urethane-anesthetized rats using in vivo extracellular recordings performed proximal to a microdialysis probe during local infusion of vehicle, the selective sGC inhibitor ODQ, or the selective AC inhibitor SQ 22536. Systemic administration of TP-10 (3.2 mg/kg) robustly increased cortically evoked spike activity in a manner that was blocked following intrastriatal infusion of ODQ (50 µm). The effects of TP-10 on evoked activity were due to accumulation of cGMP, rather than cAMP, as the AC inhibitor SQ was without effect. Consistent with these observations, studies in neuronal NO synthase (nNOS) knock-out (KO) mice confirmed that PDE10A operates downstream of nNOS to limit cGMP production and excitatory corticostriatal transmission. Thus, stimulation of PDE10A acts to attenuate corticostriatal transmission in a manner largely dependent on effects directed at the NO-sGC-cGMP signaling cascade.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/drug effects , Cyclic GMP/metabolism , Nitric Oxide Synthase Type I/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/physiology , Action Potentials/drug effects , Action Potentials/genetics , Animals , Biophysics , Corpus Striatum/cytology , Cyclic AMP/metabolism , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Neural Pathways/drug effects , Neural Pathways/physiology , Neurons/drug effects , Nitric Oxide Synthase Type I/genetics , Rats , Rats, Sprague-Dawley
16.
ACS Chem Biol ; 9(12): 2823-32, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25295858

ABSTRACT

Phosphodiesterases (PDEs) regulate the levels of the second messengers cAMP and cGMP and are important drug targets. PDE10A is highly enriched in medium spiny neurons of the striatum and is an attractive drug target for the treatment of basal ganglia diseases like schizophrenia, Parkinson's disease, or Huntington's disease. Here we describe the design, synthesis, and application of a variety of chemical biology probes, based on the first clinically tested PDE10A inhibitor MP-10, which were used to characterize the chemoproteomic profile of the clinical candidate in its native environment. A clickable photoaffinity probe was used to measure target engagement of MP-10 and revealed differences between whole cell and membrane preparations. Moreover, our results illustrate the importance of the linker design in the creation of functional probes. Biotinylated affinity probes allowed identification of drug-interaction partners in rodent and human tissue and quantitative mass spectrometry analysis revealed highly specific binding of MP-10 to PDE10A with virtually no off-target binding. The profiling of PDE10A chemical biology probes described herein illustrates a strategy by which high affinity inhibitors can be converted into probes for determining selectivity and target engagement of drug candidates in complex biological matrices from native sources.


Subject(s)
Cell Membrane/drug effects , Molecular Probes/chemistry , Neurons/drug effects , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/metabolism , Pyrazoles/chemistry , Quinolines/chemistry , Animals , Binding Sites , Cell Membrane/enzymology , Chromatography, Affinity , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Crystallography, X-Ray , Humans , Light , Models, Molecular , Molecular Probes/chemical synthesis , Neurons/cytology , Neurons/enzymology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/chemistry , Photochemical Processes , Primary Cell Culture , Protein Binding , Pyrazoles/pharmacology , Quinolines/pharmacology , Rats
17.
Neuropharmacology ; 77: 257-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24490227

ABSTRACT

Phosphodiesterase 10A (PDE10A) is highly expressed in striatal medium spiny neurons of both the direct and indirect output pathways. Similar to dopamine D2 receptor antagonists acting on indirect pathway neurons, PDE10A inhibitors have shown behavioral effects in rodent models that predict antipsychotic efficacy. These findings have supported the clinical investigation of PDE10A inhibitors as a new treatment for schizophrenia. However, PDE10A inhibitors and D2 antagonists differ in effects on direct pathway and other neurons of the basal ganglia, indicating that these two drug classes may have divergent antipsychotic efficacy and side effect profile. In the present study, we compare the behavioral effects of the selective PDE10A inhibitor MP-10 to those of the clinical standard D2 antagonist risperidone in rhesus monkeys using a standardized motor disability scale for parkinsonian primates and a newly designed "Drug Effects on Nervous System" scale to assess non-motor effects. Behavioral effects of MP-10 correlated with its plasma levels and its regulation of metabolic activity in striatal and cortical regions as measured by FDG-PET imaging. While MP-10 and risperidone broadly impacted similar behavioral domains in the primate, their effects had a different underlying basis. MP-10-treated animals retained the ability to respond but did not engage tasks, whereas risperidone-treated animals retained the motivation to respond but were unable to perform the intended actions. These findings are discussed in light of what is currently known about the modulation of striatal circuitry by these two classes of compounds, and provide insight into interpreting emerging clinical data with PDE10A inhibitors for the treatment of psychotic symptoms.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Motor Activity/drug effects , Phosphodiesterase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinolines/pharmacology , Risperidone/pharmacology , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Macaca mulatta , Male , Phosphodiesterase Inhibitors/blood , Pyrazoles/blood , Quinolines/blood , Schizophrenia/drug therapy
18.
Psychopharmacology (Berl) ; 231(10): 2189-97, 2014 May.
Article in English | MEDLINE | ID: mdl-24363077

ABSTRACT

RATIONALE: Inhibitors of phosphodiesterase 10A (PDE10A), an enzyme highly expressed in medium spiny neurons of the mammalian striatum, enhance activity in direct (dopamine D1 receptor-expressing) and indirect (D2 receptor-expressing striatal output) pathways. The ability of such agents to act to potentiate D1 receptor signaling while inhibiting D2 receptor signaling suggest that PDE10A inhibitors may have a unique antipsychotic-like behavioral profile differentiated from the D2 receptor antagonist-specific antipsychotics currently used in the treatment of schizophrenia. OBJECTIVES: To evaluate the functional consequences of PDE10A inhibitor modulation of D1 and D2 receptor pathway signaling, we compared the effects of a PDE10A inhibitor (TP-10) on D1 and D2 receptor agonist-induced disruptions in prepulse inhibition (PPI), a measure of sensorimotor gating disrupted in patients with schizophrenia. RESULTS: Our results indicate that, in rats: (1) PDE10A inhibition (TP-10, 0.32-10.0 mg/kg) has no effect on PPI disruption resulting from the mixed D1/D2 receptor agonist apomorphine (0.5 mg/kg), confirming previous report; (2) Yet, TP-10 blocked the PPI disruption induced by the D2 receptor agonist quinpirole (0.5 mg/kg); and attenuated apomorphine-induced disruptions in PPI in the presence of the D1 receptor antagonist SCH23390 (0.005 mg/kg). CONCLUSIONS: These findings indicate that TP-10 cannot block dopamine agonist-induced deficits in PPI in the presence of D1 activation and suggest that the effect of PDE10A inhibition on D1 signaling may be counterproductive in some models of antipsychotic activity. These findings, and the contribution of TP-10 effects in the direct pathway on sensorimotor gating in particular, may have implications for the potential antipsychotic efficacy of PDE10A inhibitors.


Subject(s)
Dopamine Agonists/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Sensory Gating/drug effects , Animals , Apomorphine/pharmacology , Benzazepines/pharmacology , Dopamine Antagonists/pharmacology , Male , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Reflex, Startle/drug effects , Signal Transduction/drug effects
19.
J Pharmacol Exp Ther ; 347(1): 212-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23899905

ABSTRACT

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) positive allosteric modulation (i.e., "potentiation") has been proposed to overcome cognitive impairments in schizophrenia, but AMPAR overstimulation can be excitotoxic. Thus, it is critical to define carefully a potentiator's mechanism-based therapeutic index (TI) and to determine confidently its translatability from rodents to higher-order species. Accordingly, the novel AMPAR potentiator N-{(3R,4S)-3-[4-(5-cyano-2-thienyl)phenyl]tetrahydro-2H-pyran-4-yl}propane-2-sulfonamide (PF-4778574) was characterized in a series of in vitro assays and single-dose animal studies evaluating AMPAR-mediated activities related to cognition and safety to afford an unbound brain compound concentration (Cb,u)-normalized interspecies exposure-response relationship. Because it is unknown which AMPAR subtype(s) may be selectively potentiated for an optimal TI, PF-4778574 binding affinity and functional potency were determined in rodent tissues expected to express a native mixture of AMPAR subunits and their associated proteins to afford composite pharmacological values. Functional activity was also quantified in recombinant cell lines stably expressing human GluA2 flip or flop homotetramers. Procognitive effects of PF-4778574 were evaluated in both rat electrophysiological and nonhuman primate (nhp) behavioral models of pharmacologically induced N-methyl-d-aspartate receptor hypofunction. Safety studies assessed cerebellum-based AMPAR activation (mouse) and motor coordination disruptions (mouse, dog, and nhp), as well as convulsion (mouse, rat, and dog). The resulting empirically derived exposure-response continuum for PF-4778574 defines a single-dose-based TI of 8- to 16-fold for self-limiting tremor, a readily monitorable clinical adverse event. Importantly, the Cb,u mediating each physiological effect were highly consistent across species, with efficacy and convulsion occurring at just fractions of the in vitro-derived pharmacological values.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Receptors, AMPA/agonists , Receptors, AMPA/physiology , Thiophenes/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Cells, Cultured , Dogs , Excitatory Amino Acid Agonists/therapeutic use , Female , HEK293 Cells , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Seizures/physiopathology , Seizures/prevention & control , Thiophenes/therapeutic use , Treatment Outcome
20.
J Neurosci Methods ; 215(1): 97-102, 2013 Apr 30.
Article in English | MEDLINE | ID: mdl-23419700

ABSTRACT

Research into therapeutics for neuropsychiatric disorders is increasingly focusing on drugs with new mechanisms of action, and such agents are often assessed in preclinical studies using nonhuman primates. However, researchers lack a standardised method to compare different drugs for common adverse effects on the nervous system. We have developed a new scale for this purpose, named "Drug Effects on the Nervous System" (DENS), and tested its utility in an analysis of the second-generation antipsychotic risperidone in monkeys. The behavioural effects of risperidone over a ten-fold clinically relevant exposure range were rated with the DENS scale and compared with a standard motor disability scale for primates. The ratings were correlated with projected D2 and 5-HT2A receptor occupancies over time. The DENS scale detected dose-dependent side effects of risperidone in addition to the motor effects detected with the motor disability scale, including cognitive, sensorimotor and autonomic functions. A consistent temporal association between the DENS scale changes and the projected D2 receptor occupancy was observed, and the DENS scale ratings demonstrated high inter-rater reliability. These results demonstrate the usefulness of the DENS scale as a highly sensitive, reliable and accurate method to identify common adverse effects of risperidone and potentially other neurotropics for translational studies in primates.


Subject(s)
Nervous System/drug effects , Psychotropic Drugs/adverse effects , Algorithms , Animals , Antipsychotic Agents/adverse effects , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacokinetics , Autonomic Nervous System/drug effects , Basal Ganglia Diseases/physiopathology , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Dyskinesia, Drug-Induced/physiopathology , Female , Macaca fascicularis , Macaca mulatta , Male , Observer Variation , Psychotropic Drugs/metabolism , Psychotropic Drugs/pharmacokinetics , Receptors, Drug/metabolism , Reference Standards , Risperidone/adverse effects , Risperidone/metabolism , Risperidone/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL