Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
J Mol Neurosci ; 74(1): 9, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38214771

ABSTRACT

Subarachnoid hemorrhage (SAH) due to the rupture of an intracranial aneurysm leads to delayed vasospasm and neuroischemia, which can result in profound neurologic deficit and death. Therapeutic options after SAH are currently limited to hemodynamic optimization and nimodipine, which have limited clinical efficacy. Experimental SAH results in cerebral vasospasm have demonstrated the downregulation of nitric oxide (NO)-protein kinase G (PKG) signaling elements. VP3 is a novel cell permeant phosphopeptide mimetic of VASP, a substrate of PKG and an actin-associated protein that modulates vasorelaxation in vascular smooth muscle cells. In this study, we determined that intravenous administration of high doses of VP3 did not induce systemic hypotension in rats except at the maximal soluble dose, implying that VP3 is well-tolerated and has a wide therapeutic window. Using a single cisterna magna injection rat model of SAH, we demonstrated that intravenous administration of low-dose VP3 after SAH improved neurologic deficits for up to 14 days as determined by the rotarod test. These findings suggest that strategies aimed at targeting the cerebral vasculature with VP3 may improve neurologic deficits associated with SAH.


Subject(s)
Subarachnoid Hemorrhage , Rats , Animals , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Nimodipine , Hemodynamics , Signal Transduction , Treatment Outcome , Disease Models, Animal
2.
Cancer Med ; 13(3): e6812, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38239047

ABSTRACT

BACKGROUND: It has been shown that tumor microenvironment (TME) hydroxyapatite (HAP) is typically associated with many malignancies and plays a role in tumor progression and growth. Additionally, acidosis in the TME has been reported to play a key role in selecting for a more aggressive tumor phenotype, drug resistance and desensitization to immunotherapy for many types of cancers. TME-HAP is an attractive target for tumor detection and treatment development since HAP is generally absent from normal soft tissue. We provide strong evidence that dissolution of hydroxyapatite (HAP) within the tumor microenvironment (TME-HAP) using a novel therapeutic can be used to kill cancer cells both in vitro and in vivo with minimal adverse effects. METHODS: We developed an injectable cation exchange nano particulate sulfonated polystyrene solution (NSPS) that we engineered to dissolve TME-HAP, inducing localized acute alkalosis and inhibition of tumor growth and glucose metabolism. This was evaluated in cell culture using 4T1, MDA-MB-231 triple negative breast cancer cells, MCF10 normal breast cells, and H292 lung cancer cells, and in vivo using orthotopic mouse models of cancer that contained detectable microenvironment HAP including breast (MMTV-Neu, 4T1, and MDA-MB-231), prostate (PC3) and colon (HCA7) cancer using 18 F-NaF for HAP and 18 F-FDG for glucose metabolism with PET imaging. On the other hand, H292 lung tumor cells that lacked detectable microenvironment HAP and MCF10a normal breast cells that do not produce HAP served as negative controls. Tumor microenvironment pH levels following injection of NSPS were evaluated via Chemical Exchange Saturation (CEST) MRI and via ex vivo methods. RESULTS: Within 24 h of adding the small concentration of 1X of NSPS (~7 µM), we observed significant tumor cell death (~ 10%, p < 0.05) in 4T1 and MDA-MB-231 cell cultures that contain HAP but ⟨2% in H292 and MCF10a cells that lack detectable HAP and in controls. Using CEST MRI, we found extracellular pH (pHe) in the 4T1 breast tumors, located in the mammary fat pad, to increase by nearly 10% from baseline before gradually receding back to baseline during the first hour post NSPS administration. in the tumors that contained TME-HAP in mouse models, MMTV-Neu, 4T1, and MDA-MB-231, PC3, and HCA7, there was a significant reduction (p<0.05) in 18 F-Na Fuptake post NSPS treatment as expected; 18 F- uptake in the tumor = 3.8 ± 0.5 %ID/g (percent of the injected dose per gram) at baseline compared to 1.8 ±0.5 %ID/g following one-time treatment with 100 mg/kg NSPS. Of similar importance, is that 18 F-FDG uptake in the tumors was reduced by more than 75% compared to baseline within 24 h of treatment with one-time NSPS which persisted for at least one week. Additionally, tumor growth was significantly slower (p < 0.05) in the mice treated with one-time NSPS. Toxicity showed no evidence of any adverse effects, a finding attributed to the absence of HAP in normal soft tissue and to our therapeutic NSPS having limited penetration to access HAP within skeletal bone. CONCLUSION: Dissolution of TME-HAP using our novel NSPS has the potential to provide a new treatment paradigm to enhance the management of cancer patients with poor prognosis.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Lung Neoplasms , Humans , Male , Animals , Mice , Pharmaceutical Preparations , Fluorodeoxyglucose F18 , Immunotherapy , Alkanesulfonates , Glucose , Hydroxyapatites , Tumor Microenvironment
3.
JACC Basic Transl Sci ; 8(10): 1379-1388, 2023 Oct.
Article in English | MEDLINE | ID: mdl-38094686

ABSTRACT

Ligands for the serotonin 2B receptor (5-HT2B) have shown potential to treat pulmonary arterial hypertension in preclinical models but cannot be used in humans because of predicted off-target neurological effects. The aim of this study was to develop novel systemically restricted compounds targeting 5-HT2B. Here, we show that mice treated with VU6047534 had decreased RVSP compared with control treatment in both the prevention and intervention studies using Sugen-hypoxia. VU6047534 is a novel 5-HT2B partial agonist that is peripherally restricted and able to both prevent and treat Sugen-hypoxia-induced pulmonary arterial hypertension. We have synthesized and characterized a structurally novel series of 5-HT2B ligands with high potency and selectivity for the 5-HT2B receptor subtype. Next-generation 5-HT2B ligands with similar characteristics, and predicted to be systemically restricted in humans, are currently advancing to investigational new drug-enabling studies.

4.
Mol Pharmacol ; 104(5): 195-202, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37595966

ABSTRACT

M4 muscarinic receptors are highly expressed in the striatum and cortex, brain regions that are involved in diseases such as Parkinson's disease, schizophrenia, and dystonia. Despite potential therapeutic advantages of specifically targeting the M4 receptor, it has been historically challenging to develop highly selective ligands, resulting in undesired off-target activity at other members of the muscarinic receptor family. Recently, we have reported first-in-class, potent, and selective M4 receptor antagonists. As an extension of that work, we now report the development and characterization of a radiolabeled M4 receptor antagonist, [3H]VU6013720, with high affinity (pKd of 9.5 ± 0.2 at rat M4, 9.7 at mouse M4, and 10 ± 0.1 at human M4 with atropine to define nonspecific binding) and no significant binding at the other muscarinic subtypes. Binding assays using this radioligand in rodent brain tissues demonstrate loss of specific binding in Chrm4 knockout animals. Dissociation kinetics experiments with various muscarinic ligands show differential effects on the dissociation of [3H]VU6013720 from M4 receptors, suggesting a binding site that is overlapping but may be distinct from the orthosteric site. Overall, these results demonstrate that [3H]VU6013720 is the first highly selective antagonist radioligand for the M4 receptor, representing a useful tool for studying the basic biology of M4 as well for the support of M4 receptor-based drug discovery. SIGNIFICANCE STATEMENT: This manuscript describes the development and characterization of a novel muscarinic (M) acetylcholine subtype 4 receptor antagonist radioligand, [3H]VU6013720. This ligand binds to or overlaps with the acetylcholine binding site, providing a highly selective radioligand for the M4 receptor that can be used to quantify M4 protein expression in vivo and probe the selective interactions of acetylcholine with M4 versus the other members of the muscarinic receptor family.


Subject(s)
Acetylcholine , Receptors, Muscarinic , Rats , Humans , Mice , Animals , Acetylcholine/metabolism , Receptors, Muscarinic/metabolism , Receptor, Muscarinic M4/metabolism , Atropine , Ligands , Cholinergic Agents , Muscarinic Antagonists/pharmacology , Muscarinic Antagonists/metabolism , Receptor, Muscarinic M2/metabolism , Radioligand Assay , Receptor, Muscarinic M1/metabolism
5.
Bioorg Med Chem Lett ; 56: 128479, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34838649

ABSTRACT

In this manuscript, we report a series of chiral 6-azaspiro[2.5]octanes and related spirocycles as highly potent and selective antagonists of the muscarinic acetylcholine receptor subtype 4 (mAChR4). Chiral separation and subsequent X-ray crystallographic analysis of early generation analogs revealed the R enantiomer to possess excellent human and rat M4 potency, and further structure-activity relationship (SAR) studies on this chiral scaffold led to the discovery of VU6015241 (compound 19). Compound 19 is characterized by high M4 potency and selectivity across multiple species, excellent aqueous solubility, and moderate brain exposure in rodents after intraperitoneal administration.


Subject(s)
Muscarinic Antagonists/pharmacology , Receptor, Muscarinic M4/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Muscarinic Antagonists/chemical synthesis , Muscarinic Antagonists/chemistry , Receptor, Muscarinic M4/metabolism , Structure-Activity Relationship
6.
ACS Med Chem Lett ; 12(8): 1342-1349, 2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34413964

ABSTRACT

Herein, we report the SAR leading to the discovery of VU6028418, a potent M4 mAChR antagonist with high subtype-selectivity and attractive DMPK properties in vitro and in vivo across multiple species. VU6028418 was subsequently evaluated as a preclinical candidate for the treatment of dystonia and other movement disorders. During the characterization of VU6028418, a novel use of deuterium incorporation as a means to modulate CYP inhibition was also discovered.

7.
ACS Pharmacol Transl Sci ; 4(4): 1306-1321, 2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34423268

ABSTRACT

Nonselective antagonists of muscarinic acetylcholine receptors (mAChRs) that broadly inhibit all five mAChR subtypes provide an efficacious treatment for some movement disorders, including Parkinson's disease and dystonia. Despite their efficacy in these and other central nervous system disorders, antimuscarinic therapy has limited utility due to severe adverse effects that often limit their tolerability by patients. Recent advances in understanding the roles that each mAChR subtype plays in disease pathology suggest that highly selective ligands for individual subtypes may underlie the antiparkinsonian and antidystonic efficacy observed with the use of nonselective antimuscarinic therapeutics. Our recent work has indicated that the M4 muscarinic acetylcholine receptor has several important roles in opposing aberrant neurotransmitter release, intracellular signaling pathways, and brain circuits associated with movement disorders. This raises the possibility that selective antagonists of M4 may recapitulate the efficacy of nonselective antimuscarinic therapeutics and may decrease or eliminate the adverse effects associated with these drugs. However, this has not been directly tested due to lack of selective antagonists of M4. Here, we utilize genetic mAChR knockout animals in combination with nonselective mAChR antagonists to confirm that the M4 receptor activation is required for the locomotor-stimulating and antiparkinsonian efficacy in rodent models. We also report the synthesis, discovery, and characterization of the first-in-class selective M4 antagonists VU6013720, VU6021302, and VU6021625 and confirm that these optimized compounds have antiparkinsonian and antidystonic efficacy in pharmacological and genetic models of movement disorders.

8.
Bioorg Med Chem Lett ; 37: 127838, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33556572

ABSTRACT

A high throughput screen (HTS) identified a novel, but weak (EC50 = 6.2 µM, 97% Glu Max) mGlu4 PAM chemotype based on a 1,4-thiazepane core, VU0544412. Reaction development and chemical optimization delivered a potent mGlu4 PAM VU6022296 (EC50 = 32.8 nM, 108% Glu Max) with good CNS penetration (Kp = 0.45, Kp,uu = 0.70) and enantiopreference. Finally, VU6022296 displayed robust, dose-dependent efficacy in reversing Haloperidol-Induced Catalepsy (HIC), a rodent preclinical Parkinson's disease model.


Subject(s)
Catalepsy/drug therapy , Disease Models, Animal , Drug Discovery , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Catalepsy/chemically induced , Dose-Response Relationship, Drug , Haloperidol , Mice , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
9.
JCI Insight ; 6(4)2021 02 22.
Article in English | MEDLINE | ID: mdl-33476302

ABSTRACT

The metabotropic glutamate receptor 7 (mGlu7) is a G protein-coupled receptor that has been recently linked to neurodevelopmental disorders. This association is supported by the identification of GRM7 variants in patients with autism spectrum disorder, attention deficit hyperactivity disorder, and severe developmental delay. One GRM7 mutation previously reported in 2 patients results in a single amino acid change, I154T, within the mGlu7 ligand-binding domain. Here, we report 2 new patients with this mutation who present with severe developmental delay and epilepsy. Functional studies of the mGlu7-I154T mutant reveal that this substitution resulted in significant loss of mGlu7 protein expression in HEK293A cells and in mice. We show that this occurred posttranscriptionally at the level of protein expression and trafficking. Similar to mGlu7-global KO mice, mGlu7-I154T animals exhibited reduced motor coordination, deficits in contextual fear learning, and seizures. This provides functional evidence that a disease-associated mutation affecting the mGlu7 receptor was sufficient to cause neurological dysfunction in mice and further validates GRM7 as a disease-causing gene in the human population.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease/genetics , Phenotype , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Animals , Child , Child, Preschool , Epilepsy , Fear , Female , GTP-Binding Proteins , Humans , Infant , Learning , Male , Mice , Mice, Inbred C57BL , Mutation , Neurodevelopmental Disorders/genetics , Pedigree , Seizures
10.
ACS Bio Med Chem Au ; 1(1): 21-30, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-37101980

ABSTRACT

In the course of optimizing an M1 PAM chemotype, introduction of an ether moiety unexpectedly abolished M1 PAM activity while engendering a "molecular switch" to afford a weak, pure mGlu5 PAM. Further optimization was able to deliver a potent (mGlu5 EC50 = 520 nM, 63% Glu Max), centrally penetrant (Kp = 0.83), MPEP-site binding mGlu5 PAM 17a (VU6036486) that reversed amphetamine-induced hyperlocomotion. A pronounced "magic methyl" effect was noted with a regioisomeric methyl congener, leading to a change in pharmacology to afford a potent (mGlu5 IC50 = 110 nM, 3% Glu Min), centrally penetrant (Kp = 0.94), MPEP-site binding NAM 28d (VU6044766) that displayed anxiolytic activity in a mouse marble burying assay. These data further support the growing body of literature concerning the existence of G protein-coupled receptor (GPCR) allosteric privileged structures, and the value and impact of subtle methyl group walks, as well as the highly productive fluorine walk, around allosteric ligand cores to stabilize unique GPCR conformations.

11.
Sci Signal ; 12(610)2019 12 03.
Article in English | MEDLINE | ID: mdl-31796631

ABSTRACT

Highly selective, positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach to potentially improve cognitive function in patients suffering from Alzheimer's disease and schizophrenia. Discovery programs have produced a structurally diverse range of M1 receptor PAMs with distinct pharmacological properties, including different extents of agonist activity and differences in signal bias. This includes biased M1 receptor PAMs that can potentiate coupling of the receptor to activation of phospholipase C (PLC) but not phospholipase D (PLD). However, little is known about the role of PLD in M1 receptor signaling in native systems, and it is not clear whether biased M1 PAMs display differences in modulating M1-mediated responses in native tissue. Using PLD inhibitors and PLD knockout mice, we showed that PLD was necessary for the induction of M1-dependent long-term depression (LTD) in the prefrontal cortex (PFC). Furthermore, biased M1 PAMs that did not couple to PLD not only failed to potentiate orthosteric agonist-induced LTD but also blocked M1-dependent LTD in the PFC. In contrast, biased and nonbiased M1 PAMs acted similarly in potentiating M1-dependent electrophysiological responses that were PLD independent. These findings demonstrate that PLD plays a critical role in the ability of M1 PAMs to modulate certain central nervous system (CNS) functions and that biased M1 PAMs function differently in brain regions implicated in cognition.


Subject(s)
Cerebral Cortex/enzymology , Neuronal Plasticity , Phospholipase D/genetics , Phospholipase D/metabolism , Receptor, Muscarinic M1/genetics , Receptor, Muscarinic M1/metabolism , Allosteric Site , Animals , CHO Cells , Calcium/chemistry , Cognition , Cricetinae , Cricetulus , Electrophysiology , Female , Humans , Long-Term Synaptic Depression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prefrontal Cortex/enzymology , Signal Transduction , Type C Phospholipases/metabolism
12.
ACS Chem Neurosci ; 10(3): 1035-1042, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30086237

ABSTRACT

This Letter describes the chemical optimization of a new series of muscarinic acetylcholine receptor subtype 1 (M1) positive allosteric modulators (PAMs) based on novel tricyclic triazolo- and imidazopyridine lactam cores, devoid of M1 agonism, e.g., no M1 ago-PAM activity, in high expressing recombinant cell lines. While all the new tricyclic congeners afforded excellent rat pharmacokinetic (PK) properties (CLp < 8 mL/min/kg and t1/2 > 5 h), regioisomeric triazolopyridine analogues were uniformly not CNS penetrant ( Kp < 0.05), despite a lack of hydrogen bond donors. However, removal of a single nitrogen atom to afford imidazopyridine derivatives proved to retain the excellent rat PK and provide high CNS penetration ( Kp > 2), despite inclusion of a basic nitrogen. Moreover, 24c was devoid of M1 agonism in high expressing recombinant cell lines and did not induce cholinergic seizures in vivo in mice. Interestingly, all of the new M1 PAMs across the diverse tricyclic heterocyclic cores possessed equivalent CNS MPO scores (>4.5), highlighting the value of both "medicinal chemist's eye" and experimental data, e.g., not sole reliance (or decision bias) on in silico calculated properties, for parameters as complex as CNS penetration.


Subject(s)
Drug Discovery/methods , Imidazoles/pharmacology , Lactams/pharmacology , Muscarinic Agonists/pharmacology , Pyridines/pharmacology , Receptor, Muscarinic M1/agonists , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Drug Discovery/trends , Humans , Imidazoles/chemistry , Lactams/chemistry , Mice , Muscarinic Agonists/chemistry , Pyridines/chemistry , Rats , Receptor, Muscarinic M1/physiology
13.
ACS Med Chem Lett ; 9(9): 917-922, 2018 Sep 13.
Article in English | MEDLINE | ID: mdl-30258541

ABSTRACT

Herein, we report the chemical optimization of a new series of M1 positive allosteric modulators (PAMs) based on a novel pyrrolo[2,3-b]pyridine core, developed via scaffold hopping and iterative parallel synthesis. The vast majority of analogs in this series proved to display robust cholinergic seizure activity. However, by removal of the secondary hydroxyl group, VU6007477 resulted with good rat M1 PAM potency (EC50 = 230 nM, 93% ACh max), minimal M1 agonist activity (agonist EC50 > 10 µM), good CNS penetration (rat brain/plasma K p = 0.28, K p,uu = 0.32; mouse K p = 0.16, K p,uu = 0.18), and no cholinergic adverse events (AEs, e.g., seizures). This work demonstrates that within a chemical series prone to robust M1 ago-PAM activity, SAR can result, which affords pure M1 PAMs, devoid of cholinergic toxicity/seizure liability.

14.
Psychopharmacology (Berl) ; 235(10): 2897-2913, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30054675

ABSTRACT

RATIONALE: Metabotropic glutamate receptors and muscarinic M4 receptors have been proposed as novel targets for various brain disorders, including schizophrenia. Both receptors are coupled to Go/i proteins and are expressed in brain circuits that are important in schizophrenia. Therefore, their mutual activation may be an effective treatment and allow minimizing the doses of ligands required for optimal activity. OBJECTIVES: In the present studies, subactive doses of mGlu4 and M4 activators (LSP4-2022 and VU152100, respectively) were administered to investigate the mutual interaction between mGlu4 and M4 receptors in animal models of schizophrenia. METHODS: The behavioral tests used were MK-801-induced hyperactivity, (±)-2.5-dimethoxy-4-iodoamphetamine hydrochloride (DOI)-induced head twitches, the modified forced swim test, and MK-801-induced disruptions of social interactions and novel object recognition. DOI-induced spontaneous excitatory postsynaptic currents (sEPSCs) in brain slices and positron emission tomography (PET) in were used to establish the ability of these compounds to modulate the glutamatergic and dopaminergic systems. Rotarod was used to assess putative adverse effects. RESULTS: The mutual administration of subactive doses of LSP4-2022 and VU152100 exerted similar antipsychotic-like efficacy in animals as observed for active doses of both compounds, indicating their additive actions. VU152100 inhibited the DOI-induced frequency (but not amplitude) of sEPSCs in the frontal cortex, confirming presynaptic regulation of glutamate release. Both compounds reversed amphetamine-induced decrease in D2 receptor levels in the striatum, as measured with [18F]fallypride. The compounds did not induce any motor impartments when measured in rotarod test. CONCLUSIONS: Based on our results, the simultaneous activation of M4 and mGlu4 receptors is beneficial in reversing MK-801- and amphetamine-induced schizophrenia-related changes in animals.


Subject(s)
Antipsychotic Agents/therapeutic use , Receptor, Muscarinic M4/metabolism , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Amphetamine/toxicity , Animals , Antipsychotic Agents/pharmacology , Disease Models, Animal , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Agonists/therapeutic use , Male , Mice , Motor Activity/drug effects , Motor Activity/physiology , Phosphinic Acids/pharmacology , Phosphinic Acids/therapeutic use , Receptor, Muscarinic M4/agonists , Receptors, Metabotropic Glutamate/agonists , Rodentia , Schizophrenia/chemically induced
15.
Bioorg Med Chem Lett ; 28(12): 2175-2179, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29754948

ABSTRACT

This letter describes the chemical optimization of a new series of M1 positive allosteric modulators (PAMs) based on a novel benzomorpholine core, developed via iterative parallel synthesis, and culminating in the highly utilized rodent in vivo tool compound, VU0486846 (7), devoid of adverse effect liability. This is the first report of the optimization campaign (SAR and DMPK profiling) that led to the discovery of VU0486846 and details all of the challenges faced in allosteric modulator programs (both steep and flat SAR, as well as subtle structural changes affecting CNS penetration and overall physiochemical and DMPK properties).


Subject(s)
Drug Discovery , Morpholines/pharmacology , Pyrazoles/pharmacology , Receptor, Muscarinic M1/agonists , Animals , CHO Cells , Cricetulus , Humans , Molecular Structure , Morpholines/chemical synthesis , Morpholines/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Rats , Structure-Activity Relationship
16.
ACS Chem Neurosci ; 9(9): 2218-2224, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29683646

ABSTRACT

Positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have attracted intense interest as an exciting new approach for improving the cognitive deficits in schizophrenia and Alzheimer's disease. Recent evidence suggests that the presence of intrinsic agonist activity of some M1 PAMs may reduce efficacy and contribute to adverse effect liability. However, the M1 PAM PF-06827443 was reported to have only weak agonist activity at human M1 receptors but produced M1-dependent adverse effects. We now report that PF-06827443 is an allosteric agonist in cell lines expressing rat, dog, and human M1 and use of inducible cell lines shows that agonist activity of PF-06827443 is dependent on receptor reserve. Furthermore, PF-06827443 is an agonist in native tissue preparations and induces behavioral convulsions in mice similar to other ago-PAMs. These findings suggest that PF-06827443 is a robust ago-PAM, independent of species, in cell lines and native systems.


Subject(s)
Isoindoles/pharmacology , Oxazoles/pharmacology , Prefrontal Cortex/drug effects , Receptor, Muscarinic M1/agonists , Seizures/chemically induced , Seizures/metabolism , Allosteric Regulation , Animals , CHO Cells , Calcium/metabolism , Cricetulus , Dogs , Humans , Mice , Patch-Clamp Techniques , Prefrontal Cortex/metabolism , Rats
17.
ACS Chem Neurosci ; 9(9): 2274-2285, 2018 09 19.
Article in English | MEDLINE | ID: mdl-29701957

ABSTRACT

Selective activation of the M1 subtype of muscarinic acetylcholine receptor, via positive allosteric modulation (PAM), is an exciting strategy to improve cognition in schizophrenia and Alzheimer's disease patients. However, highly potent M1 ago-PAMs, such as MK-7622, PF-06764427, and PF-06827443, can engender excessive activation of M1, leading to agonist actions in the prefrontal cortex (PFC) that impair cognitive function, induce behavioral convulsions, and result in other classic cholinergic adverse events (AEs). Here, we report a fundamentally new and highly selective M1 PAM, VU0486846. VU0486846 possesses only weak agonist activity in M1-expressing cell lines with high receptor reserve and is devoid of agonist actions in the PFC, unlike previously reported ago-PAMs MK-7622, PF-06764427, and PF-06827443. Moreover, VU0486846 shows no interaction with antagonist binding at the orthosteric acetylcholine (ACh) site (e.g., neither bitopic nor displaying negative cooperativity with [3H]-NMS binding at the orthosteric site), no seizure liability at high brain exposures, and no cholinergic AEs. However, as opposed to ago-PAMs, VU0486846 produces robust efficacy in the novel object recognition model of cognitive function. Importantly, we show for the first time that an M1 PAM can reverse the cognitive deficits induced by atypical antipsychotics, such as risperidone. These findings further strengthen the argument that compounds with modest in vitro M1 PAM activity (EC50 > 100 nM) and pure-PAM activity in native tissues display robust procognitive efficacy without AEs mediated by excessive activation of M1. Overall, the combination of compound assessment with recombinant in vitro assays (mindful of receptor reserve), native tissue systems (PFC), and phenotypic screens (behavioral convulsions) is essential to fully understand and evaluate lead compounds and enhance success in clinical development.


Subject(s)
Cognition/drug effects , Conditioning, Psychological/drug effects , Exploratory Behavior/drug effects , Morpholines/pharmacology , Prefrontal Cortex/drug effects , Pyrazoles/pharmacology , Allosteric Regulation , Animals , Antipsychotic Agents/toxicity , CHO Cells , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/physiopathology , Cricetulus , Fear , Mice , Morpholines/toxicity , Pyrazoles/toxicity , Rats , Risperidone/toxicity , Seizures/chemically induced
18.
Neuropsychopharmacology ; 43(10): 2075-2082, 2018 09.
Article in English | MEDLINE | ID: mdl-29654259

ABSTRACT

Glutamatergic transmission in the nucleus accumbens shell (NAcSh) is a substrate for reward learning and motivation. Metabotropic glutamate (mGlu) receptors regulate NAcSh synaptic strength by inducing long-term depression (LTD). Inputs from prefrontal cortex (PFC) and medio-dorsal thalamus (MDT) drive opposing motivated behaviors yet mGlu receptor regulation of these synapses is unexplored. We examined Group I mGlu receptor regulation of PFC and MDT glutamatergic synapses onto specific populations of NAc medium spiny neurons (MSNs) using D1tdTom BAC transgenic mice and optogenetics. Synaptically evoked long-term depression (LTD) at MDT-NAcSh synapses required mGlu5 but not mGlu1 and was specific for D1(+) MSNs, whereas PFC LTD was expressed at both D1(+) and D1(-) MSNs and required mGlu1 but not mGlu5. Two weeks after five daily non-contingent cocaine exposures (15 mg/kg), LTD was attenuated at MDT-D1(+) synapses but was rescued by the mGlu5-positive allosteric modulator (PAM) VU0409551. These results highlight unique plasticity mechanisms regulating specific NAcSh synapses.


Subject(s)
Nucleus Accumbens/physiology , Receptor, Metabotropic Glutamate 5/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Cocaine/pharmacology , Female , Mediodorsal Thalamic Nucleus/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Neuronal Plasticity/drug effects , Neurons/physiology , Optogenetics , Oxazoles/pharmacology , Prefrontal Cortex/physiology , Pyridines/pharmacology , Synapses/physiology , Synaptic Transmission
19.
Neuropsychopharmacology ; 43(8): 1763-1771, 2018 07.
Article in English | MEDLINE | ID: mdl-29581537

ABSTRACT

Highly selective positive allosteric modulators (PAMs) of the M1 subtype of muscarinic acetylcholine receptor have emerged as an exciting new approach for improving cognitive function in patients suffering from Alzheimer's disease and schizophrenia. However, excessive activation of M1 is known to induce seizure activity and have actions in the prefrontal cortex (PFC) that could impair cognitive function. We now report a series of pharmacological, electrophysiological, and behavioral studies in which we find that recently reported M1 PAMs, PF-06764427 and MK-7622, have robust agonist activity in cell lines and agonist effects in the mouse PFC, and have the potential to overactivate the M1 receptor and disrupt PFC function. In contrast, structurally distinct M1 PAMs (VU0453595 and VU0550164) are devoid of agonist activity in cell lines and maintain activity dependence of M1 activation in the PFC. Consistent with the previously reported effect of PF-06764427, the ago-PAM MK-7622 induces severe behavioral convulsions in mice. In contrast, VU0453595 does not induce behavioral convulsions at doses well above those required for maximal efficacy in enhancing cognitive function. Furthermore, in contrast to the robust efficacy of VU0453595, the ago-PAM MK-7622 failed to improve novel object recognition, a rodent assay of cognitive function. These findings suggest that in vivo cognition-enhancing efficacy of M1 PAMs can be observed with PAMs lacking intrinsic agonist activity and that intrinsic agonist activity of M1 PAMs may contribute to adverse effects and reduced efficacy in improving cognitive function.


Subject(s)
Cholinergic Agents/pharmacology , Nootropic Agents/pharmacology , Receptor, Muscarinic M1/metabolism , Allosteric Regulation , Animals , CHO Cells , Cricetulus , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Male , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/genetics , Recognition, Psychology/drug effects , Tissue Culture Techniques
20.
ACS Med Chem Lett ; 8(12): 1326-1330, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29259756

ABSTRACT

Herein, we report the structure-activity relationships within a series of mGlu7 NAMs based on an N-(2-(1H-1,2,4-triazol-1-yl)-5-(trifluoromethoxy)phenyl)benzamide core with excellent CNS penetration (Kp 1.9-5.8 and Kp,uu 0.4-1.4). Analogues in this series displayed steep SAR. Of these, VU6010608 (11a) emerged with robust efficacy in blocking high frequency stimulated long-term potentiation in electrophysiology studies.

SELECTION OF CITATIONS
SEARCH DETAIL
...