Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Channels (Austin) ; 16(1): 230-243, 2022 12.
Article in English | MEDLINE | ID: mdl-36239534

ABSTRACT

As part of a drug discovery effort to identify potent inhibitors of NaV1.7 for the treatment of pain, we observed that inhibitors produced unexpected cardiovascular and respiratory effects in vivo. Specifically, inhibitors administered to rodents produced changes in cardiovascular parameters and respiratory cessation. We sought to determine the mechanism of the in vivo adverse effects by studying the selectivity of the compounds on NaV1.5, NaV1.4, and NaV1.6 in in vitro and ex vivo assays. Inhibitors lacking sufficient NaV1.7 selectivity over NaV1.6 were associated with respiratory cessation after in vivo administration to rodents. Effects on respiratory rate in rats were consistent with effects in an ex vivo hemisected rat diaphragm model and in vitro NaV1.6 potency. Furthermore, direct blockade of the phrenic nerve signaling was observed at exposures known to cause respiratory cessation in rats. Collectively, these results support a significant role for NaV1.6 in phrenic nerve signaling and respiratory function.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Respiratory Insufficiency , Animals , Pain , Phrenic Nerve , Rats , Respiratory Insufficiency/drug therapy
2.
J Med Chem ; 65(1): 485-496, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34931831

ABSTRACT

Inhibitor cystine knot peptides, derived from venom, have evolved to block ion channel function but are often toxic when dosed at pharmacologically relevant levels in vivo. The article describes the design of analogues of ProTx-II that safely display systemic in vivo blocking of Nav1.7, resulting in a latency of response to thermal stimuli in rodents. The new designs achieve a better in vivo profile by improving ion channel selectivity and limiting the ability of the peptides to cause mast cell degranulation. The design rationale, structural modeling, in vitro profiles, and rat tail flick outcomes are disclosed and discussed.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/drug effects , Pain/drug therapy , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/pharmacology , Spider Venoms/chemical synthesis , Animals , Cell Degranulation/drug effects , Cystine/chemistry , Drug Design , Hot Temperature , Mast Cells/drug effects , Models, Molecular , Pain Measurement/drug effects , Rats , Spider Venoms/pharmacology
3.
Neuropharmacology ; 197: 108754, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34389398

ABSTRACT

Alzheimer's disease (AD) is a profoundly debilitating neurodegenerative disorder characterized most notably by progressive cognitive decline, but also agitation and behavioral disturbances that are extremely disruptive to patient and caregiver. Current pharmacological treatments for these symptoms have limited efficacy and significant side effects. We have recently reported the discovery of Compound 24, an M4 positive allosteric modulator (PAM) that is potent, highly selective, and devoid of cholinergic-like side effects in rats. In order to further evaluate the translatability of the effects of compound 24 in primates, here we describe the effect of Compound 24 on three behavioral and cognition assays in rhesus monkeys, the stimulant induced motor activity (SIMA) assay, the object retrieval detour task (ORD), and the visuo-spatial paired-associates learning (vsPAL) task. As far as we know, this is the first such characterization of an M4 PAM in non-human primate. Compound 24 and the clinical standard olanzapine attenuated amphetamine induced hyperactivity to a similar degree. In addition, Compound 24 demonstrated procognitive effects in scopolamine-impaired ORD and vsPAL, and these effects were of similar magnitude to donepezil. These findings suggest that M4 PAMs may be beneficial to diseases such as Alzheimer's disease and schizophrenia, which are marked by behavioral disturbances as well as deficits in cognitive function.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Behavior, Animal/drug effects , Cholinergic Agents/pharmacology , Cognition Disorders/drug therapy , Receptor, Muscarinic M4/drug effects , Schizophrenia/drug therapy , Schizophrenic Psychology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Association Learning/drug effects , Central Nervous System Stimulants , Cholinergic Agents/pharmacokinetics , Cognition Disorders/psychology , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Macaca mulatta , Male , Motor Activity/drug effects , Olanzapine/pharmacology , Orientation/drug effects
4.
Sci Transl Med ; 13(594)2021 05 19.
Article in English | MEDLINE | ID: mdl-34011626

ABSTRACT

Humans with loss-of-function mutations in the Nav1.7 channel gene (SCN9A) show profound insensitivity to pain, whereas those with gain-of-function mutations can have inherited pain syndromes. Therefore, inhibition of the Nav1.7 channel with a small molecule has been considered a promising approach for the treatment of various human pain conditions. To date, clinical studies conducted using selective Nav1.7 inhibitors have not provided analgesic efficacy sufficient to warrant further investment. Clinical studies to date used multiples of in vitro IC50 values derived from electrophysiological studies to calculate anticipated human doses. To increase the chance of clinical success, we developed rhesus macaque models of action potential propagation, nociception, and olfaction, to measure Nav1.7 target modulation in vivo. The potent and selective Nav1.7 inhibitors SSCI-1 and SSCI-2 dose-dependently blocked C-fiber nociceptor conduction in microneurography studies and inhibited withdrawal responses to noxious heat in rhesus monkeys. Pharmacological Nav1.7 inhibition also reduced odor-induced activation of the olfactory bulb (OB), measured by functional magnetic resonance imaging (fMRI) studies consistent with the anosmia reported in Nav1.7 loss-of-function patients. These data demonstrate that it is possible to measure Nav1.7 target modulation in rhesus macaques and determine the plasma concentration required to produce a predetermined level of inhibition. The calculated plasma concentration for preclinical efficacy could be used to guide human efficacious exposure estimates. Given the translatable nature of the assays used, it is anticipated that they can be also used in phase 1 clinical studies to measure target modulation and aid in the interpretation of phase 1 clinical data.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Pain , Animals , Humans , Macaca mulatta , Nociception , Nociceptors
5.
Front Pharmacol ; 12: 786078, 2021.
Article in English | MEDLINE | ID: mdl-35002718

ABSTRACT

MK-2075 is a small-molecule selective inhibitor of the NaV1.7 channel investigated for the treatment of postoperative pain. A translational strategy was developed for MK-2075 to quantitatively interrelate drug exposure, target modulation, and the desired pharmacological response in preclinical animal models for the purpose of human translation. Analgesics used as a standard of care in postoperative pain were evaluated in preclinical animal models of nociceptive behavior (mouse tail flick latency and rhesus thermode heat withdrawal) to determine the magnitude of pharmacodynamic (PD) response at plasma concentrations associated with efficacy in the clinic. MK-2075 was evaluated in those same animal models to determine the concentration of MK-2075 required to achieve the desired level of response. Translation of MK-2075 efficacious concentrations in preclinical animal models to a clinical PKPD target in humans was achieved by accounting for species differences in plasma protein binding and in vitro potency against the NaV1.7 channel. Estimates of human pharmacokinetic (PK) parameters were obtained from allometric scaling of a PK model from preclinical species and used to predict the dose required to achieve the clinical exposure. MK-2075 exposure-response in a preclinical target modulation assay (rhesus olfaction) was characterized using a computational PKPD model which included a biophase compartment to account for the observed hysteresis. Translation of this model to humans was accomplished by correcting for species differences in PK NaV1.7 potency, and plasma protein binding while assuming that the kinetics of distribution to the target site is the same between humans and rhesus monkeys. This enabled prediction of the level of target modulation anticipated to be achieved over the dosing interval at the projected clinical efficacious human dose. Integration of these efforts into the early development plan informed clinical study design and decision criteria.

6.
Pharm Res ; 37(10): 181, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32888082

ABSTRACT

PURPOSE: This work describes a staged approach to the application of pharmacokinetic-pharmacodynamic (PK-PD) modeling in the voltage-gated sodium ion channel (NaV1.7) inhibitor drug discovery effort to address strategic questions regarding in vitro to in vivo translation of target modulation. METHODS: PK-PD analysis was applied to data from a functional magnetic resonance imaging (fMRI) technique to non-invasively measure treatment mediated inhibition of olfaction signaling in non-human primates (NHPs). Initial exposure-response was evaluated using single time point data pooled across 27 compounds to inform on in vitro to in vivo correlation (IVIVC). More robust effect compartment PK-PD modeling was conducted for a subset of 10 compounds with additional PD and PK data to characterize hysteresis. RESULTS: The pooled compound exposure-response facilitated an early exploration of IVIVC with a limited dataset for each individual compound, and it suggested a 2.4-fold in vitro to in vivo scaling factor for the NaV1.7 target. Accounting for hysteresis with an effect compartment PK-PD model as compounds advanced towards preclinical development provided a more robust determination of in vivo potency values, which resulted in a statistically significant positive IVIVC with a slope of 1.057 ± 0.210, R-squared of 0.7831, and p value of 0.006. Subsequent simulations with the PK-PD model informed the design of anti-nociception efficacy studies in NHPs. CONCLUSIONS: A staged approach to PK-PD modeling and simulation enabled integration of in vitro NaV1.7 potency, plasma protein binding, and pharmacokinetics to describe the exposure-response profile and inform future study design as the NaV1.7 inhibitor effort progressed through drug discovery.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Algorithms , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Cerebrovascular Circulation , Drug Design , Drug Discovery , HEK293 Cells , Humans , In Vitro Techniques , Macaca mulatta , Magnetic Resonance Imaging , Models, Biological , Smell/drug effects , Sodium Channel Blockers/pharmacokinetics
7.
Brain Res ; 1737: 146814, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32234514

ABSTRACT

Analgesic properties of orthosteric agonists of the muscarinic M4 receptor subtype have been documented in literature reports, with evidence from pharmacological and in vivo receptor knock out (KO) studies. Constitutive M4 receptor KO mice demonstrated an increased response in the formalin pain model, supporting this hypothesis. Two novel positive allosteric modulators (PAM) of the M4 receptor, Compounds 1 and 2, were characterized in rodent models of acute nociception. Results indicated decreased time spent on nociceptive behaviors in the mouse formalin model, and efficacy in the mouse tail flick assay. The analgesic-like effects of Compounds 1 and 2 were shown to be on target, as the compounds lacked any activity in constitutive M4 KO mice, while retaining activity in wild type control littermates. The analgesic-like effects of Compounds 1 and 2 were significantly diminished in KO mice that have selective deletion of the M4 receptor in neurons that co-express the dopaminergic D1 receptor subtype, suggesting a centrally-mediated effect on nociception. The opioid antagonist naloxone did not diminish the effect of Compound 1, indicating the effects of Compound 1 are not secondarily linked to opioid pathways. Compound 1 was evaluated in the rat, where it demonstrated analgesic-like effects in tail flick and a subpopulation of spinal nociceptive sensitive neurons, suggesting some involvement of spinal mechanisms of nociceptive modulation. These studies indicate that M4 PAMs may be a tractable target for pain management assuming an appropriate safety profile, and it appears likely that both spinal and supraspinal pathways may mediate the antinociceptive-like effects.


Subject(s)
Allosteric Regulation/drug effects , Nociception/drug effects , Receptor, Muscarinic M4/agonists , Allosteric Regulation/physiology , Analgesics/pharmacology , Analgesics, Opioid/pharmacology , Animals , Cholinergic Agents/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Narcotic Antagonists/pharmacology , Nociception/physiology , Pain/metabolism , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M4/drug effects , Receptor, Muscarinic M4/metabolism
8.
J Pain Res ; 11: 735-741, 2018.
Article in English | MEDLINE | ID: mdl-29692626

ABSTRACT

INTRODUCTION: The development of novel analgesics to treat acute or chronic pain has been a challenge due to a lack of translatable measurements. Preclinical end points with improved translatability are necessary to more accurately inform clinical testing paradigms, which may help guide selection of viable drug candidates. METHODS: In this study, a nonhuman primate biomarker which is sensitive to standard analgesics at clinically relevant plasma concentrations, can differentiate analgesia from sedation and utilizes a protocol very similar to that which can be employed in human clinical studies is described. Specifically, acute heat stimuli were delivered to the volar forearm using a contact heat thermode in the same manner as the clinical setting. RESULTS: Clinically efficacious exposures of morphine, fentanyl, and tramadol produced robust analgesic effects, whereas doses of diazepam that produce sedation had no effect. CONCLUSION: We propose that this assay has predictive utility that can help improve the probability of success for developing novel analgesics.

9.
Bioorg Med Chem Lett ; 27(12): 2695-2701, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28465100

ABSTRACT

A series of substituted indoles were examined as selective inhibitors of tropomyosin-related kinase receptor A (TrkA), a therapeutic target for the treatment of pain. An SAR optimization campaign based on ALIS screening lead compound 1 is reported.


Subject(s)
Drug Design , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor, trkA/metabolism , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 27(12): 2683-2688, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28465103

ABSTRACT

Studies on human genetics have suggested that inhibitors of the Nav1.7 voltage-gated sodium channel hold considerable promise as therapies for the treatment of chronic pain syndromes. Herein, we report novel, peripherally-restricted benzoxazolinone aryl sulfonamides as potent Nav1.7 inhibitors with excellent selectivity against the Nav1.5 isoform, which is expressed in the heart muscle. Elaboration of initial lead compound 3d afforded exemplar 13, which featured attractive physicochemical properties, outstanding lipophilic ligand efficiency and pharmacological selectivity against Nav1.5 exceeding 1000-fold. Key structure-activity relationships associated with oral bioavailability were leveraged to discover compound 17, which exhibited a comparable potency/selectivity profile as well as full efficacy following oral administration in a preclinical model indicative of antinociceptive behavior.


Subject(s)
Analgesics/pharmacology , Benzoxazoles/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Sulfonamides/pharmacology , Administration, Oral , Analgesics/administration & dosage , Analgesics/chemistry , Animals , Benzoxazoles/administration & dosage , Benzoxazoles/chemistry , Biological Availability , Disease Models, Animal , Dose-Response Relationship, Drug , Formaldehyde/administration & dosage , Humans , Mice , Molecular Structure , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pain/chemically induced , Rats , Structure-Activity Relationship , Sulfonamides/administration & dosage , Sulfonamides/chemistry
11.
J Neurosci Methods ; 282: 34-42, 2017 Apr 15.
Article in English | MEDLINE | ID: mdl-28279735

ABSTRACT

BACKGROUND: NGF signaling through TrkA triggers pathways involved in a wide range of biological effects. Clinical trials targeting either NGF or TrkA are ongoing to treat various diseases in the areas of oncology, neuroscience, and for pain, but there is no described measure of target engagement of TrkA in these studies. NEW METHOD: We have developed custom ELISA assays to measure NGF-induced phosphorylation of TrkA specific for rodent and human receptors. Optimized tissue processing methods allow for detection in both the brain and in skin. In addition, TrkB and TrkC assays have been in established to evaluate selectivity against other neurotrophin receptors. RESULTS: In a preclinical NGF-induced pain model, we show that pre-dosing with a TrkA inhibitor prevents phosphorylation of TrkA in the skin at a dose that is efficacious in reversal of thermal hypersensitivity. In addition, we show data in non-human primate and human skin supporting the potential use of this approach to enable translational target engagement. Comparison with existing methods: Existing methods involve animal models expressing TrkA tumors or injection of over-expressing TrkA recombinant cells into animals. Our method can measure target engagement in both normal and disease tissues in preclinical animal models and human skin. CONCLUSIONS: We have developed methods to assess target engagement for drug programs aimed at disrupting NGF-induced TrkA signaling. This includes preclinical determination of selectivity against other neurotrophin receptors and estimation of functional peripheral restriction. Preliminary data supports this method can be translated into a clinical pharmacodynamic readout using human skin biopsies.


Subject(s)
Analgesics/pharmacokinetics , Enzyme-Linked Immunosorbent Assay/methods , Nerve Growth Factor/metabolism , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Analgesics/pharmacology , Animals , Biomarkers, Pharmacological/metabolism , Biopsy , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Female , Humans , Macaca mulatta , Male , Middle Aged , Pain/drug therapy , Pain/metabolism , Phosphorylation/drug effects , Pilot Projects , Rats , Receptor, trkB/metabolism , Receptor, trkC/metabolism , Signal Transduction/drug effects , Skin/drug effects , Skin/metabolism
12.
Proc Natl Acad Sci U S A ; 114(3): E297-E306, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28039433

ABSTRACT

Current therapies for chronic pain can have insufficient efficacy and lead to side effects, necessitating research of novel targets against pain. Although originally identified as an oncogene, Tropomyosin-related kinase A (TrkA) is linked to pain and elevated levels of NGF (the ligand for TrkA) are associated with chronic pain. Antibodies that block TrkA interaction with its ligand, NGF, are in clinical trials for pain relief. Here, we describe the identification of TrkA-specific inhibitors and the structural basis for their selectivity over other Trk family kinases. The X-ray structures reveal a binding site outside the kinase active site that uses residues from the kinase domain and the juxtamembrane region. Three modes of binding with the juxtamembrane region are characterized through a series of ligand-bound complexes. The structures indicate a critical pharmacophore on the compounds that leads to the distinct binding modes. The mode of interaction can allow TrkA selectivity over TrkB and TrkC or promiscuous, pan-Trk inhibition. This finding highlights the difficulty in characterizing the structure-activity relationship of a chemical series in the absence of structural information because of substantial differences in the interacting residues. These structures illustrate the flexibility of binding to sequences outside of-but adjacent to-the kinase domain of TrkA. This knowledge allows development of compounds with specificity for TrkA or the family of Trk proteins.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Kinetics , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Models, Molecular , Protein Conformation , Protein Kinase Inhibitors/chemical synthesis , Receptor, trkA/genetics , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/chemistry , Receptor, trkB/genetics , Receptor, trkC/antagonists & inhibitors , Receptor, trkC/chemistry , Receptor, trkC/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Structure-Activity Relationship , Surface Plasmon Resonance
13.
J Biomol Screen ; 20(6): 708-19, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25700884

ABSTRACT

GluK1, a kainate subtype of ionotropic glutamate receptors, exhibits an expression pattern in the CNS consistent with involvement in pain processing and migraine. Antagonists of GluK1 have been shown to reduce pain signaling in the spinal cord and trigeminal nerve, and are predicted to provide pain and migraine relief. We developed an ultra-high-throughput small-molecule screen to identify antagonists of GluK1. Using the calcium indicator dye fluo-4, a multimillion-member small-molecule library was screened in 1536-well plate format on the FLIPR (Fluorescent Imaging Plate Reader) Tetra against cells expressing a calcium-permeable GluK1. Following confirmation in the primary assay and subsequent counter-screen against the endogenous Par-1 receptor, 6100 compounds were selected for dose titration to assess potency and selectivity. Final triage of 1000 compounds demonstrating dose-dependent inhibition with IC50 values of less than 12 µM was performed in an automated whole-cell patch clamp electrophysiology assay. Although a weak correlation between electrophysiologically active and calcium-imaging active compounds was observed, the identification of electrophysiologically active compounds with a range of kinetic profiles revealed a broad spectrum of mechanisms of action.


Subject(s)
Drug Discovery/methods , High-Throughput Screening Assays/methods , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Automation, Laboratory , Cell Line , Dose-Response Relationship, Drug , Humans , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Reproducibility of Results , Small Molecule Libraries
14.
J Med Chem ; 57(13): 5800-16, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24914455

ABSTRACT

We have identified several series of small molecule inhibitors of TrkA with unique binding modes. The starting leads were chosen to maximize the structural and binding mode diversity derived from a high throughput screen of our internal compound collection. These leads were optimized for potency and selectivity employing a structure based drug design approach adhering to the principles of ligand efficiency to maximize binding affinity without overly relying on lipophilic interactions. This endeavor resulted in the identification of several small molecule pan-Trk inhibitor series that exhibit high selectivity for TrkA/B/C versus a diverse panel of kinases. We have also demonstrated efficacy in both inflammatory and neuropathic pain models upon oral dosing. Herein we describe the identification process, hit-to-lead progression, and binding profiles of these selective pan-Trk kinase inhibitors.


Subject(s)
Chronic Pain/drug therapy , Protein Kinase Inhibitors/chemistry , Receptor, trkA/antagonists & inhibitors , Animals , Drug Evaluation, Preclinical , Humans , Indoles/chemistry , Indoles/pharmacokinetics , Ligands , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Rats , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Triazoles/chemistry , Triazoles/pharmacokinetics , Urea/analogs & derivatives , Urea/chemistry , Urea/pharmacokinetics
16.
Bioorg Med Chem Lett ; 21(8): 2359-64, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21420857

ABSTRACT

A novel series of decahydroquinoline CB2 agonists is described. Optimization of the amide substituent led to improvements in CB2/CB1 selectivity as well as physical properties. Two key compounds were examined in the rat CFA model of acute inflammatory pain. A moderately selective CB2 agonist was active in this model. A CB2 agonist lacking functional CB1 activity was inactive in this model despite high in vivo exposure both peripherally and centrally.


Subject(s)
Amides/chemistry , Analgesics/chemistry , Quinolines/chemistry , Receptor, Cannabinoid, CB2/agonists , Amides/chemical synthesis , Amides/therapeutic use , Analgesics/chemical synthesis , Analgesics/therapeutic use , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Pain/drug therapy , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 21(8): 2354-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21420860

ABSTRACT

A new series of imidazopyridine CB2 agonists is described. Structural optimization improved CB2/CB1 selectivity in this series and conferred physical properties that facilitated high in vivo exposure, both centrally and peripherally. Administration of a highly selective CB2 agonist in a rat model of analgesia was ineffective despite substantial CNS exposure, while administration of a moderately selective CB2/CB1 agonist exhibited significant analgesic effects.


Subject(s)
Analgesics/chemistry , Pyridines/chemistry , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists , Analgesics/chemical synthesis , Analgesics/therapeutic use , Animals , Disease Models, Animal , Freund's Adjuvant/pharmacology , Humans , Hyperalgesia/drug therapy , Pyridines/chemical synthesis , Pyridines/therapeutic use , Rats , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
18.
Bioorg Med Chem Lett ; 21(9): 2646-9, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21257308

ABSTRACT

The Merck Fragment Library was screened versus acid-sensing ion channel 3 (ASIC3), a novel target for the treatment of pain. Fragment hits were optimized using two strategies, and potency was improved from 0.7 mM to 3 µM with retention of good ligand efficiency and incorporation of reasonable physical properties, off-target profile, and rat pharmacokinetics.


Subject(s)
Drug Discovery , Electrophysiological Phenomena , Nerve Tissue Proteins/antagonists & inhibitors , Acid Sensing Ion Channels , Animals , Molecular Structure , Peptide Fragments , Rats , Small Molecule Libraries , Sodium Channels
20.
Mol Pain ; 4: 48, 2008 Oct 27.
Article in English | MEDLINE | ID: mdl-18954467

ABSTRACT

BACKGROUND: Safe and effective treatment for chronic inflammatory and neuropathic pain remains a key unmet medical need for many patients. The recent discovery and description of the transient receptor potential family of receptors including TRPV1 and TRPA1 has provided a number of potential new therapeutic targets for treating chronic pain. Recent reports have suggested that TRPA1 may play an important role in acute formalin and CFA induced pain. The current study was designed to further explore the therapeutic potential of pharmacological TRPA1 antagonism to treat inflammatory and neuropathic pain. RESULTS: The in vitro potencies of HC-030031 versus cinnamaldehyde or allyl isothiocyanate (AITC or Mustard oil)-induced TRPA1 activation were 4.9 +/- 0.1 and 7.5 +/- 0.2 microM respectively (IC50). These findings were similar to the previously reported IC50 of 6.2 microM against AITC activation of TRPA1 1. In the rat, oral administration of HC-030031 reduced AITC-induced nocifensive behaviors at a dose of 100 mg/kg. Moreover, oral HC-030031 (100 mg/kg) significantly reversed mechanical hypersensitivity in the more chronic models of Complete Freunds Adjuvant (CFA)-induced inflammatory pain and the spinal nerve ligation model of neuropathic pain. CONCLUSION: Using oral administration of the selective TRPA1 antagonist HC-030031, our results demonstrated that TRPA1 plays an important role in the mechanisms responsible for mechanical hypersensitivity observed in inflammatory and neuropathic pain models. These findings suggested that TRPA1 antagonism may be a suitable new approach for the development of a potent and selective therapeutic agent to treat both inflammatory and neuropathic pain.


Subject(s)
Acetanilides/pharmacology , Analgesics/pharmacology , Calcium Channels/physiology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Neuralgia/drug therapy , Pain/drug therapy , Purines/pharmacology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/physiology , Animals , Ankyrins , Cell Line , Disease Models, Animal , Humans , Inflammation , Male , Neuralgia/etiology , Neuralgia/pathology , Pain/etiology , Pain/pathology , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel , TRPC Cation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...