Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Med Teach ; 44(1): 3-18, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34666584

ABSTRACT

INTRODUCTION: Little is known of processes by which feedback affects learners to influence achievement. This review maps what is known of how learners interact with feedback, to better understand how feedback affects learning strategies, and to explore enhancing and inhibiting factors. METHODS: Pilot searching indicated a wide range of interpretations of feedback and study designs, prompting the use of scoping methodology. Inclusion criteria comprised: (i) learners (undergraduate, postgraduate, continuing education) who regularly receive feedback, and (ii) studies that associated feedback with subsequent learner reaction. The screening was performed independently in duplicate. Data extraction and synthesis occurred via an iterative consensus approach. Self-regulatory learning theory (SRL) was used as the conceptual framework. RESULTS: Of 4253 abstracts reviewed, 232 were included in the final synthesis. Understandings of feedback are diverse; a minority adopt recognised definitions. Distinct learner responses to feedback can be categorized as cognitive, behavioural, affective, and contextual with complex, overlapping interactions. Importantly emotional responses are commonplace; factors mediating them are pivotal in learner recipience. CONCLUSION: Feedback benefits learners most when focussed on learner needs, via engagement in bi-directional dialogue. Learner emotions must be supported, with the construction of positive learner-teacher relationships. A developmental agenda is key to learner's acceptance of feedback and enhancing future learning.


Subject(s)
Students, Medical , Feedback , Humans , Learning , Students, Medical/psychology
2.
ACS Med Chem Lett ; 12(4): 593-602, 2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33859800

ABSTRACT

The gene KCNT1 encodes the sodium-activated potassium channel KNa1.1 (Slack, Slo2.2). Variants in the KCNT1 gene induce a gain-of-function (GoF) phenotype in ionic currents and cause a spectrum of intractable neurological disorders in infants and children, including epilepsy of infancy with migrating focal seizures (EIMFS) and autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE). Effective treatment options for KCNT1-related disease are absent, and novel therapies are urgently required. We describe the development of a novel class of oxadiazole KNa1.1 inhibitors, leading to the discovery of compound 31 that reduced seizures and interictal spikes in a mouse model of KCNT1 GoF.

3.
ACS Chem Neurosci ; 11(9): 1311-1323, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32212718

ABSTRACT

We aimed to develop radioligands for PET imaging of brain phosphodiesterase subtype 4D (PDE4D), a potential target for developing cognition enhancing or antidepressive drugs. Exploration of several chemical series gave four leads with high PDE4D inhibitory potency and selectivity, optimal lipophilicity, and good brain uptake. These leads featured alkoxypyridinyl cores. They were successfully labeled with carbon-11 (t1/2 = 20.4 min) for evaluation with PET in monkey. Whereas two of these radioligands did not provide PDE4D-specific signal in monkey brain, two others, [11C]T1660 and [11C]T1650, provided sizable specific signal, as judged by pharmacological challenge using rolipram or a selective PDE4D inhibitor (BPN14770) and subsequent biomathematical analysis. Specific binding was highest in prefrontal cortex, temporal cortex, and hippocampus, regions that are important for cognitive function. [11C]T1650 was progressed to evaluation in humans with PET, but the output measure of brain enzyme density (VT) increased with scan duration. This instability over time suggests that radiometabolite(s) were accumulating in the brain. BPN14770 blocked PDE4D uptake in human brain after a single dose, but the percentage occupancy was difficult to estimate because of the unreliability of measuring VT. Overall, these results show that imaging of PDE4D in primate brain is feasible but that further radioligand refinement is needed, most likely to avoid problematic radiometabolites.


Subject(s)
Brain , Positron-Emission Tomography , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Radiopharmaceuticals , Rolipram/pharmacology
4.
Bioorg Med Chem Lett ; 27(21): 4805-4811, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29029933

ABSTRACT

The discovery and selection of a highly potent and selective NaV1.7 inhibitor PF-06456384, designed specifically for intravenous infusion, is disclosed. Extensive in vitro pharmacology and ADME profiling followed by in vivo preclinical PK and efficacy model data are discussed. A proposed protein-ligand binding mode for this compound is also provided to rationalise the high levels of potency and selectivity over inhibition of related sodium channels. To further support the proposed binding mode, potent conjugates are described which illustrate the potential for development of chemical probes to enable further target evaluation.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/chemistry , Piperidines/chemistry , Pyridines/chemistry , Sulfonamides/chemistry , Voltage-Gated Sodium Channel Blockers/chemistry , Animals , Binding Sites , Dogs , Half-Life , Hepatocytes/metabolism , Humans , Infusions, Intravenous , Inhibitory Concentration 50 , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/drug therapy , Pain/pathology , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Protein Binding , Protein Structure, Tertiary , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Rats , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Thiadiazoles , Voltage-Gated Sodium Channel Blockers/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/therapeutic use
5.
J Med Chem ; 60(16): 7029-7042, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28682065

ABSTRACT

A series of acidic diaryl ether heterocyclic sulfonamides that are potent and subtype selective NaV1.7 inhibitors is described. Optimization of early lead matter focused on removal of structural alerts, improving metabolic stability and reducing cytochrome P450 inhibition driven drug-drug interaction concerns to deliver the desired balance of preclinical in vitro properties. Concerns over nonmetabolic routes of clearance, variable clearance in preclinical species, and subsequent low confidence human pharmacokinetic predictions led to the decision to conduct a human microdose study to determine clinical pharmacokinetics. The design strategies and results from preclinical PK and clinical human microdose PK data are described leading to the discovery of the first subtype selective NaV1.7 inhibitor clinical candidate PF-05089771 (34) which binds to a site in the voltage sensing domain.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel/metabolism , Phenyl Ethers/pharmacology , Sulfonamides/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Cell Line , Cytochrome P-450 CYP2C9/metabolism , Cytochrome P-450 CYP2C9 Inhibitors/chemical synthesis , Cytochrome P-450 CYP2C9 Inhibitors/chemistry , Cytochrome P-450 CYP2C9 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP2C9 Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/chemical synthesis , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Design , Humans , Microsomes, Liver/metabolism , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Phenyl Ethers/chemical synthesis , Phenyl Ethers/chemistry , Phenyl Ethers/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Voltage-Gated Sodium Channel Blockers/chemical synthesis , Voltage-Gated Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channel Blockers/pharmacokinetics
6.
ACS Med Chem Lett ; 8(6): 666-671, 2017 Jun 08.
Article in English | MEDLINE | ID: mdl-28626530

ABSTRACT

A series of TRPA1 antagonists is described which has as its core structure an indazole moiety. The physical properties and in vitro DMPK profiles are discussed. Good in vivo exposure was obtained with several analogs, allowing efficacy to be assessed in rodent models of inflammatory pain. Two compounds showed significant activity in these models when administered either systemically or topically. Protein chimeras were constructed to indicate compounds from the series bound in the S5 region of the channel, and a computational docking model was used to propose a binding mode for example compounds.

7.
PLoS One ; 11(4): e0152405, 2016.
Article in English | MEDLINE | ID: mdl-27050761

ABSTRACT

Human genetic studies show that the voltage gated sodium channel 1.7 (Nav1.7) is a key molecular determinant of pain sensation. However, defining the Nav1.7 contribution to nociceptive signalling has been hampered by a lack of selective inhibitors. Here we report two potent and selective arylsulfonamide Nav1.7 inhibitors; PF-05198007 and PF-05089771, which we have used to directly interrogate Nav1.7's role in nociceptor physiology. We report that Nav1.7 is the predominant functional TTX-sensitive Nav in mouse and human nociceptors and contributes to the initiation and the upstroke phase of the nociceptor action potential. Moreover, we confirm a role for Nav1.7 in influencing synaptic transmission in the dorsal horn of the spinal cord as well as peripheral neuropeptide release in the skin. These findings demonstrate multiple contributions of Nav1.7 to nociceptor signalling and shed new light on the relative functional contribution of this channel to peripheral and central noxious signal transmission.


Subject(s)
Axons/physiology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Presynaptic Terminals/physiology , Action Potentials , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , HEK293 Cells , Humans , Male , Mice , NAV1.7 Voltage-Gated Sodium Channel/physiology , Patch-Clamp Techniques , Phenyl Ethers/pharmacology , Sulfonamides/pharmacology
8.
Channels (Austin) ; 9(6): 360-6, 2015.
Article in English | MEDLINE | ID: mdl-26646477

ABSTRACT

Voltage-gated sodium (NaV) channels are a family of transmembrane ion channel proteins. They function by forming a gated, water-filled pore to help establish and control cell membrane potential via control of the flow of ions between the intracellular and the extracellular environments. Blockade of NaVs has been successfully accomplished in the clinic to enable control of pathological firing patterns that occur in a diverse range of conditions such as chronic pain, epilepsy, and cardiac arrhythmias. First generation sodium channel modulator drugs, despite low inherent subtype selectivity, preferentially act on over-excited cells which reduces undesirable side effects in the clinic. However, the limited therapeutic indices observed with the first generation demanded a new generation of sodium channel inhibitors. The structure, function and the state of the art in sodium channel modulator drug discovery are discussed in this chapter.


Subject(s)
Analgesics/pharmacology , Anti-Arrhythmia Agents/pharmacology , Anticonvulsants/pharmacology , Drug Discovery/methods , Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/metabolism , Amino Acid Sequence , Analgesics/chemistry , Animals , Anti-Arrhythmia Agents/chemistry , Anticonvulsants/chemistry , Humans , Molecular Sequence Data , Sodium Channel Blockers/chemistry
9.
Bioorg Med Chem Lett ; 24(16): 3690-9, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25060923

ABSTRACT

Voltage-gated sodium channels (Navs) are an important family of transmembrane ion channel proteins and Nav drug discovery is an exciting field. Pharmaceutical investment in Navs for pain therapeutics has expanded exponentially due to genetic data such as SCN10A mutations and an improved ability to establish an effective screen sequence for example IonWorks Barracuda®, Synchropatch® and Qube®. Moreover, emerging clinical data (AZD-3161, XEN402, CNV1014802, PF-05089771, PF-04531083) combined with recent breakthroughs in Nav structural biology pave the way for a future of fruitful prospective Nav drug discovery.


Subject(s)
Pain/drug therapy , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Voltage-Gated Sodium Channels/metabolism , Animals , Drug Discovery , Humans , Sodium Channel Blockers/chemistry , Voltage-Gated Sodium Channels/chemistry
10.
Proc Natl Acad Sci U S A ; 110(29): E2724-32, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818614

ABSTRACT

Voltage-gated sodium (Nav) channels play a fundamental role in the generation and propagation of electrical impulses in excitable cells. Here we describe two unique structurally related nanomolar potent small molecule Nav channel inhibitors that exhibit up to 1,000-fold selectivity for human Nav1.3/Nav1.1 (ICA-121431, IC50, 19 nM) or Nav1.7 (PF-04856264, IC50, 28 nM) vs. other TTX-sensitive or resistant (i.e., Nav1.5) sodium channels. Using both chimeras and single point mutations, we demonstrate that this unique class of sodium channel inhibitor interacts with the S1-S4 voltage sensor segment of homologous Domain 4. Amino acid residues in the "extracellular" facing regions of the S2 and S3 transmembrane segments of Nav1.3 and Nav1.7 seem to be major determinants of Nav subtype selectivity and to confer differences in species sensitivity to these inhibitors. The unique interaction region on the Domain 4 voltage sensor segment is distinct from the structural domains forming the channel pore, as well as previously characterized interaction sites for other small molecule inhibitors, including local anesthetics and TTX. However, this interaction region does include at least one amino acid residue [E1559 (Nav1.3)/D1586 (Nav1.7)] that is important for Site 3 α-scorpion and anemone polypeptide toxin modulators of Nav channel inactivation. The present study provides a potential framework for identifying subtype selective small molecule sodium channel inhibitors targeting interaction sites away from the pore region.


Subject(s)
Acetamides/pharmacology , Electrophysiological Phenomena/physiology , NAV1.3 Voltage-Gated Sodium Channel/metabolism , Thiazoles/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Amino Acid Motifs/genetics , Binding Sites/genetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , NAV1.3 Voltage-Gated Sodium Channel/genetics , Patch-Clamp Techniques , Sequence Alignment
11.
Bioorg Med Chem ; 18(22): 7816-25, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20965738

ABSTRACT

Na(v)1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons. It has been implicated in the pathophysiology of inflammatory and neuropathic pain, and we envisioned that selective blockade of Na(v)1.8 would be analgesic, while reducing adverse events typically associated with non-selective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 6-aryl-2-pyrazinecarboxamides, which are potent blockers of the human Na(v)1.8 channel and also block TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons. Selected derivatives display selectivity versus human Na(v)1.2. We further demonstrate that an example from this series is orally bioavailable and produces antinociceptive activity in vivo in a rodent model of neuropathic pain following oral administration.


Subject(s)
Neuralgia/drug therapy , Pyrazines/chemistry , Sodium Channel Blockers/chemistry , Sodium Channels/chemistry , Administration, Oral , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Ganglia, Spinal/cytology , Humans , Microsomes/metabolism , NAV1.8 Voltage-Gated Sodium Channel , Neurons/metabolism , Pyrazines/pharmacokinetics , Pyrazines/therapeutic use , Rats , Sodium Channel Blockers/pharmacokinetics , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 20(22): 6812-5, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20855211

ABSTRACT

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Subject(s)
Niacinamide/pharmacology , Sodium Channel Blockers/pharmacology , Administration, Oral , Animals , Biological Availability , Niacinamide/chemistry , Niacinamide/pharmacokinetics , Rats , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacokinetics , Structure-Activity Relationship
13.
Neuropharmacology ; 59(3): 201-7, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20566409

ABSTRACT

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Na(v)1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Na(v)1.8 channel blockers. A-887826 exemplifies this series and potently (IC(50)=11nM) blocked recombinant human Na(v)1.8 channels. A-887826 was approximately 3 fold less potent to block Na(v)1.2, approximately 10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na(+)) currents and was >30 fold less potent to block Na(V)1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na(+)) currents (IC(50)=8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freund's adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by approximately 4mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Na(v)1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Na(v)1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Na(v)1.8 sodium channels in pathological pain states.


Subject(s)
Hyperalgesia/drug therapy , Pain Threshold/drug effects , Sodium Channel Blockers/pharmacology , Sodium Channel Blockers/therapeutic use , Sodium Channels/metabolism , Animals , Biophysics , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation/methods , Ganglia, Spinal/cytology , Humans , Hyperalgesia/etiology , Male , Membrane Potentials/drug effects , Morpholines/chemistry , Morpholines/pharmacology , Morpholines/therapeutic use , NAV1.8 Voltage-Gated Sodium Channel , Neuralgia/complications , Neuralgia/etiology , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Niacinamide/pharmacology , Niacinamide/therapeutic use , Patch-Clamp Techniques/methods , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/chemistry , Sodium Channels/drug effects , Spinal Cord Injuries/complications , Tetrodotoxin/pharmacology , Transfection/methods
14.
Bioorg Med Chem ; 16(12): 6379-86, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18501613

ABSTRACT

The synthesis and pharmacological characterization of a novel furan-based class of voltage-gated sodium channel blockers is reported. Compounds were evaluated for their ability to block the tetrodotoxin-resistant sodium channel Na(v)1.8 (PN3) as well as the Na(v)1.2 and Na(v)1.5 subtypes. Benchmark compounds from this series possessed enhanced potency, oral bioavailability, and robust efficacy in a rodent model of neuropathic pain, together with improved CNS and cardiovascular safety profiles compared to the clinically used sodium channel blockers mexiletine and lamotrigine.


Subject(s)
Analgesics, Non-Narcotic/chemistry , Analgesics, Non-Narcotic/pharmacology , Furans/chemistry , Furans/pharmacology , Neuralgia/drug therapy , Piperazines/chemistry , Piperazines/pharmacology , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Analgesics, Non-Narcotic/chemical synthesis , Animals , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Furans/chemical synthesis , Humans , Male , Mice , Piperazines/chemical synthesis , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/chemical synthesis , Structure-Activity Relationship
15.
J Med Chem ; 51(3): 407-16, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18176998

ABSTRACT

Nav1.8 (also known as PN3) is a tetrodotoxin-resistant (TTx-r) voltage-gated sodium channel (VGSC) that is highly expressed on small diameter sensory neurons and has been implicated in the pathophysiology of inflammatory and neuropathic pain. Recent studies using an Nav1.8 antisense oligonucleotide in an animal model of chronic pain indicated that selective blockade of Nav1.8 was analgesic and could provide effective analgesia with a reduction in the adverse events associated with nonselective VGSC blocking therapeutic agents. Herein, we describe the preparation and characterization of a series of 5-substituted 2-furfuramides, which are potent, voltage-dependent blockers (IC50 < 10 nM) of the human Nav1.8 channel. Selected derivatives, such as 7 and 27, also blocked TTx-r sodium currents in rat dorsal root ganglia (DRG) neurons with comparable potency and displayed >100-fold selectivity versus human sodium (Nav1.2, Nav1.5, Nav1.7) and human ether-a-go-go (hERG) channels. Following systemic administration, compounds 7 and 27 dose-dependently reduced neuropathic and inflammatory pain in experimental rodent models.


Subject(s)
Amides/chemical synthesis , Analgesics/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Furans/chemical synthesis , Sodium Channel Blockers/chemical synthesis , Sodium Channels/physiology , Amides/chemistry , Amides/pharmacology , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Line , Cricetinae , Cricetulus , Furans/chemistry , Furans/pharmacokinetics , Furans/pharmacology , Ganglia, Spinal/cytology , Humans , In Vitro Techniques , Male , Mice , NAV1.8 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/physiology , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Pain/etiology , Patch-Clamp Techniques , Peripheral Nervous System Diseases/drug therapy , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Sodium Channel Blockers/pharmacokinetics , Sodium Channel Blockers/pharmacology , Structure-Activity Relationship
16.
Proc Natl Acad Sci U S A ; 104(20): 8520-5, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17483457

ABSTRACT

Activation of tetrodotoxin-resistant sodium channels contributes to action potential electrogenesis in neurons. Antisense oligonucleotide studies directed against Na(v)1.8 have shown that this channel contributes to experimental inflammatory and neuropathic pain. We report here the discovery of A-803467, a sodium channel blocker that potently blocks tetrodotoxin-resistant currents (IC(50) = 140 nM) and the generation of spontaneous and electrically evoked action potentials in vitro in rat dorsal root ganglion neurons. In recombinant cell lines, A-803467 potently blocked human Na(v)1.8 (IC(50) = 8 nM) and was >100-fold selective vs. human Na(v)1.2, Na(v)1.3, Na(v)1.5, and Na(v)1.7 (IC(50) values >or=1 microM). A-803467 (20 mg/kg, i.v.) blocked mechanically evoked firing of wide dynamic range neurons in the rat spinal dorsal horn. A-803467 also dose-dependently reduced mechanical allodynia in a variety of rat pain models including: spinal nerve ligation (ED(50) = 47 mg/kg, i.p.), sciatic nerve injury (ED(50) = 85 mg/kg, i.p.), capsaicin-induced secondary mechanical allodynia (ED(50) approximately 100 mg/kg, i.p.), and thermal hyperalgesia after intraplantar complete Freund's adjuvant injection (ED(50) = 41 mg/kg, i.p.). A-803467 was inactive against formalin-induced nociception and acute thermal and postoperative pain. These data demonstrate that acute and selective pharmacological blockade of Na(v)1.8 sodium channels in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


Subject(s)
Aniline Compounds/pharmacology , Aniline Compounds/pharmacokinetics , Furans/pharmacology , Furans/pharmacokinetics , Mononeuropathies/therapy , Nerve Tissue Proteins/antagonists & inhibitors , Pain Management , Pain/pathology , Sodium Channel Blockers/pharmacology , Sodium Channels/metabolism , Action Potentials/drug effects , Analgesics/pharmacology , Aniline Compounds/administration & dosage , Aniline Compounds/chemistry , Animals , Capsaicin/pharmacology , Evoked Potentials/drug effects , Furans/administration & dosage , Furans/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Humans , Inflammation , Kinetics , Male , NAV1.8 Voltage-Gated Sodium Channel , Neurons/cytology , Neurons/drug effects , Pain/chemically induced , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacokinetics
17.
Channels (Austin) ; 1(3): 152-3, 2007.
Article in English | MEDLINE | ID: mdl-18690030

ABSTRACT

Sodium channels are key proteins in regulating neuronal excitability and accumulating data suggest that specific subtypes of voltage-dependent sodium channels are important in signaling various types of pain. Consistent with this theme, Jarvis et al.(7) recently reported the identification of a subtype-selective Na(v)1.8 blocker that was active in several pre-clinical models of pain. During the course of these studies compounds were also identified that showed large differences in potency when tested on Na(v)1.8 channels from different species. This addendum illustrates one of these compounds along with the potency correlation between recombinant and native tetrodotoxin-resistant sodium channels for additional examples. These data show that significant differences can be observed for sodium channel blockers across species and highlight the importance of considering this possibility when searching for new compounds and research tools to probe sodium channel function.


Subject(s)
Analgesics/pharmacology , Aniline Compounds/pharmacology , Furans/pharmacology , Pain/drug therapy , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , NAV1.8 Voltage-Gated Sodium Channel , Pain/metabolism , Recombinant Proteins/antagonists & inhibitors , Sodium Channels/metabolism , Species Specificity
18.
Curr Opin Chem Biol ; 7(3): 395-401, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12826128

ABSTRACT

Screening in a 'well-less' or lawn format provides a means to screen large compound collections against many targets in a fast, versatile and cost effective manner. The development of generic lawn format assays to screen various gene families against large compound collections should facilitate the identification of hits and tools to use in drug discovery and chemogenomic endeavours. Lawn format holds particular promise for screening GPCRs and selected enzyme families with potential use in other gene families.


Subject(s)
Combinatorial Chemistry Techniques/methods , Drug Evaluation, Preclinical/methods , Animals , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cells, Cultured , Drug Design , Enzymes/chemistry , Humans , Protein Binding
19.
J Comb Chem ; 5(2): 110-7, 2003.
Article in English | MEDLINE | ID: mdl-12625700

ABSTRACT

The high-throughput manual solid-phase parallel synthesis of libraries comprising thousands of discrete samples using pellicular supports (i.e. SynPhase crowns and lanterns) and a suite of novel tools and techniques is described. Key aspects of this approach include the combination of a split-split-split synthesis strategy with spatial encoding to differentiate thousands of crowns, the rapid washing and filtration of up to 48 reaction vessels in parallel, the application of an inexpensive and environmentally friendly technique to remove trifluoroacetic acid from sixteen 96-well plates in parallel, and a high-throughput method for removing cleaved crowns from reusable pin racks. Tens of thousands of discrete samples have been produced in-house using this conceptually and operationally straightforward strategy.

SELECTION OF CITATIONS
SEARCH DETAIL
...