Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 379(1): 85-95, 2021 10.
Article in English | MEDLINE | ID: mdl-34253646

ABSTRACT

Current therapies for gastroparesis metoclopramide and domperidone carry risks of extrapyramidal symptoms and life-threatening cardiac arrhythmias. Trazpiroben, a novel, potent dopamine D2/D3 receptor antagonist, has low brain permeation and very low affinity for human ether-à-go-go-related gene (hERG) channel inhibition, potentially improving on safety profiles of existing therapies. Trazpiroben demonstrated the following receptor affinities: high for D2 and D3, moderate for D4, and minimal for D1 and D5 It demonstrated moderate affinity for adrenergic α 1B (α 1B) and 5-hydroxytryptamine (5HT) 2A receptors and low potential for off-target adverse events (AEs). Trazpiroben potently inhibited dopamine-activated D2L receptor activation of cognate G-proteins in human embryonic kidney 293 cell membranes and was a neutral D2L receptor antagonist. In vivo, trazpiroben dose-dependently increased prolactin release in orally dosed rat (0.1-1 mg/kg). Additionally, multiple oral doses in the rat (100 mg/kg) and dog (50 mg/kg) for 3 days produced robust plasma exposures and prolactin increases in both species. Trazpiroben inhibited retching/vomiting in the dog with apomorphine-induced emesis with a potency (0.1-1 mg/kg) like that of trazpiroben-mediated prolactin increases in rat. Oral trazpiroben (1, 10, and 30 mg/kg) did not affect rat rotarod performance, suggesting low brain penetration. Trazpiroben concentrations were low in cerebrospinal fluid versus plasma after multiple oral doses for 4 days in rat and dog. Trazpiroben weakly inhibited the hERG channel current (concentration causing half-maximal inhibition of control-specific binding of 15.6 µM), indicating little potential for disrupting cardiac rhythm. Overall, trazpiroben is a potent D2/D3 receptor antagonist designed to avoid the serious potential AEs associated with current gastroparesis therapies. SIGNIFICANCE STATEMENT: Trazpiroben is a novel, potent dopamine D2/D3 selective receptor antagonist designed to avoid adverse effects associated with the current pharmacological therapies metoclopramide and domperidone. Preclinical studies have demonstrated low brain penetration and weak affinity for the hERG channel, indicating that trazpiroben is not expected to be associated with central nervous system or cardiovascular safety issues. With these pharmacological properties, trazpiroben may represent a viable new treatment option for gastroparesis because of a potentially improved safety profile relative to existing therapies.


Subject(s)
Dopamine D2 Receptor Antagonists/therapeutic use , Gastroparesis/drug therapy , Receptors, Dopamine D3/antagonists & inhibitors , Triazoles/therapeutic use , Animals , Antiemetics/pharmacology , Antiemetics/therapeutic use , CHO Cells , Cricetinae , Cricetulus , Dogs , Domperidone/analogs & derivatives , Domperidone/pharmacology , Domperidone/therapeutic use , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacology , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists/chemistry , Dopamine D2 Receptor Antagonists/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Humans , Male , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Triazoles/pharmacology
2.
J Physiol ; 597(17): 4615-4625, 2019 09.
Article in English | MEDLINE | ID: mdl-31246276

ABSTRACT

KEY POINTS: Skeletal muscle fatigue limits performance in various physical activities, with exercise intolerance being a key symptom in a broad spectrum of diseases. We investigated whether a small molecule fast skeletal troponin activator (FSTA), CK-2066260, can mitigate muscle fatigue by reducing the cytosolic free [Ca2+ ] required to produce a given submaximal force and hence decreasing the energy requirement. Isolated intact single mouse muscle fibres and rat muscles in-situ treated with CK-2066260 showed improved muscle endurance., which was accompanied by decreased ATP demand and reduced glycogen usage. CK-2066260 treatment improved in-vivo exercise capacity in healthy rats and in a rat model of peripheral artery insufficiency. In conclusion, we show that the FSTA CK-2066260 effectively counteracts muscle fatigue in rodent skeletal muscle in vitro, in situ, and in vivo. This may translate to humans and provide a promising pharmacological treatment to patients suffering from severe muscle weakness and exercise intolerance. ABSTRACT: Skeletal muscle fatigue limits performance during physical exercise and exacerbated muscle fatigue is a prominent symptom among a broad spectrum of diseases. The present study investigated whether skeletal muscle fatigue is affected by the fast skeletal muscle troponin activator (FSTA) CK-2066260, which increases myofibrillar Ca2+ sensitivity and amplifies the submaximal force response. Because more force is produced for a given Ca2+ , we hypothesized that CK-2066260 could mitigate muscle fatigue by reducing the energetic cost of muscle activation. Isolated single mouse muscle fibres were fatigued by 100 repeated 350 ms contractions while measuring force and the cytosolic free [Ca2+ ] or [Mg2+ ] ([Mg2+ ]i ). When starting fatiguing stimulation at matching forces (i.e. lower stimulation frequency with CK-2066260): force was decreased by ∼50% with and by ∼75% without CK-2066260; [Mg2+ ]i was increased by ∼10% with and ∼32% without CK-2066260, reflecting a larger decrease in [ATP] in the latter. The glycogen content in in situ stimulated rat muscles fatigued by repeated contractions at matching forces was about two times higher with than without CK-2066260. Voluntary exercise capacity, assessed by rats performing rotarod exercise and treadmill running, was improved in the presence of CK-2066260. CK-2066260 treatment also increased skeletal muscle fatigue resistance and exercise performance in a rat model of peripheral artery insufficiency. In conclusion, we demonstrate that the FSTA CK-2066260 mitigates skeletal muscle fatigue by reducing the metabolic cost of force generation.


Subject(s)
Muscle Contraction/physiology , Muscle Fatigue/physiology , Muscle Fibers, Fast-Twitch/metabolism , Troponin/metabolism , Animals , Calcium/metabolism , Female , Glycogen/metabolism , Male , Mice , Mice, Inbred C57BL , Myofibrils/metabolism , Physical Conditioning, Animal/physiology , Rats , Rats, Sprague-Dawley
3.
Pharmacol Res Perspect ; 6(3): e00400, 2018 06.
Article in English | MEDLINE | ID: mdl-29736245

ABSTRACT

Revefenacin (TD-4208) is a novel, long-acting, and lung-selective muscarinic cholinergic receptor (mAChR) antagonist in development as a nebulized inhalation solution for the treatment of chronic obstructive pulmonary disease (COPD) patients. This study evaluated the pharmacology of revefenacin at human recombinant mAChRs and in airway tissues from rats, guinea pigs, and humans. At human recombinant mAChRs, revefenacin displayed high affinity (pKI = 8.2-9.8) and behaved as a competitive antagonist (pKI, apparent = 9.4-10.9) at the five human recombinant mAChRs. Kinetic studies demonstrated that revefenacin dissociated significantly slower from the hM3 (t1/2 = 82 minutes) compared to the hM 2 (t1/2 = 6.9 minutes) mAChR at 37°C, thereby making it kinetically selective for the former subtype. Similarly, in functional studies, revefenacin-mediated antagonism of acetylcholine (ACh)-evoked calcium mobilization responses were reversed less rapidly at hM3 compared to the hM2 mAChR. In isolated tracheal tissues from rat and guinea pig and isolated bronchial tissues from humans, revefenacin potently antagonized mAChR-mediated contractile responses. Furthermore, the antagonistic effects of revefenacin in rat, guinea pig, and human airway tissues were slowly reversible (t1/2 of 13.3, >16, and >10 hours, respectively). These data demonstrate that revefenacin is a potent, high affinity, and selective functional mAChR antagonist with kinetic selectivity for the hM3 receptor and produces potent and long-lasting antagonism of mAChR-mediated contractile responses in rat, guinea pig, and human airway tissue. These data suggest that revefenacin has the potential to be a potent once-daily dosed inhaled bronchodilator in COPD patients.


Subject(s)
Benzamides/pharmacology , Bronchi/physiology , Carbamates/pharmacology , Muscarinic Antagonists/pharmacology , Recombinant Proteins/metabolism , Trachea/physiology , Administration, Inhalation , Animals , Bronchi/drug effects , Guinea Pigs , Humans , Nebulizers and Vaporizers , Rats , Trachea/drug effects
4.
J Physiol ; 595(5): 1657-1670, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27869319

ABSTRACT

KEY POINTS: We report that the small molecule CK-2066260 selectively slows the off-rate of Ca2+ from fast skeletal muscle troponin, leading to increased myofibrillar Ca2+ sensitivity in fast skeletal muscle. Rodents dosed with CK-2066260 show increased hindlimb muscle force and power in response to submaximal rates of nerve stimulation in situ. CK-2066260 has no effect on free cytosolic [Ca2+ ] during contractions of isolated muscle fibres. We conclude that fast skeletal muscle troponin sensitizers constitute a potential therapy to address an unmet need of improving muscle function in conditions of weakness and premature muscle fatigue. ABSTRACT: Skeletal muscle dysfunction occurs in many diseases and can lead to muscle weakness and premature muscle fatigue. Here we show that the fast skeletal troponin activator, CK-2066260, counteracts muscle weakness by increasing troponin Ca2+ affinity, thereby increasing myofibrillar Ca2+ sensitivity. Exposure to CK-2066260 resulted in a concentration-dependent increase in the Ca2+ sensitivity of ATPase activity in isolated myofibrils and reconstituted hybrid sarcomeres containing fast skeletal muscle troponin C. Stopped-flow experiments revealed a ∼2.7-fold decrease in the Ca2+ off-rate of isolated troponin complexes in the presence of CK-2066260 (6 vs. 17 s-1 under control conditions). Isolated mouse flexor digitorum brevis fibres showed a rapidly developing, reversible and concentration-dependent force increase at submaximal stimulation frequencies. This force increase was not accompanied by any changes in the free cytosolic [Ca2+ ] or its kinetics. CK-2066260 induced a slowing of relaxation, which was markedly larger at 26°C than at 31°C and could be linked to the decreased Ca2+ off-rate of troponin C. Rats dosed with CK-2066260 showed increased hindlimb isometric and isokinetic force in response to submaximal rates of nerve stimulation in situ producing significantly higher absolute forces at low isokinetic velocities, whereas there was no difference in force at the highest velocities. Overall muscle power was increased and the findings are consistent with a lack of effect on crossbridge kinetics. In conclusion, CK-2066260 acts as a fast skeletal troponin activator that may be used to increase muscle force and power in conditions of muscle weakness.


Subject(s)
Calcium/physiology , Imidazoles/pharmacology , Muscle Fibers, Fast-Twitch/drug effects , Myofibrils/drug effects , Pyrazines/pharmacology , Adenosine Triphosphatases/physiology , Animals , Cattle , Female , Hindlimb/drug effects , Hindlimb/physiology , Mice, Inbred C57BL , Muscle Fibers, Fast-Twitch/physiology , Myofibrils/physiology , Rabbits , Rats, Sprague-Dawley , Troponin C/physiology
6.
J Pharmacol Exp Ther ; 353(1): 159-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25678535

ABSTRACT

Heart failure-mediated skeletal myopathy, which is characterized by muscle atrophy and muscle metabolism dysfunction, often manifests as dyspnea and limb muscle fatigue. We have previously demonstrated that increasing Ca(2+) sensitivity of the sarcomere by a small-molecule fast skeletal troponin activator improves skeletal muscle force and exercise performance in healthy rats and models of neuromuscular disease. The objective of this study was to investigate the effect of a novel fast skeletal troponin activator, CK-2127107 (2-aminoalkyl-5-N-heteroarylpyrimidine), on skeletal muscle function and exercise performance in rats exhibiting heart failure-mediated skeletal myopathy. Rats underwent a left anterior descending coronary artery ligation, resulting in myocardial infarction and a progressive decline in cardiac function [left anterior descending coronary artery heart failure (LAD-HF)]. Compared with sham-operated control rats, LAD-HF rat hindlimb and diaphragm muscles exhibited significant muscle atrophy. Fatigability was increased during repeated in situ isokinetic plantar flexor muscle contractions. CK-2127107 produced a leftward shift in the force-Ca(2+) relationship of skinned, single diaphragm, and extensor digitorum longus fibers. Exercise performance, which was assessed by rotarod running, was lower in vehicle-treated LAD-HF rats than in sham controls (116 ± 22 versus 193 ± 31 seconds, respectively; mean ± S.E.M.; P = 0.04). In the LAD-HF rats, a single oral dose of CK-2127107 (10 mg/kg p.o.) increased running time compared with vehicle treatment (283 ± 47 versus 116 ± 22 seconds; P = 0.0004). In summary, CK-2127107 substantially increases exercise performance in this heart failure model, suggesting that modulation of skeletal muscle function by a fast skeletal troponin activator may be a useful therapeutic in heart failure-associated exercise intolerance.


Subject(s)
Heart Failure, Systolic/physiopathology , Muscle, Skeletal/drug effects , Physical Conditioning, Animal , Pyrimidines/pharmacology , Troponin/metabolism , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Cattle , Female , Heart Failure, Systolic/complications , Heart Failure, Systolic/metabolism , Muscle Contraction/drug effects , Muscle Fibers, Fast-Twitch/drug effects , Muscle Fibers, Fast-Twitch/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myofibrils/drug effects , Myofibrils/metabolism , Rabbits , Rats, Sprague-Dawley , Rotarod Performance Test
7.
J Med Chem ; 58(6): 2609-22, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25629394

ABSTRACT

Through application of our multivalent approach to drug discovery we previously reported the first discovery of dual pharmacology MABA bronchodilators, exemplified by 1. Herein we describe the subsequent lead optimization of both muscarinic antagonist and ß2 agonist activities, through modification of the linker motif, to achieve 24 h duration of action in a guinea pig bronchoprotection model. Concomitantly we targeted high lung selectivities, low systemic exposures and identified crystalline forms suitable for inhalation devices. This article culminates with the discovery of our first clinical candidate 12f (TD-5959, GSK961081, batefenterol). In a phase 2b trial, batefenterol produced statistical and clinically significant differences compared to placebo and numerically greater improvements in the primary end point of trough FEV1 compared to salmeterol after 4 weeks of dosing in patients with moderate to severe chronic obstructive pulmonary disease (COPD).


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Carbamates/therapeutic use , Drug Discovery , Lung/drug effects , Muscarinic Antagonists/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinolones/therapeutic use , Administration, Inhalation , Adrenergic beta-2 Receptor Agonists/administration & dosage , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Agonists/pharmacokinetics , Animals , CHO Cells , Carbamates/administration & dosage , Carbamates/chemistry , Carbamates/pharmacokinetics , Cricetulus , Guinea Pigs , HEK293 Cells , Humans , Models, Molecular , Muscarinic Antagonists/administration & dosage , Muscarinic Antagonists/chemistry , Muscarinic Antagonists/pharmacokinetics , Quinolones/administration & dosage , Quinolones/chemistry , Quinolones/pharmacokinetics
8.
J Pharmacol Exp Ther ; 351(1): 190-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25100753

ABSTRACT

The objective of the present studies was to characterize the pharmacologic properties of GSK-961081 [TD-5959; (R)-1-(3-((2-chloro-4-(((2-hydroxy-2-(8-hydroxy-2-oxo-1,2-dihydroquinolin-5-yl)ethyl)amino)methyl)-5-methoxyphenyl)amino)-3-oxopropyl) piperidin-4-yl [1,1'-biphenyl]-2-ylcarbamate], a novel first-in-class inhaled bifunctional compound possessing both muscarinic antagonist (MA) and ß2-adrenoceptor agonist (BA) properties (MABA). In competition radioligand binding studies at human recombinant receptors, GSK-961081 displayed high affinity for hM2 (Ki = 1.4 nM), hM3 muscarinic receptors (Ki = 1.3 nM) and hß2-adrenoceptors (Ki = 3.7 nM). GSK-961081 behaved as a potent hß2-adrenoceptor agonist (EC50 = 0.29 nM for stimulation of cAMP levels) with 440- and 320-fold functional selectivity over hß1- and hß3-adrenoceptors, respectively. In guinea pig isolated tracheal tissues, GSK-961081 produced smooth muscle relaxation through MA (EC50 = 50.2 nM), BA (EC50=24.6 nM), and MABA (EC50 = 11 nM) mechanisms. In the guinea pig bronchoprotection assay, inhaled GSK-961081 produced potent, dose-dependent inhibition of bronchoconstrictor responses via MA (ED50 = 33.9 µg/ml), BA (ED50 = 14.1 µg/ml), and MABA (ED50 = 6.4 µg/ml) mechanisms. Significant bronchoprotective effects of GSK-961081 were evident in guinea pigs via MA, BA, and MABA mechanisms for up to 7 days after dosing. The lung selectivity index of GSK-961081 in guinea pigs was 55- to 110-fold greater than that of tiotropium with respect to systemic antimuscarinic antisialagogue effects and was 10-fold greater than that of salmeterol with respect to systemic ß2-adrenoceptor hypotensive effects. These preclinical findings studies suggest that GSK-961081 has the potential to be a promising next-generation inhaled lung-selective bronchodilator for the treatment of airway diseases, including chronic obstructive pulmonary disease.


Subject(s)
Adrenergic beta-2 Receptor Antagonists/pharmacology , Bronchodilator Agents/pharmacology , Carbamates/pharmacology , Muscarinic Antagonists/pharmacology , Quinolones/pharmacology , Adrenergic beta-2 Receptor Antagonists/adverse effects , Adrenergic beta-2 Receptor Antagonists/pharmacokinetics , Albuterol/analogs & derivatives , Albuterol/pharmacokinetics , Albuterol/pharmacology , Animals , Bronchodilator Agents/adverse effects , Bronchodilator Agents/pharmacokinetics , CHO Cells , Carbamates/adverse effects , Carbamates/pharmacokinetics , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Drug Evaluation, Preclinical , Guinea Pigs , HEK293 Cells , Humans , Lung/drug effects , Lung/physiology , Muscarinic Antagonists/adverse effects , Muscarinic Antagonists/pharmacokinetics , Muscle Relaxation , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Protein Binding , Quinolones/adverse effects , Quinolones/pharmacokinetics , Receptors, Adrenergic, beta/metabolism , Receptors, Muscarinic/metabolism , Salmeterol Xinafoate , Scopolamine Derivatives/pharmacokinetics , Scopolamine Derivatives/pharmacology , Tiotropium Bromide , Tissue Distribution , Trachea/drug effects , Trachea/physiology
9.
PLoS One ; 9(5): e96921, 2014.
Article in English | MEDLINE | ID: mdl-24805850

ABSTRACT

Amyotrophic Lateral Sclerosis (ALS) is a motor neuron disease characterized by progressive motor neuron loss resulting in muscle atrophy, declining muscle function, and eventual paralysis. Patients typically die from respiratory failure 3 to 5 years from the onset of symptoms. Tirasemtiv is a fast skeletal troponin activator that sensitizes the sarcomere to calcium; this mechanism of action amplifies the response of muscle to neuromuscular input producing greater force when nerve input is reduced. Here, we demonstrate that a single dose of tirasemtiv significantly increases submaximal isometric force, forelimb grip strength, grid hang time, and rotarod performance in a female transgenic mouse model (B6SJL-SOD1 G93A) of ALS with functional deficits. Additionally, diaphragm force and tidal volume are significantly higher in tirasemtiv-treated female B6SJL-SOD1 G93A mice. These results support the potential of fast skeletal troponin activators to improve muscle function in neuromuscular diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Imidazoles/administration & dosage , Motor Neurons/drug effects , Muscle Strength/drug effects , Pyrazines/administration & dosage , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Motor Neurons/pathology , Muscle Strength/genetics , Muscle, Skeletal/drug effects , Troponin/genetics , Troponin/metabolism
10.
Brain ; 136(Pt 6): 1718-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23715096

ABSTRACT

Nebulin--a giant sarcomeric protein--plays a pivotal role in skeletal muscle contractility by specifying thin filament length and function. Although mutations in the gene encoding nebulin (NEB) are a frequent cause of nemaline myopathy, the most common non-dystrophic congenital myopathy, the mechanisms by which mutations in NEB cause muscle weakness remain largely unknown. To better understand these mechanisms, we have generated a mouse model in which Neb exon 55 is deleted (Neb(ΔExon55)) to replicate a founder mutation seen frequently in patients with nemaline myopathy with Ashkenazi Jewish heritage. Neb(ΔExon55) mice are born close to Mendelian ratios, but show growth retardation after birth. Electron microscopy studies show nemaline bodies--a hallmark feature of nemaline myopathy--in muscle fibres from Neb(ΔExon55) mice. Western blotting studies with nebulin-specific antibodies reveal reduced nebulin levels in muscle from Neb(ΔExon55) mice, and immunofluorescence confocal microscopy studies with tropomodulin antibodies and phalloidin reveal that thin filament length is significantly reduced. In line with reduced thin filament length, the maximal force generating capacity of permeabilized muscle fibres and single myofibrils is reduced in Neb(ΔExon55) mice with a more pronounced reduction at longer sarcomere lengths. Finally, in Neb(ΔExon55) mice the regulation of contraction is impaired, as evidenced by marked changes in crossbridge cycling kinetics and by a reduction of the calcium sensitivity of force generation. A novel drug that facilitates calcium binding to the thin filament significantly augmented the calcium sensitivity of submaximal force to levels that exceed those observed in untreated control muscle. In conclusion, we have characterized the first nebulin-based nemaline myopathy model, which recapitulates important features of the phenotype observed in patients harbouring this particular mutation, and which has severe muscle weakness caused by thin filament dysfunction.


Subject(s)
Disease Models, Animal , Exons/genetics , Muscle Proteins/genetics , Myopathies, Nemaline/genetics , Severity of Illness Index , Animals , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Weakness/genetics , Muscle Weakness/pathology , Myopathies, Nemaline/pathology
11.
J Med Genet ; 50(6): 383-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23572184

ABSTRACT

BACKGROUND: Nemaline myopathy-the most common non-dystrophic congenital myopathy-is caused by mutations in thin filament genes, of which the nebulin gene is the most frequently affected one. The nebulin gene codes for the giant sarcomeric protein nebulin, which plays a crucial role in skeletal muscle contractile performance. Muscle weakness is a hallmark feature of nemaline myopathy patients with nebulin mutations, and is caused by changes in contractile protein function, including a lower calcium-sensitivity of force generation. To date no therapy exists to treat muscle weakness in nemaline myopathy. Here, we studied the ability of the novel fast skeletal muscle troponin activator, CK-2066260, to augment force generation at submaximal calcium levels in muscle cells from nemaline myopathy patients with nebulin mutations. METHODS: Contractile protein function was determined in permeabilised muscle cells isolated from frozen patient biopsies. The effect of 5 µM CK-2066260 on force production was assessed. RESULTS: Nebulin protein concentrations were severely reduced in muscle cells from these patients compared to controls, while myofibrillar ultrastructure was largely preserved. Both maximal active tension and the calcium-sensitivity of force generation were lower in patients compared to controls. Importantly, CK-2066260 greatly increased the calcium-sensitivity of force generation-without affecting the cooperativity of activation-in patients to levels that exceed those observed in untreated control muscle. CONCLUSIONS: Fast skeletal troponin activation is a therapeutic mechanism to augment contractile protein function in nemaline myopathy patients with nebulin mutations and with other neuromuscular diseases.


Subject(s)
Imidazoles/pharmacology , Muscle Proteins/genetics , Muscle Strength/drug effects , Mutation/genetics , Myopathies, Nemaline/physiopathology , Pyrazines/pharmacology , Troponin/metabolism , Adult , Biopsy , Calcium/metabolism , Child, Preschool , Humans , Imidazoles/administration & dosage , Infant , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Myopathies, Nemaline/drug therapy , Myopathies, Nemaline/genetics , Pyrazines/administration & dosage , Treatment Outcome , Troponin/drug effects , Young Adult
12.
Muscle Nerve ; 48(2): 279-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23512724

ABSTRACT

INTRODUCTION: Tirasemtiv is a novel small molecule activator of the fast skeletal muscle troponin complex that produces sensitization of the sarcomere to calcium. Tirasemtiv is currently in Phase II clinical trials for neuromuscular disease. METHODS: We conducted a blinded, randomized, placebo-controlled preclinical study of the effect of tirasemtiv on forearm grip strength, endurance, respiratory physiology, and muscle pathology in adequate sample sizes of the Lama2(dy-2J) mouse model of congenital muscular dystrophy. RESULTS: Mice receiving a high dose of tirasemtiv had significantly higher muscle fiber cross-sectional area and respiratory response to CO2 stimulation at 16 weeks than mice on low dose or placebo. There were no changes in muscle pathology, serum creatine kinase, strength, endurance, or respiration following long-term treatment. CONCLUSIONS: We conclude that tirasemtiv influences the structure of the skeletal muscle fiber in this model of muscular dystrophy but does not impact muscle function, as evaluated in this study.


Subject(s)
Imidazoles/therapeutic use , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/genetics , Pyrazines/therapeutic use , Troponin/metabolism , Analysis of Variance , Animals , Creatine Kinase/blood , Disease Models, Animal , Female , Imidazoles/pharmacology , Laminin/genetics , Longitudinal Studies , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Fibers, Skeletal/drug effects , Muscle Strength/drug effects , Muscle Strength/physiology , Muscular Dystrophies/blood , Muscular Dystrophies/drug therapy , Muscular Dystrophies/pathology , Mutation/genetics , Physical Endurance/drug effects , Physical Endurance/genetics , Pyrazines/pharmacology , Respiratory Physiological Phenomena/drug effects , Respiratory Physiological Phenomena/genetics
13.
PLoS One ; 8(2): e55861, 2013.
Article in English | MEDLINE | ID: mdl-23437068

ABSTRACT

The effect of the fast skeletal muscle troponin activator, CK-2066260, on calcium-induced force development was studied in skinned fast skeletal muscle fibers from wildtype (WT) and nebulin deficient (NEB KO) mice. Nebulin is a sarcomeric protein that when absent (NEB KO mouse) or present at low levels (nemaline myopathy (NM) patients with NEB mutations) causes muscle weakness. We studied the effect of fast skeletal troponin activation on WT muscle and tested whether it might be a therapeutic mechanism to increase muscle strength in nebulin deficient muscle. We measured tension-pCa relations with and without added CK-2066260. Maximal active tension in NEB KO tibialis cranialis fibers in the absence of CK-2066260 was ∼60% less than in WT fibers, consistent with earlier work. CK-2066260 shifted the tension-calcium relationship leftwards, with the largest relative increase (up to 8-fold) at low to intermediate calcium levels. This was a general effect that was present in both WT and NEB KO fiber bundles. At pCa levels above ∼6.0 (i.e., calcium concentrations <1 µM), CK-2066260 increased tension of NEB KO fibers to beyond that of WT fibers. Crossbridge cycling kinetics were studied by measuring k(tr) (rate constant of force redevelopment following a rapid shortening/restretch). CK-2066260 greatly increased k(tr) at submaximal activation levels in both WT and NEB KO fiber bundles. We also studied the sarcomere length (SL) dependence of the CK-2066260 effect (SL 2.1 µm and 2.6 µm) and found that in the NEB KO fibers, CK-2066260 had a larger effect on calcium sensitivity at the long SL. We conclude that fast skeletal muscle troponin activation increases force at submaximal activation in both wildtype and NEB KO fiber bundles and, importantly, that this troponin activation is a potential therapeutic mechanism for increasing force in NM and other skeletal muscle diseases with loss of muscle strength.


Subject(s)
Muscle Fatigue/physiology , Muscle Fibers, Fast-Twitch/physiology , Muscle Proteins/deficiency , Troponin/metabolism , Animals , Biomechanical Phenomena , Calcium/metabolism , In Vitro Techniques , Kinetics , Mice , Mice, Knockout , Muscle Proteins/metabolism , Sarcomeres/metabolism
15.
Eur J Pharmacol ; 605(1-3): 145-52, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19168050

ABSTRACT

Existing antimuscarinic drugs for overactive bladder have high affinity for M(3)/M(1) muscarinic receptors and consequently produce M(3)/M(1)-mediated adverse effects including dry mouth, constipation, mydriasis and somnolence. TD-6301 is a M(2/4) muscarinic receptor-selective antagonist developed for the treatment of overactive bladder. The present studies characterize the in vitro and in vivo pharmacological properties of this molecule in comparison to other marketed antimuscarinics agents. In radioligand binding studies, TD-6301 was found to possess high affinity for human M(2) muscarinic receptor (K(i)=0.36 nM) and was 31, 36, 2 and 128-fold selective for the human M(2) muscarinic receptor compared to the M(1), M(3), M(4) and M(5) muscarinic receptors, respectively. The in vivo bladder selectivity of TD-6301 in rats was determined to be 26, 28, >100, 16 and 0.4-fold, respectively, assessed by comparing its potency for inhibition of volume-induced bladder contractions to that for inhibition of oxotremorine-induced salivation, inhibition of small-intestinal transit, decreases in locomotor activity, increases in pupil diameter and increases in heart rate. TD-6301 was more potent in inhibiting volume-induced bladder contractions (ID(50)=0.075 mg/kg) compared to oxotremorine-induced salivation (ID(50)=1.0 mg/kg) resulting in a bladder/salivary gland selectivity ratio greater than that observed for tolterodine, oxybutynin, darifenacin and solifenacin. The preclinical properties of TD-6301 suggest that this molecule is likely to be efficacious in overactive bladder patients with a lower propensity to cause M(3) muscarinic receptor mediated adverse effects.


Subject(s)
Muscarinic Antagonists/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Receptor, Muscarinic M2/antagonists & inhibitors , Receptor, Muscarinic M4/antagonists & inhibitors , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Male , Muscarinic Antagonists/administration & dosage , Muscarinic Antagonists/toxicity , Muscle Contraction/drug effects , Radioligand Assay , Rats , Rats, Sprague-Dawley , Salivary Glands/drug effects , Salivary Glands/metabolism , Salivation/drug effects , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Urinary Bladder, Overactive/drug therapy
16.
Mol Pharmacol ; 72(2): 291-302, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478612

ABSTRACT

THRX-160209 is a potent antagonist at the M(2) muscarinic acetylcholine (ACh) receptor subtype that was designed using a multivalent strategy, simultaneously targeting the orthosteric site and a nearby site known to bind allosteric ligands. In this report, we describe three characteristics of THRX-160209 binding that are consistent with a multivalent interaction: 1) an apparent affinity of the multivalent ligand for the M2 receptor subtype (apparent pK(I) = 9.51 +/- 0.22) that was several orders of magnitude greater than its two monovalent components (apparent pK(I) values < 6.0), 2) specificity of THRX-160209 for the M2 receptor subtype compared with the closely related M4 (apparent pK(I) = 8.78 +/- 0.24) and M1, M3, and M5 receptors (apparent pK(I) values 10-fold) of the dissociation rate of tritium-labeled THRX-160209 from M2 receptors by competing monovalent ligands that are known to interact with either the orthosteric site (e.g., atropine) or a well characterized allosteric site (e.g., obidoxime) on the receptor. In complementary kinetic studies assessing allosteric modulation of the receptor, unlabeled THRX-160209 retarded dissociation of [3H]N-methyl scopolamine (NMS). The effects of THRX-160209 on retardation of [3H]NMS dissociation were competitively inhibited by obidoxime, suggesting that obidoxime and THRX-160209 bind to an overlapping region coincident with other typical muscarinic allosteric agents, such as 3-methyl-5-[7-[4-[(4S)-4-methyl-1,3-oxazolidin-2-yl]phenoxy]heptyl]-1,2-oxazole (W84) and gallamine. Taken together, these data are consistent with the hypothesis that THRX-160209 binds in a multivalent manner to the M2 receptor, simultaneously occupying the orthosteric site and a spatially distinct allosteric site.


Subject(s)
Benzeneacetamides/metabolism , Muscarinic Antagonists/metabolism , Piperidines/metabolism , Receptor, Muscarinic M2/metabolism , Allosteric Site , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Ligands , N-Methylscopolamine/metabolism , Obidoxime Chloride/pharmacology , Radioligand Assay , Receptor, Muscarinic M2/chemistry
17.
Curr Opin Investig Drugs ; 5(1): 40-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14983972

ABSTRACT

Antimuscarinic drugs have been the mainstay in the treatment of overactive bladder (OAB) for over two decades. An ideal antimuscarinic medicine is one that can normalize bladder function without interfering with parasympathetic regulation of other organs. Currently, extended-release formulations of tolterodine (tolterodine-ER) and oxybutynin (oxybutynin-ER and oxybutynin-TDS) serve as the cornerstone in the pharmacotherapy of OAB. Although these products represent a significant improvement over older agents, especially with respect to convenience of dosing schedule, their tolerability concerns and modest efficacy make them less than ideal therapies. Advances in our understanding of muscarinic receptor pharmacology have raised optimism in our ability to widen the therapeutic index and increase the efficacy of antimuscarinics by selectively targeting one or more of the five muscarinic subtypes. A structurally diverse group of molecules, having varying receptor-selectivity profiles (non-selective, M3 selective, M2 selective, M2 sparing and M5 sparing), are in development for OAB. Results of clinical trials with these drugs must be awaited before their therapeutic value can be accurately judged.


Subject(s)
Muscarinic Antagonists/therapeutic use , Urinary Bladder Diseases/drug therapy , Urination Disorders/drug therapy , Animals , Drug Design , Humans , Muscarinic Antagonists/chemistry , Receptors, Muscarinic/classification , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Structure-Activity Relationship , Urinary Bladder/metabolism , Urinary Bladder/physiopathology , Urinary Bladder Diseases/physiopathology , Urinary Bladder, Neurogenic/drug therapy , Urinary Bladder, Neurogenic/physiopathology , Urinary Incontinence/drug therapy , Urinary Incontinence/physiopathology , Urination Disorders/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...