Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Pharmacol Exp Ther ; 348(3): 421-31, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24399854

ABSTRACT

Bile acids (BAs) and BA receptors, including G protein-coupled bile acid receptor 1 (GPBAR1), represent novel targets for the treatment of metabolic and inflammatory disorders. However, BAs elicit myriad effects on cardiovascular function, although this has not been specifically ascribed to GPBAR1. This study was designed to test whether stimulation of GPBAR1 elicits effects on cardiovascular function that are mechanism based that can be identified in acute ex vivo and in vivo cardiovascular models, to delineate whether effects were due to pathways known to be modulated by BAs, and to establish whether a therapeutic window between in vivo cardiovascular liabilities and on-target efficacy could be defined. The results demonstrated that the infusion of three structurally diverse and selective GPBAR1 agonists produced marked reductions in vascular tone and blood pressure in dog, but not in rat, as well as reflex tachycardia and a positive inotropic response, effects that manifested in an enhanced cardiac output. Changes in cardiovascular function were unrelated to modulation of the levothyroxine/thyroxine axis and were nitric oxide independent. A direct effect on vascular tone was confirmed in dog isolated vascular rings, whereby concentration-dependent decreases in tension that were tightly correlated with reductions in vascular tone observed in vivo and were blocked by iberiotoxin. Compound concentrations in which cardiovascular effects occurred, both ex vivo and in vivo, could not be separated from those necessary for modulation of GPBAR1-mediated efficacy, resulting in project termination. These results are the first to clearly demonstrate direct and potent peripheral arterial vasodilation due to GPBAR1 stimulation in vivo through activation of large conductance Ca(2+) activated potassium channel K(Ca)1.1.


Subject(s)
Arteries/drug effects , Receptors, G-Protein-Coupled/agonists , Vasodilation/drug effects , Animals , Arteries/physiology , Atrial Natriuretic Factor/blood , CHO Cells , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Cytokines/blood , Dinitrofluorobenzene/analogs & derivatives , Dogs , Endothelin-1/blood , Humans , Imidazoles/pharmacology , In Vitro Techniques , Male , Nitric Oxide/biosynthesis , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Thyroxine/blood , Triazoles/pharmacology
2.
J Pharmacol Exp Ther ; 340(3): 492-500, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22128344

ABSTRACT

We previously reported the discovery of a novel ribosomal S6 kinase 2 (RSK2) inhibitor, (R)-5-Methyl-1-oxo-2,3,4,5-tetrahydro-1H-[1,4]diazepino[1,2-a] indole-8-carboxylic acid [1-(3-dimethylamino-propyl)-1H-benzoimidazol-2-yl]-amide (BIX 02565), with high potency (IC(50) = 1.1 nM) targeted for the treatment of heart failure. In the present study, we report that despite nanomolar potency at the target, BIX 02565 elicits off-target binding at multiple adrenergic receptor subtypes that are important in the control of vascular tone and cardiac function. To elucidate in vivo the functional consequence of receptor binding, we characterized the cardiovascular (CV) profile of the compound in an anesthetized rat CV screen and telemetry-instrumented conscious rats. Infusion of BIX 02565 (1, 3, and 10 mg/kg) in the rat CV screen resulted in a precipitous decrease in both mean arterial pressure (MAP; to -65 ± 6 mm Hg below baseline) and heart rate (-93 ± 13 beats/min). In telemetry-instrumented rats, BIX 02565 (30, 100, and 300 mg/kg p.o. QD for 4 days) elicited concentration-dependent decreases in MAP after each dose (to -39 ± 4 mm Hg on day 4 at T(max)); analysis by Demming regression demonstrated strong correlation independent of route of administration and influence of anesthesia. Because of pronounced off-target effects of BIX 02565 on cardiovascular function, a high-throughput selectivity screen at adrenergic α(1A) and α(2A) was performed for 30 additional RSK2 inhibitors in a novel chemical series; a wide range of adrenergic binding was achieved (0-92% inhibition), allowing for differentiation within the series. Eleven lead compounds with differential binding were advanced to the rat CV screen for in vivo profiling. This led to the identification of potent RSK2 inhibitors (cellular IC(50) <0.14 nM) without relevant α(1A) and α(2A) inhibition and no adverse cardiovascular effects in vivo.


Subject(s)
Azepines/pharmacology , Benzimidazoles/pharmacology , Blood Pressure/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Drug Discovery , Male , Rats , Rats, Sprague-Dawley
3.
J Cardiovasc Pharmacol ; 59(4): 369-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22179024

ABSTRACT

The strategic integration of in vivo cardiovascular models is important during lead optimization to enable a wide therapeutic index for cardiovascular safety. However, under what conditions (eg, species, route of administration, anesthesia) studies should be performed to drive go/no-go is open to interpretation. Two compounds, torcetrapib and a novel steroid hormone mimetic (SHM-1121X), both with off-target cardiovascular liabilities, were profiled in 4 in vivo cardiovascular models. Overlapping plasma concentrations of torcetrapib were achieved in all models tested; values ranged from therapeutic to supratherapeutic. In anesthetized rats, intravenous torcetrapib elicited dose-dependent increases in mean arterial pressure (MAP; 2-18 mm Hg above vehicle during the low- and high-dose infusion), and in anesthetized dogs, torcetrapib increased MAP from 4 to 22 mm Hg. In conscious rats, a single oral dose of torcetrapib increased MAP from 10 to 18 mm Hg in the low-dose and high-dose groups, respectively, whereas in conscious dogs, MAP increased from 3 to 12 mm Hg. SHM-1121X produced marked hypotension in the same models. Pharmacokinetic-pharmacodynamic analysis demonstrated strong correlation across the models tested for both compounds. Results suggest that equivalency across models allows for flexibility to address key issues and enable go/no-go during lead optimization without concern for discordant results. The predictive value of each model was validated with torcetrapib and, when put into practice, led to a decisive no-go for SHM-1121X.


Subject(s)
Anesthesia/methods , Models, Animal , Quinolines/pharmacology , Administration, Oral , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/pharmacology , Blood Pressure/drug effects , Cardiovascular System/drug effects , Cardiovascular System/metabolism , Dogs , Dose-Response Relationship, Drug , Infusions, Intravenous , Male , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Species Specificity
4.
Front Pharmacol ; 7: 368, 2016.
Article in English | MEDLINE | ID: mdl-27790142

ABSTRACT

Establishing a wide therapeutic index (TI) for pre-clinical safety is important during lead optimization (LO) in research, prior to clinical development, although is often limited by a molecules physiochemical characteristics. Recent advances in the application of the innovative vibrating mesh spray-drying technology to prepare amorphous solid dispersions may offer an opportunity to achieve high plasma concentrations of poorly soluble NCEs to enable testing and establishment of a wide TI in safety pharmacology studies. While some of the amorphous solid dispersion carriers are generally recognized as safe for clinical use, whether they are sufficiently benign to enable in vivo pharmacology studies has not been sufficiently demonstrated. Thus, the physical properties, and effect in a battery of in vivo safety pharmacology models, were assessed in three classes of polymers employed as spray-dried dispersion carriers. The polymers (HPMC-AS, Eudragit, PVAP) displayed low affinity with acetone/methanol, suitable for solvent-based spray drying. The water sorption of the polymers was moderate, and the degree of hysteresis of HPMC-AS was smaller than Eudragit and PVAP indicating the intermolecular interaction of water-cellulose molecules is weaker than water-acrylate or water-polyvinyl molecules. The polymer particles were well-suspended without aggregation with a mean particle size less than 3 µm in an aqueous vehicle. When tested in conscious Wistar Han rats in safety pharmacology models (n = 6-8/dose/polymer) investigating effects on CNS, gastrointestinal, and cardiovascular function, no liabilities were identified at any dose tested (30-300 mg/kg PO, suspension). In brief, the polymers had no effect in a modified Irwin test that included observational and evoked endpoints related to stereotypies, excitation, sedation, pain/anesthesia, autonomic balance, reflexes, and others. No effect of the polymers on gastric emptying or intestinal transit was observed when measured using a barium sulfate tracer material. Finally, in telemetry-instrumented rats the polymers had no effect on acute or 24-h mean blood pressure and heart rate values at doses up to 300 mg/kg. Thus, the properties of the three enteric polymers are appropriate as spray-dried dispersion carriers and were benign in a battery of safety pharmacology studies, demonstrating their applicability to enable in vivo safety pharmacology profiling of poorly soluble molecules during LO.

5.
PLoS One ; 9(8): e105883, 2014.
Article in English | MEDLINE | ID: mdl-25170619

ABSTRACT

Inflammation is associated with immune cells infiltrating into the inflammatory site and pain. CC chemokine receptor 1 (CCR1) mediates trafficking of leukocytes to sites of inflammation. However, the contribution of CCR1 to pain is incompletely understood. Here we report an unexpected discovery that CCR1-mediated trafficking of neutrophils and CCR1 activity on non-hematopoietic cells both modulate pain. Using a genetic approach (CCR1-/- animals) and pharmacological inhibition of CCR1 with selective inhibitors, we show significant reductions in pain responses using the acetic acid-induced writhing and complete Freund's adjuvant-induced mechanical hyperalgesia models. Reductions in writhing correlated with reduced trafficking of myeloid cells into the peritoneal cavity. We show that CCR1 is highly expressed on circulating neutrophils and their depletion decreases acetic acid-induced writhing. However, administration of neutrophils into the peritoneal cavity did not enhance acetic acid-induced writhing in wild-type (WT) or CCR1-/- mice. Additionally, selective knockout of CCR1 in either the hematopoietic or non-hematopoietic compartments also reduced writhing. Together these data suggest that CCR1 functions to significantly modulate pain by controlling neutrophil trafficking to the inflammatory site and having an unexpected role on non-hematopoietic cells. As inflammatory diseases are often accompanied with infiltrating immune cells at the inflammatory site and pain, CCR1 antagonism may provide a dual benefit by restricting leukocyte trafficking and reducing pain.


Subject(s)
Neutrophil Infiltration/immunology , Neutrophils/immunology , Pain/immunology , Receptors, CCR1/immunology , Acetic Acid , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Cell Movement/genetics , Cell Movement/immunology , Flow Cytometry , Freund's Adjuvant , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/immunology , Leukocytes/immunology , Leukocytes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neutrophil Infiltration/genetics , Neutrophils/metabolism , Pain/chemically induced , Pain/genetics , Pain Measurement/methods , Peritonitis/genetics , Peritonitis/immunology , Peritonitis/metabolism , Receptors, CCR1/antagonists & inhibitors , Receptors, CCR1/genetics
6.
PLoS One ; 7(12): e52985, 2012.
Article in English | MEDLINE | ID: mdl-23285242

ABSTRACT

Sphingosine-1-phospate (S1P) and S1P receptor agonists elicit mechanism-based effects on cardiovascular function in vivo. Indeed, FTY720 (non-selective S1P(X) receptor agonist) produces modest hypertension in patients (2-3 mmHg in 1-yr trial) as well as acute bradycardia independent of changes in blood pressure. However, the precise receptor subtypes responsible is controversial, likely dependent upon the cardiovascular response in question (e.g. bradycardia, hypertension), and perhaps even species-dependent since functional differences in rodent, rabbit, and human have been suggested. Thus, we characterized the S1P receptor subtype specificity for each compound in vitro and, in vivo, the cardiovascular effects of FTY720 and the more selective S1P1,5 agonist, BAF312, were tested during acute i.v. infusion in anesthetized rats and after oral administration for 10 days in telemetry-instrumented conscious rats. Acute i.v. infusion of FTY720 (0.1, 0.3, 1.0 mg/kg/20 min) or BAF312 (0.5, 1.5, 5.0 mg/kg/20 min) elicited acute bradycardia in anesthetized rats demonstrating an S1P1 mediated mechanism-of-action. However, while FTY720 (0.5, 1.5, 5.0 mg/kg/d) elicited dose-dependent hypertension after multiple days of oral administration in rat at clinically relevant plasma concentrations (24-hr mean blood pressure = 8.4, 12.8, 16.2 mmHg above baseline vs. 3 mmHg in vehicle controls), BAF312 (0.3, 3.0, 30.0 mg/kg/d) had no significant effect on blood pressure at any dose tested suggesting that hypertension produced by FTY720 is mediated S1P3 receptors. In summary, in vitro selectivity results in combination with studies performed in anesthetized and conscious rats administered two clinically tested S1P agonists, FTY720 or BAF312, suggest that S1P1 receptors mediate bradycardia while hypertension is mediated by S1P3 receptor activation.


Subject(s)
Azetidines/adverse effects , Benzyl Compounds/adverse effects , Bradycardia/chemically induced , Hypertension/chemically induced , Propylene Glycols/adverse effects , Receptors, Lysosphingolipid/agonists , Sphingosine/analogs & derivatives , Animals , Azetidines/pharmacology , Benzyl Compounds/pharmacology , Bradycardia/pathology , Cells, Cultured , Drug Evaluation, Preclinical , Fingolimod Hydrochloride , Humans , Hypertension/pathology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Male , Propylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/classification , Sphingosine/adverse effects , Sphingosine/pharmacology , Substrate Specificity
7.
J Pharmacol Toxicol Methods ; 64(1): 89-96, 2011.
Article in English | MEDLINE | ID: mdl-21376127

ABSTRACT

INTRODUCTION: Long-term administration of non-selective matrix metalloproteinase (MMP) inhibitors, such as marimastat, in humans elicits musculoskeletal syndrome (MSS), a syndrome characterized by joint damage including pain, stiffness, and inflammation. This pathology is a significant obstacle to the clinical development of MMP inhibitors and in pre-clinical models MSS can be verified only after terminal histopathology. Consequently, we devised a longitudinal and functional readout of MSS in conscious rats treated with marimastat that was validated against terminal histological assessment. METHODS: MSS was induced by minipump infusion of marimastat (5-10mg/kg/day). In marimastat-treated or vehicle-control groups, three possible functional biomarkers were assessed: paw volume (PV), landing foot splay separation (LFSS), and rotarod performance (n=6 rats/group for each endpoint). RESULTS: Histologically, fibrosis scores in the synovium and ligament increased from 0 on Day 1 (D1) to 4.6±0.2 and 4.7±0.1, respectively, on D15; growth plate thickness was also elevated from 215.0±6.3µm (D1) to 253.3±8.0µm (D15). While neither PV nor LFSS were correlative with MSS histopathology, marimastat (10mg/kg/day) reduced rotarod performance from 180±0s (D0) to 135±30s (D9) using a constant speed protocol (10rpm, 180s) and from 180±0s (D0) to 96±6s (D6) employing a variable speed protocol (increasing from 5 to 25rpm over 180s). DISCUSSION: Results of the present study demonstrate that rotarod performance can be used as a predictive longitudinal, in vivo functional biomarker of MSS concomitant with histological evidence of joint damage to effectively facilitate compound selection during drug discovery. Moreover, for targets with a mechanistic risk for MSS, the model is also conducive to inclusion in secondary pharmacodynamic studies during lead optimization to identify the best (safest) compounds for advancement into clinical trials.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase Inhibitors , Animals , Biomarkers, Pharmacological/analysis , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/toxicity , Hydroxamic Acids/toxicity , Joints/drug effects , Joints/pathology , Longitudinal Studies , Male , Musculoskeletal Diseases/chemically induced , Musculoskeletal Diseases/diagnosis , Musculoskeletal Diseases/pathology , Musculoskeletal System/drug effects , Musculoskeletal System/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL