Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Bioorg Med Chem Lett ; 22(9): 3208-12, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22465635

ABSTRACT

In an effort to identify multi-targeted kinase inhibitors with a novel spectrum of kinase activity, a screen of Abbott proprietary KDR inhibitors against a broad panel of kinases was conducted and revealed a series of thienopyridine ureas with promising activity against the Aurora kinases. Modification of the diphenyl urea and C7 moiety of these compounds provided potent inhibitors with good pharmacokinetic profiles that were efficacious in mouse tumor models after oral dosing. Compound 2 (ABT-348) of this series is currently undergoing Phase I clinical trials in solid and hematological cancer populations.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Urea/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Mice , Protein Kinase Inhibitors/chemistry , Vascular Endothelial Growth Factor A
2.
J Cardiovasc Pharmacol ; 54(6): 543-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770671

ABSTRACT

Torcetrapib is a cholesteryl ester transfer protein inhibitor with an undesired response of increasing arterial pressure in humans. Pressor responses to torcetrapib have been demonstrated in multiple preclinical species. However, these studies have not related plasma concentrations to observed effects. Our purpose was to 1) characterize the cardiovascular responses of torcetrapib in conscious and anesthetized dogs with measured plasma concentrations; and 2) characterize the hemodynamic effects contributing to hypertension using comprehensively instrumented anesthetized dogs. Torcetrapib was dosed orally (3, 30 mg/kg) and intravenously (0.01, 0.33, 0.1 mg/kg) in conscious and anesthetized dogs, respectively. Mean arterial pressure and heart rate were monitored in both models; additional parameters were measured in anesthetized dogs. Plasma drug concentrations were assessed in both models. In conscious and anesthetized dogs, torcetrapib increased mean arterial pressure 25 and 18 mm Hg and heart rate 35 and 21 beats/min, at 2.94 and 3.99 microg/mL, respectively. In anesthetized dogs, torcetrapib increased pulmonary arterial pressure, both systemic and pulmonary hypertension driven by increases in vascular resistance. The compound increased rate pressure product and myocardial contractility while decreasing time to systolic pressure recovery and ejection time. Thus, torcetrapib-induced pressor responses are mediated by systemic and pulmonary vasoconstriction and are associated with increased myocardial oxygen consumption and positive inotropy.


Subject(s)
Anesthesia , Cardiovascular System/drug effects , Hemodynamics/drug effects , Pentobarbital/administration & dosage , Quinolines/pharmacology , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiac Output/drug effects , Cardiac Output/physiology , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Dogs , Electrocardiography , Heart Rate/drug effects , Heart Rate/physiology , Hemodynamics/physiology , Male , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Quinolines/administration & dosage , Quinolines/blood , Quinolines/pharmacokinetics , Telemetry , Vascular Resistance/drug effects , Vascular Resistance/physiology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
3.
J Med Chem ; 49(7): 2339-52, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16570930

ABSTRACT

An inactin-anesthetized rat cardiovascular (CV) assay was employed in a screening mode to triage multiple classes of melanin-concentrating hormone receptor 1 (MCHr1) antagonists. Lead identification was based on a compound profile producing high drug concentration in both plasma (>40 microM) and brain (>20 microg/g) with <15% change in cardiovascular endpoints. As a result of these stringent requirements, lead optimization activities on multiple classes of MCHr1 antagonists were terminated. After providing evidence that the cardiovascular liabilities were not a function of MCHr1 antagonism, continued screening identified the chromone-substituted aminopiperidine amides as a class of MCHr1 antagonists that demonstrated a safe cardiovascular profile at high drug concentrations in both plasma and brain. The high incidence of adverse cardiovascular effects associated with an array of MCHr1 antagonists of significant chemical diversity, combined with the stringent safety requirements for antiobesity drugs, highlight the importance of incorporating cardiovascular safety assessment early in the lead selection process.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Cardiovascular System/drug effects , Chromones/chemical synthesis , Piperidines/chemical synthesis , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/blood , Blood Pressure/drug effects , Brain/metabolism , Cell Line, Tumor , Chromones/adverse effects , Chromones/blood , Dogs , Indazoles/adverse effects , Indazoles/blood , Indazoles/chemical synthesis , Male , Mice , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Piperidines/adverse effects , Piperidines/blood , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Tissue Distribution
4.
J Med Chem ; 49(22): 6439-42, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064063

ABSTRACT

Dipeptidyl peptidase IV (DPP4) deactivates glucose-regulating hormones such as GLP-1 and GIP, thus, DPP4 inhibition has become a useful therapy for type 2 diabetes. Optimization of the high-throughput screening lead 6 led to the discovery of 25 (ABT-341), a highly potent, selective, and orally bioavailable DPP4 inhibitor. When dosed orally, 25 dose-dependently reduced glucose excursion in ZDF rats. Amide 25 is safe in a battery of in vitro and in vivo tests and may represent a new therapeutic agent for the treatment of type 2 diabetes.


Subject(s)
Biphenyl Compounds/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/metabolism , Hypoglycemic Agents/pharmacology , Serine Proteinase Inhibitors/pharmacology , Triazoles/pharmacology , Animals , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacokinetics , Cyclohexenes/chemistry , Diabetes Mellitus, Type 2/genetics , Dose-Response Relationship, Drug , Drug Design , Drug Evaluation, Preclinical , Female , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Models, Molecular , Rats , Rats, Zucker , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/pharmacokinetics , X-Ray Diffraction
5.
J Med Chem ; 49(22): 6569-84, 2006 Nov 02.
Article in English | MEDLINE | ID: mdl-17064075

ABSTRACT

Evaluation of multiple structurally distinct series of melanin concentrating hormone receptor 1 antagonists in an anesthetized rat cardiovascualar assay led to the identification of a chromone-2-carboxamide series as having excellent safety against the chosen cardiovascular endpoints at high drug concentrations in the plasma and brain. Optimization of this series led to considerable improvements in affinity, functional potency, and pharmacokinetic profile. This led to the identification of a 7-fluorochromone-2-carboxamide (22) that was orally efficacious in a diet-induced obese mouse model, retained a favorable cardiovascular profile in rat, and demonstrated dramatic improvement in effects on mean arterial pressure in our dog cardiovascular model compared to other series reported by our group. However, this analogue also led to prolongation of the QT interval in the dog that was linked to affinity for hERG channel and unexpectedly potent functional blockade of this ion channel.


Subject(s)
Benzodioxoles/pharmacology , Cardiovascular Diseases/chemically induced , Chromones/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Acylation , Animals , Area Under Curve , Benzodioxoles/pharmacokinetics , Benzodioxoles/toxicity , Blood Pressure/drug effects , Body Weight/drug effects , Calcium/metabolism , Cell Line , Chromones/pharmacokinetics , Chromones/toxicity , Dogs , Electrocardiography/drug effects , Female , Half-Life , Heart Rate/drug effects , Indicators and Reagents , Mice , Mice, Inbred C57BL , Potassium Channels/drug effects , Potassium Channels/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
6.
J Med Chem ; 48(19): 5888-91, 2005 Sep 22.
Article in English | MEDLINE | ID: mdl-16161992

ABSTRACT

4-(1-Benzo[1,3]dioxol-5-ylmethylpiperidine-4-ylmethyl)-6-chlorochromen-2-one (7) is a potent, orally bioavailable melanin concentrating hormone receptor 1 (MCHr1) antagonist that causes dose-dependent weight loss in diet-induced obese mice. Further evaluation of 7 in an anesthetized dog model of cardiovascular safety revealed adverse hemodynamic effects at a plasma concentration comparable to the minimally effective therapeutic concentration. These results highlight the need for scrutiny of the cardiovascular safety profile of MCHr1 antagonists.


Subject(s)
Coumarins/chemical synthesis , Piperidines/chemical synthesis , Receptors, Pituitary Hormone/antagonists & inhibitors , Receptors, Somatostatin/antagonists & inhibitors , Administration, Oral , Animals , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacology , Biological Availability , Blood Pressure/drug effects , Brain/metabolism , Cell Line, Tumor , Coumarins/adverse effects , Coumarins/pharmacology , Dogs , Eating/drug effects , Energy Metabolism , Humans , Male , Mice , Mice, Obese , Myocardial Contraction/drug effects , Piperidines/adverse effects , Piperidines/pharmacology , Radioligand Assay , Structure-Activity Relationship
7.
Am J Physiol Heart Circ Physiol ; 294(1): H238-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17982006

ABSTRACT

Levosimendan enhances cardiac contractility via Ca(2+) sensitization and induces vasodilation through the activation of ATP-dependent K(+) and large-conductance Ca(2+)-dependent K(+) channels. However, the hemodynamic effects of levosimendan, as well as its metabolites, OR-1896 and OR-1855, relative to plasma concentrations achieved, are not well defined. Thus levosimendan, OR-1896, OR-1855, or vehicle was infused at 0.01, 0.03, 0.1, and 0.3 mumol.kg(-1).30 min(-1), targeting therapeutic to supratherapeutic concentrations of total levosimendan (62.6 ng/ml). Results were compared with those of the beta(1)-agonist dobutamine and the phosphodiesterase 3 inhibitor milrinone. Peak concentrations of levosimendan, OR-1896, and OR-1855 were 455 +/- 21, 126 +/- 6, and 136 +/- 6 ng/ml, respectively. Levosimendan and OR-1896 produced dose-dependent reductions in mean arterial pressure (-31 +/- 2 and -42 +/- 3 mmHg, respectively) and systemic resistance without affecting pulse pressure, effects paralleled by increases in heart rate; OR-1855 produced no effect at any dose tested. Dobutamine, but not milrinone, increased mean arterial pressure and pulse pressure (17 +/- 2 and 23 +/- 2 mmHg, respectively). Regarding potency to elicit reductions in time to peak pressure and time to systolic pressure recovery: OR-1896 > levosimendan > milrinone > dobutamine. Levosimendan and OR-1896 elicited dose-dependent increases in change in pressure over time (118 +/- 10 and 133 +/- 13%, respectively), concomitant with reductions in left ventricular end-diastolic pressure and ejection time. However, neither levosimendan nor OR-1896 produced increases in myocardial oxygen consumption at inotropic and vasodilatory concentrations, whereas dobutamine increased myocardial oxygen consumption (79% above baseline). Effects of the levosimendan and OR-1896 were limited to the systemic circulation; neither compound produced changes in pulmonary pressure, whereas dobutamine produced profound increases (74 +/- 13%). Thus levosimendan and OR-1896 are hemodynamically active in the anesthetized dog at concentrations observed clinically and elicit cardiovascular effects consistent with activation of both K(+) channels and Ca(2+) sensitization, whereas OR-1855 is inactive on endpoints measured in this study.


Subject(s)
Acetamides/pharmacology , Cardiotonic Agents/pharmacology , Cardiovascular System/drug effects , Dobutamine/pharmacology , Hydrazones/pharmacology , Milrinone/pharmacology , Oxygen Consumption/drug effects , Pyridazines/pharmacology , Vascular Resistance/drug effects , Vasodilator Agents/pharmacology , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cardiac Output/drug effects , Cardiotonic Agents/blood , Cardiovascular System/metabolism , Dogs , Dose-Response Relationship, Drug , Heart Rate/drug effects , Hydrazones/blood , Male , Myocardial Contraction/drug effects , Potassium/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Pulmonary Circulation/drug effects , Pyridazines/blood , Simendan , Time Factors , Vasodilator Agents/blood , Ventricular Function, Left/drug effects
8.
Clin Exp Pharmacol Physiol ; 32(12): 1020-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16445566

ABSTRACT

Iron-deficiency anaemia, a complication of end-stage renal disease (ESRD), is often treated with parenteral iron therapies that have been shown to produce dose-limiting hypotension in patients. ABT-870 (iron-(III)-hydroxide-oligosaccharide) is comprised of elemental iron complexed with oligosaccharide, a composition that we hypothesised would allow the hypotensive effects of parenteral iron therapy to be overcome, thus allowing a rapid rate of infusion to be well tolerated. Mean arterial pressure (MAP) and heart rate (HR) were monitored in anaesthetized dogs following the infusion of ABT-870 and iron sucrose administered at doses of 7.1 and 21.3 mg/kg using a rapid 30 s infusion. ABT-870 and iron sucrose were also monitored at doses of 7.1, 21.3 and 50 mg/kg administered over a 10 min period. Sodium ferric gluconate complex (SFGC) was administered in an identical fashion at doses of 12.5 and 31.2 mg/kg. A 30 s rapid infusion of ABT-870 at doses of 7.1 and 14.3 mg/kg or a 10 min infusion of ABT-870 at doses of 7.1 and 21.3 mg/kg produced little effect on MAP and HR. Infusion of the highest dose of ABT-870 (50 mg/kg) produced no consistent hypotension, but did produce an increase in HR (maximal increase 35 +/- 9 b.p.m.), an effect that lasted only 15 min. A 30 s rapid infusion of iron sucrose at 7.1 mg/kg produced modest increases in MAP and HR (5 +/- 1 mmHg and 5 +/- 2 b.p.m., respectively). However, rapid infusion of iron sucrose at 14.3 mg/kg produced hypotension (to -8 +/- 1 mmHg below baseline) and exerted variable, biphasic effects on HR ranging from -16 to +50 b.p.m. Although 10 min infusion of iron sucrose at 7.1 mg/kg exerted little effect on MAP and HR, at doses of 21.3 and 50 mg/kg iron sucrose elicited a profound dose-dependent decrease in MAP (-34 +/- 11 and -83 +/- 5 mmHg, respectively) and a pronounced increase in HR ranging from 32 to 49 b.p.m. above baseline. A 10 min infusion of SFGC at doses of 12.5 and 31.2 mg/kg produced a dose-dependent decrease in MAP (-28 +/- 18 and -67 +/- 12 mmHg below baseline) and a marked increase in HR (26 +/- 11 and 94 +/- 15 b.p.m. above baseline). In conclusion, unlike iron sucrose and SFGC, high doses of ABT-870 failed to exert consistent hypotensive effects. These data demonstrate that ABT-870 may have a substantial therapeutic window and considerable clinical potential for iron-replacement therapy.


Subject(s)
Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Ferric Compounds/pharmacology , Heart Rate/drug effects , Hematinics/pharmacology , Iron Compounds/pharmacology , Oligosaccharides/pharmacology , Animals , Dextrans , Dogs , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Infusions, Intravenous , Male , Oligosaccharides/administration & dosage
9.
J Cardiovasc Pharmacol ; 46(2): 232-40, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16044036

ABSTRACT

The pharmacological profile of the novel dihydropyridine K channel opener (KCO), (9R)-9-(3-iodo-4-methylphenyl)-5,9-dihydro-3H-furo[3,4-b]pyrano[4,3-e]pyridine-1,8(4H,7H)-dione (A-325100), is described in numerous in vitro assays. Furthermore, the cardiovascular effects of A-325100 are characterized in both the anesthetized and conscious dog. In vitro, A-325100 selectively activated KATP currents and potently relaxed vascular smooth muscle (IC50 between 7.69x10 M and 7.78x10 M), an effect that was abolished by glyburide. Moreover, A-325100 did not interact with L-type Ca2+ channels at concentrations up to 30 microM. In anesthetized dogs A-325100 produced a dose-dependent reduction in systemic vascular resistance and mean arterial pressure concomitant with dose-dependent increases in dP/dtmax and heart rate. In conscious telemetry-instrumented dogs oral administration of A-325100 produced a similar response profile, including dose-dependent reductions in MAP and increases in heart rate and dP/dtmax. When concentration-dependent changes in MAP, heart rate, and dP/dtmax were compared relative to circulating plasma concentrations, A-325100 produced similar effects in both the anesthetized and conscious dog. In conclusion, the present study provides the first pharmacological description of the novel and selective tricyclic dihydropyridine KCO, A-325100. When studied in vivo, A-325100 produced similar concentration-dependent cardiovascular effects in both models consistent with its mode of action and independent of route of administration. Thus, these data demonstrate that the hemodynamic effects of vasoactive compounds, such as KCOs, can be effectively profiled in both the conscious and anesthetized dog.


Subject(s)
Dihydropyridines/pharmacology , Hemodynamics/drug effects , Ion Channel Gating/drug effects , Potassium Channels/metabolism , Pyrones/pharmacology , Adenosine Triphosphate/metabolism , Anesthesia , Animals , Aorta, Thoracic/drug effects , Blood Pressure/drug effects , Dihydropyridines/blood , Dogs , Dose-Response Relationship, Drug , Guinea Pigs , Heart Rate/drug effects , In Vitro Techniques , Ligands , Male , Muscle Relaxation/drug effects , Muscle, Smooth/drug effects , Patch-Clamp Techniques , Portal Vein/drug effects , Pyrones/blood , Radioligand Assay , Rats , Rats, Sprague-Dawley , Urinary Bladder/drug effects , Urinary Bladder/physiology
10.
Am J Physiol Heart Circ Physiol ; 283(4): H1555-61, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12234809

ABSTRACT

The role of endothelin-B (ET(B)) receptors in circulatory homeostasis is ambiguous, reflecting vasodilator and constrictor effects ascribed to the receptor and diuretic and natriuretic responses that could oppose the hypertensive effects of ET excess. With the use of conscious, telemetry-instrumented cynomolgus monkeys, we characterized the hypertension produced by ET(B) blockade and the role of ET(A) receptors in mediating this response. Mean arterial pressure (MAP) and heart rate (HR) were measured 24 h/day for 24 days under control conditions and during administration of the ET(B)-selective antagonist A-192621 (0.1, 1.0, and 10 mg/kg bid, 4 days/dose) followed by coadministration of the ET(A) antagonist atrasentan (5 mg/kg bid) + A-192621 (10 mg/kg bid) for another 4 days. High-dose ET(B) blockade increased MAP from 79 +/- 3 (control) to 87 +/- 3 and 89 +/- 3 mmHg on the first and fourth day, respectively; HR was unchanged, and plasma ET-1 concentration increased from 2.1 +/- 0.3 pg/ml (control) to 7.24 +/- 0.99 and 11.03 +/- 2.37 pg/ml. Atrasentan + A-192621 (10 mg/kg) decreased MAP from hypertensive levels (89 +/- 3) to 75 +/- 2 and 71 +/- 4 mmHg on the first and fourth day, respectively; plasma ET-1 and HR increased to 26.64 +/- 3.72 and 28.65 +/- 2.89 pg/ml and 113 +/- 5 (control) to 132 +/- 5 and 133 +/- 7 beats/min. Thus systemic ET(B) blockade produces a sustained hypertension in conscious nonhuman primates, which is mediated by ET(A) receptors. These data suggest an importance clearance function for ET(B) receptors, one that influences arterial pressure homeostasis indirectly by reducing plasma ET-1 levels and minimizing ET(A) activation.


Subject(s)
Endothelin Receptor Antagonists , Hypertension/physiopathology , Receptors, Endothelin/physiology , Animals , Antineoplastic Agents/pharmacology , Atrasentan , Blood Pressure/drug effects , Blood Pressure/physiology , Consciousness , Endothelin-1/blood , Heart Rate/drug effects , Heart Rate/physiology , Homeostasis , Hypertension/chemically induced , Macaca fascicularis , Male , Pyrrolidines/pharmacology , Receptor, Endothelin A , Receptor, Endothelin B , Telemetry
11.
J Cardiovasc Pharmacol ; 44(2): 137-47, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15243293

ABSTRACT

The therapeutic utility of KATP channel opening agents (KCOs) in the treatment of overactive bladder may be limited by hypotension as a result of insufficient selectivity in vivo for bladder versus vasculature smooth muscle. Recently, we demonstrated that the putative uroselective KCOs, A-278637, ZD-6169, and WAY-133537 suppress unstable bladder contraction in an in vivo pre-clinical pig model of detrusor instability secondary to partial outlet obstruction. In the present study in the anesthetized dog we targeted plasma concentrations 3-, 10-, and 30-fold above a common index of in vivo efficacy (EC35) for suppression of unstable bladder contraction in pigs, to provide a comprehensive cardiovascular profile of these compounds. When compared at similar multiples of efficacy, dose-dependent reductions in SVR were greater in ZD-6169 and WAY-133537-treated animals versus A-278637. A-278637, unlike ZD-6169 or WAY-133537, produced no effect on MAP at concentrations 10-fold above the EC35. At concentrations 30-fold above the EC35, MAP in A-278637-treated animals was reduced -11% from baseline versus -24% and -42% for ZD-6169 and WAY-133537. Accordingly, at plasma concentrations approximately 30-fold above the EC35 reflex-mediated increases in HR were modest for A-278637-treated animals (15% above baseline) versus ZD-6169 (22%) or WAY-133537 (35%). Increases in both dP/dt and cardiac output occurred at lower therapeutic multiples and were greater in magnitude for animals treated with WAY-133537 (66% and 64% above baseline, respectively, 60 minutes into compound infusion) and ZD-6169 (10% and 13%) versus A-278637 (-2% and 6%). Thus, A-278637 exerted lesser effects on cardiovascular function at equivalent multiples of the EC35 than either ZD-6169 or WAY-133537. These data suggest that A-278637 possesses a greater functional selectivity for urinary bladder versus vascular smooth muscle in vivo and that A-278637 may exhibit a more favorable therapeutic index than either ZD-6169 or WAY-133537.


Subject(s)
Amides/pharmacology , Benzophenones/pharmacology , Cyclic S-Oxides/pharmacology , Cyclobutanes/pharmacology , Nifedipine/pharmacology , Nitriles/pharmacology , Potassium Channels/physiology , Quinolones/pharmacology , Adenosine Triphosphate/chemistry , Amides/blood , Animals , Benzophenones/blood , Blood Pressure/drug effects , Cyclic S-Oxides/blood , Cyclobutanes/blood , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electrocardiography/methods , Heart Rate/drug effects , Hypotension/chemically induced , Infusions, Intravenous , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Nifedipine/blood , Nitriles/blood , Pharmaceutical Vehicles/administration & dosage , Pharmaceutical Vehicles/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Potassium Channels/drug effects , Quinolones/blood , Tachycardia/chemically induced , Urinary Bladder/drug effects , Urinary Bladder/physiology , Urinary Incontinence/drug therapy , Urinary Incontinence/physiopathology , Vascular Resistance/drug effects
12.
CNS Drug Rev ; 10(2): 167-82, 2004.
Article in English | MEDLINE | ID: mdl-15179445

ABSTRACT

ABT-089 [2-methyl-3-(2-(S)-pyrrolidinylmethoxy)pyridine dihydrochloride salt] is a selective neuronal nicotinic receptor (NNR) modulator with cognitive enhancing properties in animal models of cognitive functioning. Amongst NNR subtypes, ABT-089 shows selectivity for the cytisine binding site on the alpha4beta2 receptor subtype as compared to the alpha-bungarotoxin (alpha-BgT) binding sites on the alpha7 and alpha1beta1deltagamma receptor subtypes. In functional in vitro electrophysiological and cation flux assays, ABT-089 displays differential activity including agonism, partial agonism and antagonism depending upon the NNR subtype and assay. ABT-089 is as potent and efficacious as (-)-nicotine at evoking acetylcholine (ACh) release from hippocampal synaptosomes. Furthermore, ABT-089 is neuroprotective against excitotoxic glutamate insults, with even greater potency seen after chronic treatment. Similarly, ABT-089 is effective in models of cognitive functioning, including enhancement of baseline functioning as well as improvement of impaired cognitive functioning seen following septal lesioning and natural aging. In neuroprotective assays the compound is most potent by chronic administration. In stark contrast to the positive effects in the cognitive models, ABT-089 shows little propensity to induce adverse effects such as ataxia, hypothermia, seizures, cardiovascular or gastrointestinal side effects. Together these data suggest that ABT-089 is a NNR modulator with the potential for treating cognitive disorders with markedly limited adverse cardiovascular and gastrointestinal side effects.


Subject(s)
Cognition Disorders/drug therapy , Digestive System/drug effects , Learning/drug effects , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Pyrrolidines/pharmacology , Animals , Cardiovascular System/drug effects , Dogs , Dose-Response Relationship, Drug , Humans , Mice , Nicotinic Agonists/chemistry , Nicotinic Agonists/therapeutic use , Pyridines/chemistry , Pyridines/therapeutic use , Pyrrolidines/chemistry , Pyrrolidines/therapeutic use , Rats
SELECTION OF CITATIONS
SEARCH DETAIL