Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
ChemMedChem ; 4(5): 853-65, 2009 May.
Article in English | MEDLINE | ID: mdl-19263460

ABSTRACT

Soluble guanylate cyclase (sGC) is a key signal-transduction enzyme activated by nitric oxide (NO). Impairments of the NO-sGC signaling pathway have been implicated in the pathogenesis of cardiovascular and other diseases. Direct stimulation of sGC represents a promising therapeutic strategy particularly for the treatment of pulmonary hypertension (PH), a disabling disease associated with a poor prognosis. Previous sGC stimulators such as the pyrazolopyridines BAY 41-2272 and BAY 41-8543 demonstrated beneficial effects in experimental models of PH, but were associated with unfavorable drug metabolism and pharmacokinetic (DMPK) properties. Herein we disclose an extended SAR exploration of this compound class to address these issues. Our efforts led to the identification of the potent sGC stimulator riociguat, which exhibits an improved DMPK profile and exerts strong effects on pulmonary hemodynamics and exercise capacity in patients with PH. Riociguat is currently being investigated in phase III clinical trials for the oral treatment of PH.


Subject(s)
Pyrimidines/chemistry , Receptors, Cytoplasmic and Nuclear/agonists , Administration, Oral , Animals , Dogs , Drug Discovery , Female , Guanylate Cyclase/metabolism , Hypertension, Pulmonary/drug therapy , Morpholines/chemistry , Morpholines/pharmacology , Nitric Oxide/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rabbits , Rats , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Soluble Guanylyl Cyclase , Structure-Activity Relationship
2.
Arzneimittelforschung ; 58(2): 71-80, 2008.
Article in English | MEDLINE | ID: mdl-18412020

ABSTRACT

UNLABELLED: Liver cirrhosis is a chronic disease with high mortality rate and need for effective pharmacological intervention. The fibrotic remodelling of liver tissue is crucially dependent on hepatic stellate cell activation. Activation of hepatic stellate cells is reduced by an increase in cyclic guanosine monophosphate (cGMP). Stable cGMP analogues also reduce the contractile response of hepatic stellate cells. However, cGMP production is downregulated in the cirrhotic liver due to the reduced activity of the endothelial nitric oxide synthase. OBJECTIVE: Here we report that the novel activator of soluble guanylate cyclase (sGC), BAY 60-2770 (4-({(4-carboxybutyl) [2- (5-fluoro-2-{[4'-(trifluoromethyl) biphenyl-4-yl]methoxy}phenyl)ethyl] amino}methyl)benzoic acid), which increases the activity of sGC in a nitric oxide-independent manner, attenuates liver fibrosis in two rat models. METHODS: The compound was studied in the pig serum model and the carbon tetrachloride model. Fibrosis was assessed by estimating the increase in fibrous collagen by micromorphometry of histological sections stained with Sirius Red/Fast Green and by measuring total hepatic collagen. RESULTS: BAY 60-2770, on a recombinant sGC reporter cell line, stimulated the luminescence signals with an EC50 value of 5.4 +/- 1.2 nmol/L. In the presence of [1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; 10 pmol/L) the EC50 was shifted to 0.39 +/- 0.11 nmol/L. In both fibrosis models, once daily oral administration of BAY 60-2770 concomittantly with the fibrotic stimulus prevented 60-75% of fibrosis, the lowest effective dose being 0.1 mg/kg in the pig serum model and 0.3 mg/kg in the carbon tetrachloride model. The treatment was well tolerated by all animals. The doses used were devoid of any significant influence on systemic blood pressure. CONCLUSION: Nitric oxide-independent activation of sGC might be an innovative therapeutic approach for the treatment of liver fibrosis of necro-inflammatory and immunological origin.


Subject(s)
Benzoates/pharmacology , Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Hydrocarbons, Fluorinated/pharmacology , Liver Cirrhosis, Experimental/enzymology , Nitric Oxide/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Biphenyl Compounds , Blood Pressure/drug effects , Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride Poisoning/enzymology , Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chemistry, Pharmaceutical , Collagen/biosynthesis , Disease Progression , Enzyme Activation/drug effects , Female , Guanylate Cyclase/genetics , Hydroxyproline/metabolism , Liver/enzymology , Liver/pathology , Liver Cirrhosis, Experimental/drug therapy , Liver Cirrhosis, Experimental/pathology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Serum , Soluble Guanylyl Cyclase , Swine
3.
J Org Chem ; 70(23): 9463-9, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16268621

ABSTRACT

[Reaction: see text]. A regioselective and efficient approach toward 6-amino-5-benzoyl-1-substituted 2(1H)-pyridinones by reaction of acyclic ketene aminals with propiolic acid ester was developed. The effect of the solvent and temperature on the regioselectivity of the reaction and the compatibility of the target compounds to functional group manipulations was examined. Substrates with an ortho substituent build atropisomers due to the restricted rotation around the C-N bond. The enantiomers were separated, and the barrier of rotation was determined experimentally. Quantum chemical calculations allowed a ranking of the barrier heights, and a new mechanism of rotation by deformation of the central pyridinone moiety is proposed.


Subject(s)
Pyridones/chemical synthesis , Models, Chemical , Molecular Conformation , Molecular Structure , Stereoisomerism
4.
Anal Biochem ; 339(1): 104-12, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15766716

ABSTRACT

We have established a rapid, homogeneous, cell-based, and highly sensitive assay for guanosine 3'-5'-cyclic monophosphate (cGMP) that is suitable for fully automated ultra-high-throughput screening. In this assay system, cGMP production is monitored in living cells via Ca2+ influx through the olfactory cyclic nucleotide-gated cation channel CNGA2, acting as the intracellular cGMP sensor. A stably transfected Chinese hamster ovary (CHO) cell line was generated recombinantly expressing soluble guanylate cyclase, CNGA2, and aequorin as a luminescence indicator for the intracellular calcium concentration. This cell line was used to screen more than 900,000 compounds in an automated ultra-high-throughput screening assay using 1536-well microtiter plates. In this way, we have been able to identify BAY 58-2667, a member of a new class of amino dicarboxylic acids that directly activate soluble guanylate cyclase. The assay system allows the real-time cGMP detection within living cells and makes it possible to screen for activators and inhibitors of enzymes involved in the nitric oxide/cGMP pathway.


Subject(s)
Cyclic GMP/metabolism , Drug Evaluation, Preclinical , Guanylate Cyclase/metabolism , Ion Channels/metabolism , Nitric Oxide/metabolism , Signal Transduction , Animals , Benzoates/pharmacology , CHO Cells , Calcium/metabolism , Cattle , Cricetinae , Cyclic Nucleotide-Gated Cation Channels , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Ion Channels/genetics , Olfactory Bulb/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
Br J Pharmacol ; 136(5): 773-83, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12086987

ABSTRACT

1. Soluble guanylyl cyclase (sGC) is the only proven receptor for the ubiquitous biological messenger nitric oxide (NO) and is intimately involved in many signal transduction pathways, most notably in regulating vascular tone and platelet function. sGC is a heterodimeric (alpha/ss) protein that converts GTP to cyclic GMP; NO binds to its prosthetic haem group. Here, we report the discovery of a novel sGC activating compound, its interaction with a previously unrecognized regulatory site and its therapeutic implications. 2. Through a high-throughput screen we identified BAY 58-2667, an amino dicarboxylic acid which potently activates sGC in an NO-independent manner. In contrast to NO, YC-1 and BAY 41-2272, the sGC stimulators described recently, BAY 58-2667 activates the enzyme even after it has been oxidized by the sGC inhibitor ODQ or rendered haem deficient. 3. Binding studies with radiolabelled BAY 58-2667 show a high affinity site on the enzyme. 4. Using photoaffinity labelling studies we identified the amino acids 371 (alpha-subunit) and 231 - 310 (ss-subunit) as target regions for BAY 58-2667. 5. sGC activation by BAY 58-2667 results in an antiplatelet activity both in vitro and in vivo and a potent vasorelaxation which is not influenced by nitrate tolerance. 6. BAY 58-2667 shows a potent antihypertensive effect in conscious spontaneously hypertensive rats. In anaesthetized dogs the hemodynamic effects of BAY 58-2667 and GTN are very similar on the arterial and venous system. 7. This novel type of sGC activator is a valuable research tool and may offer a new approach for treating cardiovascular diseases.


Subject(s)
Cardiovascular System/metabolism , Heme/metabolism , Nitric Oxide/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Cardiovascular System/drug effects , Dogs , Enzyme Activators/chemistry , Enzyme Activators/metabolism , Enzyme Activators/pharmacology , Female , Guanylate Cyclase , In Vitro Techniques , Male , Rabbits , Rats , Rats, Inbred SHR , Rats, Wistar , Soluble Guanylyl Cyclase
6.
Br J Pharmacol ; 135(2): 333-43, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11815368

ABSTRACT

BAY 41-8543 is a novel, highly specific and so far the most potent NO-independent stimulator of sGC. Here we report the effects of BAY 41-8543 on the isolated enzyme, endothelial cells, platelets, isolated vessels and Langendorff heart preparation. BAY 41-8543 stimulates the recombinant sGC concentration-dependently from 0.0001 microM to 100 microM up to 92-fold. In combination, BAY 41-8543 and NO have synergistic effects over a wide range of concentrations. Similar results are shown in implying that BAY 41-8543 stimulates the sGC directly and furthermore makes the enzyme more sensitive to its endogenous activator NO. In vitro, BAY 41-8543 is a potent relaxing agent of aortas, saphenous arteries, coronary arteries and veins with IC(50)-values in the nM range. In the rat heart Langendorff preparation, BAY 41-8543 potently reduces coronary perfusion pressure from 10(-9) to 10(-6) g ml(-1) without any effect on left ventricular pressure and heart rate. BAY 41-8543 is effective even under nitrate tolerance conditions proved by the same vasorelaxing effect on aortic rings taken either from normal or nitrate-tolerant rats. BAY 41-8543 is a potent inhibitor of collagen-mediated aggregation in washed human platelets (IC(50)=0.09 microM). In plasma, BAY 41-8543 inhibits collagen-mediated aggregation better than ADP-induced aggregation, but has no effect on the thrombin pathway. BAY 41-8543 is also a potent direct stimulator of the cyclic GMP/PKG/VASP pathway in platelets and synergizes with NO over a wide range of concentrations. These results suggest that BAY 41-8543 is on the one hand an invaluable tool for studying sGC signaling in vitro and on the other hand its unique profile may offer a novel approach for treating cardiovascular diseases.


Subject(s)
Enzyme Activators/pharmacology , Guanylate Cyclase/metabolism , Morpholines/pharmacology , Nitric Oxide , Pyrimidines/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Coronary Vessels/drug effects , Coronary Vessels/enzymology , Dogs , Dose-Response Relationship, Drug , Enzyme Activators/chemistry , Female , Femoral Vein/drug effects , Femoral Vein/enzymology , Heart/drug effects , Heart/physiology , Humans , Male , Nitric Oxide/physiology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Rabbits , Rats , Rats, Wistar , Swine , Vasodilation/drug effects , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...