Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 274(1): 78-86, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24200993

ABSTRACT

Drugs that block the cardiac K(+) channel encoded by the human ether-à-go-go gene (hERG) have been associated with QT interval prolongation leading to proarrhythmia, and in some cases, sudden cardiac death. Because of special structural features of the hERG K(+) channel, it has become a promiscuous target that interacts with pharmaceuticals of widely varying chemical structures and a reason for concern in the pharmaceutical industry. The structural diversity suggests that multiple binding sites are available on the channel with possible allosteric interactions between them. In the present study, three reference compounds and nine compounds of a previously disclosed series were evaluated for their allosteric effects on the binding of [(3)H]astemizole and [(3)H]dofetilide to the hERG K(+) channel. LUF6200 was identified as an allosteric inhibitor in dissociation assays with both radioligands, yielding similar EC50 values in the low micromolar range. However, potassium ions increased the binding of the two radioligands in a concentration-dependent manner, and their EC50 values were not significantly different, indicating that potassium ions behaved as allosteric enhancers. Furthermore, addition of potassium ions resulted in a concentration-dependent leftward shift of the LUF6200 response curve, suggesting positive cooperativity and distinct allosteric sites for them. In conclusion, our investigations provide evidence for allosteric modulation of the hERG K(+) channel, which is discussed in the light of findings on other ion channels.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Phenethylamines/metabolism , Potassium Channel Blockers/metabolism , Radioligand Assay/methods , Sulfonamides/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Dose-Response Relationship, Drug , ERG1 Potassium Channel , HEK293 Cells , Humans , Phenethylamines/chemistry , Phenethylamines/pharmacology , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Protein Binding/physiology , Sulfonamides/chemistry , Sulfonamides/pharmacology
2.
J Med Chem ; 56(23): 9427-40, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24224763

ABSTRACT

Cardiotoxicity is a side effect that plagues modern drug design and is very often due to the off-target blockade of the human ether-à-go-go related gene (hERG) potassium channel. To better understand the structural determinants of this blockade, we designed and synthesized a series of 40 derivatives of clofilium, a class III antiarrhythmic agent. These were evaluated in radioligand binding and patch-clamp assays to establish structure-affinity relationships (SAR) for this potassium channel. Efforts were especially focused on studying the influence of the structural rigidity and the nature of the linkers composing the clofilium scaffold. It was shown that introducing triple bonds and oxygen atoms in the n-butyl linker of the molecule greatly reduced affinity without significantly modifying the pKa of the essential basic nitrogen. These findings could prove useful in the first stages of drug discovery as a systematic way of reducing the risk of hERG K(+) channel blockade-induced cardiotoxicity.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Ether-A-Go-Go Potassium Channels/drug effects , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , Quaternary Ammonium Compounds/chemistry , Structure-Activity Relationship
3.
J Med Chem ; 56(7): 2828-40, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23473309

ABSTRACT

Drug-induced blockade of the human ether-a-go-go-related gene K(+) channel (hERG) represents one of the major antitarget concerns in pharmaceutical industry. SAR studies of this ion channel have shed light on the structural requirements for hERG interaction but most importantly may reveal drug design principles to reduce hERG affinity. In the present study, a novel library of neutral and positively charged heteroaromatic derivatives of the class III antiarrhythmic agent dofetilide was synthesized and assessed for hERG affinity in radioligand binding and manual patch clamp assays. Structural modifications of the pyridine moiety, side chain, and peripheral aromatic moieties were evaluated, thereby revealing approaches for reducing hERG binding affinity. In particular, we found that the extra rigidity imposed close to the positively charged pyridine moiety can be very efficient in decreasing hERG affinity.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Phenethylamines/pharmacology , Potassium Channel Blockers/pharmacology , Sulfonamides/pharmacology , ERG1 Potassium Channel , HEK293 Cells , Humans , Patch-Clamp Techniques , Phenethylamines/chemistry , Potassium Channel Blockers/chemistry , Radioligand Assay , Sulfonamides/chemistry
4.
ChemMedChem ; 7(1): 107-13, 2012 Jan 02.
Article in English | MEDLINE | ID: mdl-21919210

ABSTRACT

Cardiotoxicity is a common side effect of a large variety of drugs that is often caused by off-target human ether-à-go-go-related gene (hERG) potassium channel blockade. In this study, we designed and synthesized a series of derivatives of the class III antiarrhythmic agent E-4031. These compounds where evaluated in a radioligand binding assay and automated patch clamp assay to establish structure-activity relationships (SAR) for their inhibition of the hERG K(+) channel. Structural modifications of E-4031 were made by altering the peripheral aromatic moieties with a series of distinct substituents. Additionally, we synthesized several derivatives with a quaternary nitrogen and modified the center of the molecule by introduction of an additional nitrogen and deletion of the carbonyl oxygen. Some modifications caused a great increase in affinity for the hERG K(+) channel, while other seemingly minor changes led to a strongly diminished affinity. Structures with quaternary amines carrying an additional aromatic moiety were found to be highly active in radioligand binding assay. A decrease in affinity was achieved by introducing an amide functionality in the central scaffold without directly interfering with the pK(a) of the essential basic amine. The knowledge gained from this study could be used in early stages of drug discovery and drug development to avoid or circumvent hERG K(+) channel blockade, thereby reducing the risk of cardiotoxicity, related arrhythmias and sudden death.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Anti-Arrhythmia Agents/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Piperidines/chemistry , Piperidines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Anti-Arrhythmia Agents/chemical synthesis , Arrhythmias, Cardiac/drug therapy , HEK293 Cells , Humans , Piperidines/chemical synthesis , Pyridines/chemical synthesis , Structure-Activity Relationship
5.
Biochem Pharmacol ; 80(8): 1180-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20599769

ABSTRACT

The adenosine A(1) receptor is a promising therapeutic target for neurological disorders such as cognition deficits and is involved in cardiovascular preconditioning. Classically adenosine receptor agonists were all derivatives of adenosine, and thought to require a D-ribose moiety. More recently, however, the discovery of non-adenosine agonists for the human adenosine A(1) receptor (hA(1)R) has challenged this dogma (Beukers et al., 2004). In this study we characterize the tritiated form of one of these compounds, [(3)H]LUF5834, as the first non-ribose partial agonist radioligand with nanomolar affinity for the hA(1)R. Due to its partial agonist efficacy, [(3)H]LUF5834 labeled both G protein-coupled and uncoupled receptors with a similar high affinity. Using [(3)H]LUF5834 we performed competition binding experiments to characterize a range of A(1)R ligands varying in efficacy from the full agonist CPA to the inverse agonist DPCPX. Surprisingly, in the control condition both agonists and inverse agonists displayed biphasic isotherms. With the addition of 1mM GTP the high affinity isotherm of agonists or the low affinity isotherm of inverse agonists was lost revealing the mechanism of action of such inverse agonists at the A(1)R. Consequently, [(3)H]LUF5834 represents a novel high affinity radioligand for the A(1)R and may prove a useful tool to provide estimates of inverse agonist efficacy at this receptor.


Subject(s)
Adenosine A1 Receptor Agonists , Aminopyridines/pharmacology , Imidazoles/pharmacology , Adenine/analogs & derivatives , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Aminopyridines/chemistry , Animals , Cell Line , Cell Membrane , Cricetinae , Gene Expression Regulation , Humans , Imidazoles/chemistry , Radioligand Assay , Receptor, Adenosine A1/genetics , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism
6.
ChemMedChem ; 5(5): 716-29, 2010 May 03.
Article in English | MEDLINE | ID: mdl-20349498

ABSTRACT

Ligand-based in silico hERG models were generated for 2 644 compounds using linear discriminant analysis (LDA) and support vector machines (SVM). As a result, the dataset used for the model generation is the largest publicly available (see Supporting Information). Extended connectivity fingerprints (ECFPs) and functional class fingerprints (FCFPs) were used to describe chemical space. All models showed area under curve (AUC) values ranging from 0.89 to 0.94 in a fivefold cross-validation, indicating high model consistency. Models correctly predicted 80 % of an additional, external test set; Y-scrambling was also performed to rule out chance correlation. Additionally models based on patch clamp data and radioligand binding data were generated separately to analyze their predictive ability when compared to combined models. To experimentally validate the models, 50 of the predicted hERG blockers from the Chembridge database and ten of the predicted non-hERG blockers from an in-house compound library were selected for biological evaluation. Out of those 50 predicted hERG blockers, tested at a concentration of 10 microM, 18 compounds showed more than 50 % displacement of [(3)H]astemizole binding to cell membranes expressing the hERG channel. K(i) values of four of the selected binders were determined to be in the micromolar and high nanomolar range (K(i) (VH01)=2.0 microM, K(i) (VH06)=0.15 microM, K(i) (VH19)=1.1 microM and K(i) (VH47)=18 microM). Of these four compounds, VH01 and VH47 showed also a second, even higher affinity binding site with K(i) values of 7.4 nM and 36 nM, respectively. In the case of non-hERG blockers, all ten compounds tested were found to be inactive, showing less than 50 % displacement of [(3)H]astemizole binding at 10 microM. These experimentally validated models were then used to virtually screen commercial compound databases to evaluate whether they contain hERG blockers. 109 784 (23 %) of Chembridge, 133 175 (38 %) of Chemdiv, 111 737 (31 %) of Asinex and 11 116 (18 %) of the Maybridge database were predicted to be hERG blockers by at least two of the models, a prediction which could, for example, be used as a pre-filtering tool for compounds with potential hERG liabilities.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Models, Chemical , Potassium Channel Blockers/chemistry , Databases, Factual , Discriminant Analysis , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Ligands , Models, Molecular , Potassium Channel Blockers/pharmacology
7.
ChemMedChem ; 4(10): 1722-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19725081

ABSTRACT

In this study we followed a new approach to analyze molecular substructures required for hERG channel blockade. We designed and synthesized 40 analogues of dofetilide (1), a potent hERG potassium channel blocker, and established structure-activity relationships (SAR) for their interaction with this important cardiotoxicity-related off-target. Structural modifications to dofetilide were made by diversifying the substituents on the phenyl rings and the protonated nitrogen and by varying the carbon chain length. The analogues were evaluated in a radioligand binding assay and SAR data were derived with the aim to specify structural features that give rise to hERG toxicity.


Subject(s)
Drug Design , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Phenethylamines/chemistry , Potassium Channel Blockers/chemistry , Sulfonamides/chemistry , Cell Line , ERG1 Potassium Channel , Humans , Molecular Structure , Phenethylamines/chemical synthesis , Phenethylamines/pharmacology , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology
8.
Purinergic Signal ; 4(1): 21-37, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18368531

ABSTRACT

Until now, more than 800 distinct G protein-coupled receptors (GPCRs) have been identified in the human genome. The four subtypes of the adenosine receptor (A(1), A(2A), A(2B) and A(3) receptor) belong to this large family of GPCRs that represent the most widely targeted pharmacological protein class. Since adenosine receptors are widespread throughout the body and involved in a variety of physiological processes and diseases, there is great interest in understanding how the different subtypes are regulated, as a basis for designing therapeutic drugs that either avoid or make use of this regulation. The major GPCR regulatory pathway involves phosphorylation of activated receptors by G protein-coupled receptor kinases (GRKs), a process that is followed by binding of arrestin proteins. This prevents receptors from activating downstream heterotrimeric G protein pathways, but at the same time allows activation of arrestin-dependent signalling pathways. Upon agonist treatment, adenosine receptor subtypes are differently regulated. For instance, the A(1)Rs are not (readily) phosphorylated and internalize slowly, showing a typical half-life of several hours, whereas the A(2A)R and A(2B)R undergo much faster downregulation, usually shorter than 1 h. The A(3)R is subject to even faster downregulation, often a matter of minutes. The fast desensitization of the A(3)R after agonist exposure may be therapeutically equivalent to antagonist occupancy of the receptor. This review describes the process of desensitization and internalization of the different adenosine subtypes in cell systems, tissues and in vivo studies. In addition, molecular mechanisms involved in adenosine receptor desensitization are discussed.

9.
Eur J Pharmacol ; 522(1-3): 1-8, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16214128

ABSTRACT

To study the effect of allosteric modulators on the internalization of human adenosine A(1) receptors, the receptor was equipped with a C-terminal yellow fluorescent protein tag. The introduction of this tag did not affect the radioligand binding properties of the receptor. CHO cells stably expressing this receptor were subjected during 16 h to varying concentrations of the agonist N(6)-cyclopentyladenosine (CPA) in the absence or presence of 10 microM of the allosteric enhancer PD 81,723 ((2-amino-4,5-dimethyl-3-thienyl)-[3-(trifluoromethyl)phenyl]methanone) or the allosteric inhibitor SCH-202676 (N-(2,3-diphenyl-1,2,4-thiadiazol-5(2H)-ylidene)methanamine). CPA itself was able to internalize 25% and 40% of the receptors at a concentration of 400 nM or 4 muM, respectively. Addition of either PD 81,723 or SCH-202676 alone had no effect on internalization. However, with PD 81,723 a slight amount of internalization was obtained already at 40 nM of CPA and at 400 nM CPA 59% of the receptors internalized. SCH-202676 on the other hand effectively prevented CPA-induced internalization of the receptor.


Subject(s)
Endocytosis/physiology , Receptor, Adenosine A1/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Allosteric Regulation , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Endocytosis/drug effects , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Radioligand Assay , Receptor, Adenosine A1/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thiadiazoles , Thiazoles/pharmacology , Thiophenes/pharmacology , Transfection , Tritium , Xanthines/metabolism
10.
Eur J Pharmacol ; 499(1-2): 91-8, 2004 Sep 19.
Article in English | MEDLINE | ID: mdl-15363955

ABSTRACT

We studied fusion proteins between the human adenosine A1 receptor and different 351Cys-mutated G(i1) alpha-subunits (A1-Gialpha) with respect to two important concepts in receptor pharmacology, i.e. allosteric modulation and constitutive activity/inverse agonism. The aim of our study was twofold. We first analysed whether such fusion products are still subject to allosteric modulation, and, secondly, we investigated the potential utility of the fusion proteins to study constitutive receptor activity. We determined the pharmacological profile of nine different A1-Gialpha fusion proteins in radioligand binding studies. In addition, we performed [35S]GTPgammaS binding experiments to study receptor and G protein activation of selected A1-Gialpha fusion proteins. Compared to unfused adenosine A1 receptors, the affinity of N6-cyclopentyladenosine (CPA) at wild-type A1-Gialpha fusion proteins (351Cys) increased more than eightfold, while the affinity of 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) did not change significantly. Furthermore, we showed that the allosteric enhancer of agonist binding, PD81,723 (2-amino-4,5-dimethyl-3-thienyl-[3-(trifluoromethyl)-phenyl]methanone), elicited similar effects on ligand binding; i.e. CPA binding to the A1-Gialpha fusion proteins was enhanced, whereas the affinity of DPCPX was hardly affected. Moreover, sodium ions were unable to decrease agonist binding to the majority of the A1-Gialpha fusion proteins, presumably because they exhibit their effect through uncoupling of the R-G complex. From [35S]GTPgammaS binding experiments, we learned that all the A1-Gialpha fusion proteins tested had a higher basal receptor activity than the unfused adenosine A1 receptor, thereby providing improved conditions to observe inverse agonism. Moreover, the maximal CPA-induced stimulation of basal [35S]GTPgammaS binding was increased for the five A1-Gialpha fusion proteins tested, whereas the inhibition induced by 8-cyclopentyltheophylline (CPT) was more pronounced at 351Cys, 351Ile, and 351Val A1-Gialpha fusion proteins. Thus, the maximal receptor (de)activation depended on the amino acid at position 351 of the Gi alpha-subunit. In conclusion, A1-Gialpha fusion proteins, especially with 351Cys and 351Ile, can be used as research tools to investigate inverse agonism, due to their increased readout window in [35S]GTPgammaS binding experiments.


Subject(s)
Adenosine/analogs & derivatives , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Receptor, Adenosine A1/metabolism , Recombinant Fusion Proteins/metabolism , Adenosine/metabolism , Adenosine/pharmacology , Allosteric Site/genetics , Animals , Binding, Competitive/drug effects , COS Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cysteine/genetics , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Mutation , Radioligand Assay , Receptor, Adenosine A1/genetics , Recombinant Fusion Proteins/genetics , Sodium Chloride/pharmacology , Sulfur Radioisotopes , Thiophenes/pharmacology , Transfection , Tritium , Xanthines/metabolism
11.
J Med Chem ; 46(8): 1492-503, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12672250

ABSTRACT

Four subtypes of adenosine receptors are currently known, that is, A(1), A(2A), A(2B), and A(3) receptors. Interestingly, quite substantial species differences exist especially between human and rat A(3) receptors. As a result, ligands such as CCPA, which are very selective for the rat A(1) receptor versus the human A(3) receptor, are substantially less selective when the human A(1) and A(3) receptors are compared. New 2-substituted and 2,N(6)-disubstituted adenosines were synthesized, and their affinities for the human adenosine A(1), A(2A), A(2B), and A(3) receptors were determined. Although large substituents on the C2-position are generally thought to yield adenosine A(2A) receptor selective ligands, the reported series of 2-triazeno-substituted adenosines had a very high affinity for the A(1) receptor. For example, 2-(3-phenylaminocarbonyltriazene-1-yl)adenosine had an affinity of 6.1 +/- 1.3 nM for the human adenosine A(1) receptor. Introduction of a diphenethyl substituent at the N(6)-position of this compound resulted in a high-affinity agonist, 3.1 +/- 0.9 nM, for the human adenosine A(1) receptor with 316- and 45-fold selectivity versus the human A(2A) and human A(3) receptors, respectively. The most selective, high-affinity human adenosine A(1) receptor agonist was the disubstituted compound N(6)-cyclopentyl-2-(3-phenylaminocarbonyltriazene-1-yl)adenosine (TCPA). TCPA had an affinity of 2.8 +/- 0.8 nM for the human adenosine A(1) receptor and was 75-fold and 214-fold selective versus the human A(2A) and human A(3) receptors, respectively. In addition, TCPA was a full agonist and inhibited the forskolin-induced cAMP production of CHO cells stably transfected with the human adenosine A(1) receptor with an IC(50) of 1.5 +/- 0.5 nM.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Purinergic P1 Receptor Agonists , Triazenes/chemical synthesis , Adenosine/chemistry , Adenosine/pharmacology , Animals , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Humans , Radioligand Assay , Structure-Activity Relationship , Triazenes/chemistry , Triazenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...