Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Fundam Clin Pharmacol ; 37(1): 147-157, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35981720

ABSTRACT

Binding kinetics has become a popular topic in pharmacology due to its potential contribution to the selectivity and duration of drug action. Yet, the overall kinetic aspects of complex binding mechanisms are still merely described in terms of elaborate algebraic equations. Interestingly, it has been recommended some 10 years ago to examine such mechanisms in terms of binding fluxes instead of the conventional rate constants. Alike the velocity of product formation in enzymology, those fluxes refer to the velocity by which one target species converts into another one. Novel binding flux-based approaches are utilized to get a better visual insight into the "competition" between two drugs/ligands for a single target as well as between induced fit- and conformational selection pathways for a single ligand within a thermodynamic cycle. The present data were obtained by differential equation-based simulations. Early on, the ligand-binding steps "race" to equilibrium (i.e., when their forward and reverse fluxes are equal) at their individual pace. The overall/global equilibrium is only reached later on. For the competition association assays, this parting might produce a transient "overshoot" of one of the bound target species. A similar overshoot may also show up within a thermodynamic cycle and, at first glance, suggest that the induced fit pathway dominates. Yet, present findings show that under certain circumstances, it could rather be the other way round. Novel binding flux-based approaches offer visually attractive insights into crucial aspects of "complex" binding mechanisms under non-equilibrium conditions.


Subject(s)
Protein Conformation , Ligands , Kinetics , Protein Binding
2.
Pharmaceuticals (Basel) ; 14(12)2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34959665

ABSTRACT

Treatment of neuroendocrine tumours with the radiolabelled somatostatin receptor subtype 2 (SST2) peptide agonist [177Lu]Lu-DOTA-TATE is effective and well-established. Recent studies suggest improved therapeutic efficacy using the SST2 peptide antagonist [177Lu]Lu-OPS201. However, little is known about the cellular mechanisms that lead to the observed differences. In the present in vitro study, we compared kinetic binding, saturation binding, competition binding, cellular uptake and release of [177Lu]Lu-OPS201 versus [177Lu]Lu-DOTA-TATE using HEK cells stably transfected with the human SST2. While [177Lu]Lu-OPS201 and [177Lu]Lu-DOTA-TATE exhibited comparable affinity (KD, 0.15 ± 0.003 and 0.08 ± 0.02 nM, respectively), [177Lu]Lu-OPS201 recognized four times more binding sites than [177Lu]Lu-DOTA-TATE. Competition assays demonstrated that a high concentration of the agonist displaced only 30% of [177Lu]Lu-OPS201 bound to HEK-SST2 cell membranes; an indication that the antagonist binds to additional sites that are not recognized by the agonist. [177Lu]Lu-OPS201 showed faster association and slower dissociation than [177Lu]Lu-DOTA-TATE. Whereas most of [177Lu]Lu-OPS201 remained at the cell surface, [177Lu]Lu-DOTA-TATE was almost completely internalised inside the cell. The present data identified distinct differences between [177Lu]Lu-OPS201 and [177Lu]Lu-DOTA-TATE regarding the recognition of receptor binding sites (higher for [177Lu]Lu-OPS201) and their kinetics (faster association and slower dissociation of [177Lu]Lu-OPS201) that explain, to a great extent, the improved therapeutic efficacy of [177Lu]Lu-OPS201 compared to [177Lu]Lu-DOTA-TATE.

3.
Pharmacol Res Perspect ; 9(5): e00847, 2021 10.
Article in English | MEDLINE | ID: mdl-34459109

ABSTRACT

Induced fit- (IF) and conformational selection (CS) binding mechanisms have long been regarded to be mutually exclusive. Yet, they are now increasingly considered to produce the final ligand-target complex alongside within a thermodynamic cycle. This viewpoint benefited from the introduction of binding fluxes as a tool for analyzing the overall behavior of such cycle. This study aims to provide more vivid and applicable insights into this emerging field. In this respect, combining differential equation- based simulations and hitherto little explored alternative modes of calculation provide concordant information about the intricate workings of such cycle. In line with previous reports, we observe that the relative contribution of IF increases with the ligand concentration at equilibrium. Yet the baseline contribution may vary from one case to another and simulations as well as calculations show that this parameter is essentially regulated by the dissociation rate of both pathways. Closer attention should be paid to how the contributions of IF and CS compare at physiologically relevant drug/ligand concentrations. To this end, a simple equation discloses how changing a limited set of "microscopic" rate constants can extend the concentration range at which CS contributes most effectively. Finally, it could also be beneficial to extend the utilization of flux- based approaches to more physiologically relevant time scales and alternative binding models.


Subject(s)
Computer Simulation , Pharmacokinetics , Protein Binding , Ligands
4.
Trends Pharmacol Sci ; 41(12): 923-932, 2020 12.
Article in English | MEDLINE | ID: mdl-33153779

ABSTRACT

A decade ago, many high-affinity drugs were thought to bind to their target via an induced-fit pathway instead of conformational selection. Yet, both pathways make up part of a thermodynamic cycle, and, owing to binding flux-based approaches, it is now rather considered that they act in parallel and also that their relative contribution to the final ligand-target complex depends on the ligand concentration. Those approaches are of increasing interest, but published data still merely refer to the peculiar situation of equilibrium binding. This article draws attention to the benefit of extending those approaches to address more physiological nonequilibrium binding conditions and in vivo situations. For the presented example, they help to apprehend transient experimental manifestations of a 'conventional' thermodynamic cycle.


Subject(s)
Protein Conformation , Humans , Kinetics , Ligands , Protein Binding , Thermodynamics
5.
Elife ; 82019 01 03.
Article in English | MEDLINE | ID: mdl-30605082

ABSTRACT

PDZ domain scaffold proteins are molecular modules orchestrating cellular signalling in space and time. Here, we investigate assembly of PDZ scaffolds using supported cell membrane sheets, a unique experimental setup enabling direct access to the intracellular face of the cell membrane. Our data demonstrate how multivalent protein-protein and protein-lipid interactions provide critical avidity for the strong binding between the PDZ domain scaffold proteins, PICK1 and PSD-95, and their cognate transmembrane binding partners. The kinetics of the binding were remarkably slow and binding strength two-three orders of magnitude higher than the intrinsic affinity for the isolated PDZ interaction. Interestingly, discrete changes in the intrinsic PICK1 PDZ affinity did not affect overall binding strength but instead revealed dual scaffold modes for PICK1. Our data supported by simulations suggest that intrinsic PDZ domain affinities are finely tuned and encode specific cellular responses, enabling multiplexed cellular functions of PDZ scaffolds.


Subject(s)
Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Disks Large Homolog 4 Protein/metabolism , PDZ Domains , Allosteric Site , Amino Acid Motifs , Animals , Binding Sites , HEK293 Cells , Hippocampus/metabolism , Humans , Kinetics , Ligands , Mutation , Neurons/metabolism , Protein Binding , Protein Domains , Rats , Recombinant Proteins/metabolism , Signal Transduction , Thermodynamics
6.
Medchemcomm ; 9(9): 1426-1438, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30288218

ABSTRACT

Review articles on binding kinetics essentially focus on drugs that dissociate slowly from their target since this is required for the successful treatment of many pathophysiological conditions. Recently, the therapeutic benefit of a high k on (i.e. the second order association rate constant) has also been linked to fast association and to a fast clinical action. Other studies, however, called this assertion into question since additional factors, like the dosing paradigm and the binding mechanism, are important as well. The still ongoing reticence about integrating binding kinetics in lead optimization programs motivated us to critically review the link between the drug's kinetic rate constants and their in vitro and in vivo target occupancy profile, with special focus on k on. The presented simulations tally with a positive link between a drug's effective/observed association rate (which is quite easy to determine in vitro) and the swiftness of its clinical action. On the other hand, the simulations show that the k on-concept should not be confounded with the effective association process since increasing this parameter only enhances the drug's in vitro and in vivo association under certain conditions: the binding mechanism should be suitable, rebinding (and thus the factors within the target's micro-environment that favour this mechanism) should not be too prominent and the dosage should not be kept in par with the drug's affinity. Otherwise, increasing k on could be ineffective or even be counter-productive.

7.
Br J Pharmacol ; 174(23): 4233-4246, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28838028

ABSTRACT

BACKGROUND AND PURPOSE: Optimal drug therapy often requires long-lasting target occupancy While this attribute was usually linked to the drug's pharmacokinetic properties, the dissociation rate is now increasingly recognized to contribute as well. Nearly all the earlier pharmacokinetic-pharmacodynamic (PK-PD) simulations encompassed single-step binding drugs and focused on koff . However, 'micro'-PK mechanisms and more complex binding mechanisms like bivalent- and induced-fit binding may contribute as well. Corresponding binding models are presently explored. EXPERIMENTAL APPROACH: We compared the 24 h in vivo occupancy over time profiles of prototype bivalent- and induced-fit-like binding drugs (A and B) after one or repeated daily dosings, both without and with rebinding. Special attention was focused on the effect of each of the microscopic rate constants on the occupancy profiles and on the metrics to represent those profiles. KEY RESULTS: Although both models can be represented by the same mathematical formulation, drugs A and B display quite different occupancy profiles, even though they have the same potency. These differences can be attributed to the different effects of their microscopic rate constants on their composite koff and also on their susceptibility to experience rebinding. This also affects how the occupancy profiles of bivalent- and induced-fit-like binders progress when repeating the dosings and by changing the dosage. CONCLUSIONS AND IMPLICATIONS: Closer attention should be paid to more complex binding models in PK-PD simulations. This may help pharmacologists and medicinal chemists to improve the translation of in vitro kinetic measurements from preclinical screening programmes into clinical efficiency.


Subject(s)
Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Computer Simulation , Dose-Response Relationship, Drug , Humans , Pharmaceutical Preparations/administration & dosage , Protein Binding , Time Factors
8.
Br J Clin Pharmacol ; 82(3): 673-82, 2016 09.
Article in English | MEDLINE | ID: mdl-27135195

ABSTRACT

The time course of the beneficial pharmacological effect of a drug has long been considered to depend merely on the temporal fluctuation of its free concentration. Only in the last decade has it become widely accepted that target-binding kinetics can also affect in vivo pharmacological activity. Although current reviews still essentially focus on genuine dissociation rates, evidence is accumulating that additional micro-pharmacokinetic (PK) and -pharmacodynamic (PD) mechanisms, in which the cell membrane plays a central role, may also increase the residence time of a drug on its target. The present review provides a compilation of otherwise widely dispersed information on this topic. The cell membrane can intervene in drug binding via the following three major mechanisms: (i) by acting as a sink/repository for the drug; (ii) by modulating the conformation of the drug and even by participating in the binding process; and (iii) by facilitating the approach (and rebinding) of the drug to the target. To highlight these mechanisms, we focus on drugs that are currently used in clinical therapy, such as the antihypertensive angiotensin II type 1 receptor antagonist candesartan, the atypical antipsychotic agent clozapine and the bronchodilator salmeterol. Although the role of cell membranes in PK-PD modelling is gaining increasing interest, many issues remain unresolved. It is likely that novel biophysical and computational approaches will provide improved insights in the near future.


Subject(s)
Cell Membrane/metabolism , Protein Binding , Benzimidazoles/pharmacokinetics , Benzimidazoles/pharmacology , Biphenyl Compounds , Clozapine/pharmacokinetics , Clozapine/pharmacology , Humans , Models, Biological , Salmeterol Xinafoate/pharmacokinetics , Salmeterol Xinafoate/pharmacology , Tetrazoles/pharmacokinetics , Tetrazoles/pharmacology
9.
Assay Drug Dev Technol ; 14(3): 180-93, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27078680

ABSTRACT

Intracerebroventricular injection of angiotensin IV, a ligand of insulin-regulated aminopeptidase (IRAP), has been shown to improve cognitive functions in several animal models. Consequently, IRAP is considered a potential target for treatment of cognitive disorders. To identify nonpeptidic IRAP inhibitors, we adapted an established enzymatic assay based on membrane preparations from Chinese hamster ovary cells and a synthetic peptide-like substrate for high-throughput screening purposes. The 384-well microplate-based absorbance assay was used to screen a diverse set of 10,500 compounds for their inhibitory capacity of IRAP. The assay performance was robust with Z'-values ranging from 0.81 to 0.91, and the screen resulted in 23 compounds that displayed greater than 60% inhibition at a compound concentration of 10 µM. After hit confirmation experiments, purity analysis, and promiscuity investigations, three structurally different compounds were considered particularly interesting as starting points for the development of small-molecule-based IRAP inhibitors. After resynthesis, all three compounds confirmed low µM activity and were shown to be rapidly reversible. Additional characterization included activity in a fluorescence-based orthogonal assay and in the presence of a nonionic detergent and a reducing agent, respectively. Importantly, the characterized compounds also showed inhibition of the human ortholog, prompting our further interest in these novel IRAP inhibitors.


Subject(s)
Cystinyl Aminopeptidase/antagonists & inhibitors , High-Throughput Screening Assays , Protease Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Protease Inhibitors/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship
10.
Br J Pharmacol ; 173(15): 2319-34, 2016 08.
Article in English | MEDLINE | ID: mdl-27129075

ABSTRACT

BACKGROUND AND PURPOSE: Optimal drug therapy often requires continuing high levels of target occupancy. Besides the traditional pharmacokinetic contribution, target binding kinetics is increasingly considered to play an important role as well. While most attention has been focused on the dissociation rate of the complex, recent reports expressed doubt about the unreserved translatability of this pharmacodynamic property into clinical efficacy. 'Micro'-pharmacokinetic mechanisms like drug rebinding and partitioning into the cell membrane may constitute a potential fix. EXPERIMENTAL APPROACH: Simulations were based on solving differential equations. KEY RESULTS: Based on a selected range of association and dissociation rate constants, kon and koff , and rebinding potencies of the drugs as variables, their effects on the temporal in vivo occupancy profile of their targets, after one or multiple repetitive dosings, have here been simulated. CONCLUSIONS AND IMPLICATIONS: Most strikingly, the simulations show that, when rebinding is also taken into account, increasing kon may produce closely the same outcome as decreasing koff when dosing is performed in accordance with the therapeutically most relevant constant [Lmax ]/KD ratio paradigm. Also, under certain conditions, rebinding may produce closely the same outcome as invoking slow diffusion of the drug between the plasma compartment and a target-containing 'effect' compartment. Although the present simulations should only be regarded as a 'proof of principle', these findings may help pharmacologists and medicinal chemists to devise ex vivo and in vitro binding kinetic assays that are more relevant and translatable to in vivo settings.


Subject(s)
Pharmaceutical Preparations/metabolism , Binding Sites , Dose-Response Relationship, Drug , Kinetics
11.
Br J Pharmacol ; 173(8): 1268-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26808227

ABSTRACT

'Induced-fit' binding of drugs to a target may lead to high affinity, selectivity and a long residence time, and this mechanism has been proposed to apply to many drugs with high clinical efficacy. It is a multistep process that initially involves the binding of a drug to its target to form a loose RL complex and a subsequent isomerization/conformational change to yield a tighter binding R'L state. Equations with the same mathematical form may also describe the binding of bivalent antibodies and related synthetic drugs. Based on a selected range of 'microscopic' rate constants and variables such as the ligand concentration and incubation time, we have simulated the experimental manifestations that may go along with induced-fit binding. Overall, they validate different experimental procedures that have been used over the years to identify such binding mechanisms. However, they also reveal that each of these manifestations only becomes perceptible at particular combinations of rate constants. The simulations also show that the durable nature of R'L and the propensity of R'L to be formed repeatedly before the ligand dissociates will increase the residence time. This review may help pharmacologists and medicinal chemists obtain preliminary indications for identifying an induced-fit mechanism.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II Type 1 Receptor Blockers/chemistry , Animals , Binding Sites/drug effects , Humans , Ligands
12.
Curr Top Med Chem ; 15(24): 2504-22, 2015.
Article in English | MEDLINE | ID: mdl-26126905

ABSTRACT

Binding kinetics are the rates of association and dissociation of a drug-protein complex and are important molecular descriptors for the optimization of drug binding to G-protein coupled receptors (GPCRs). There are now many examples of binding kinetics in GPCR drug discovery. In this report, the first principles and examples of binding kinetics in GPCR drug discovery are reviewed. Addressed are the influence of binding kinetics on the translation of binding to the therapeutic window in the context of the equilibrium state of the system and molecular mechanisms of slow binding including induced fit, displacement of water, rebinding and heterovalency.


Subject(s)
Drug Discovery , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites/drug effects , Humans , Kinetics
13.
Expert Opin Drug Discov ; 10(10): 1085-98, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26165720

ABSTRACT

INTRODUCTION: Optimal drug therapy often requires continuing high levels of target occupancy. Besides the traditional pharmacokinetic (PK) contribution thereto, drug-target interactions that comprise successive 'microscopic' steps as well as the intervention of the cell membrane and other 'micro'-anatomical structures nearby may help attaining this objective. AREAS COVERED: This article reviews the 'micro'-pharmacodynamic (PD) and PK mechanisms that may increase a drug's residence time. Special focus is on induced-fit- and bivalent ligand binding models as well as on the ability of the plasma membrane surrounding the target to act as a repository for the drug (e.g., microkinetic model), to actively participate in the binding process (e.g., exosite model) and, along with microanatomical elements like synapses and interstitial spaces, to act on the drug's diffusion properties (reduction in dimensionality and drug-rebinding models). EXPERT OPINION: The PK profile, as well as the target dissociation kinetics of a drug, may fail to account for its long-lasting efficiency in intact tissues and in vivo. This lacuna could potentially be alleviated by incorporating some of the enumerated 'microscopic' mechanisms and, to unveil them, dedicated experiments on sufficiently physiologically relevant biological material like cell monolayers can already be implemented early on in the lead optimization process.


Subject(s)
Models, Biological , Pharmaceutical Preparations/administration & dosage , Pharmacokinetics , Cell Membrane/metabolism , Diffusion , Drug Delivery Systems , Humans , Ligands , Pharmaceutical Preparations/metabolism , Synapses/metabolism , Tissue Distribution
14.
Drug Discov Today Technol ; 17: 28-34, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26724334

ABSTRACT

Radioligand binding assays on intact cells offer distinct advantages to those on membrane suspensions. Major pharmacological properties like drug affinity and binding kinetics are more physiologically relevant. Complex mechanisms can be studied with a wider choice of experimental approaches and so provide insights into induced-fit type binding, receptor internalisation and even into pharmacomicrokinetic phenomena like drug rebinding and partitioning into the membrane. Hence, intact cell binding constitutes a valuable addition to the pharmacologist's toolbox.


Subject(s)
Pharmaceutical Preparations/metabolism , Proteins/metabolism , Radioligand Assay , Animals , Drug Evaluation, Preclinical , Humans , Ligands , Protein Binding
15.
Br J Pharmacol ; 172(9): 2300-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25537684

ABSTRACT

BACKGROUND AND PURPOSE: Non-competitive drugs that confer allosteric modulation of orthosteric ligand binding are of increasing interest as therapeutic agents. Sought-after advantages include a ceiling level to drug effect and greater receptor-subtype selectivity. It is thus important to determine the mode of interaction of newly identified receptor ligands early in the drug discovery process and binding studies with labelled orthosteric ligands constitute a traditional approach for this. According to the general allosteric ternary complex model, allosteric ligands that exhibit negative cooperativity may generate distinctive 'competition' curves: they will not reach baseline levels and their nadir will increase in par with the orthosteric ligand concentration. This behaviour is often considered a key hallmark of allosteric interactions. EXPERIMENTAL APPROACH: The present study is based on differential equation-based simulations. KEY RESULTS: The differential equation-based simulations revealed that the same 'competition binding' pattern was also obtained when a monovalent ligand binds to one of the target sites of a heterobivalent ligand, even if this process is exempt of allosteric interactions. This pattern was not strictly reciprocal when the binding of each of the ligands was recorded. The prominence of this phenomenon may vary from one heterobivalent ligand to another and we suggest that this phenomenon may take place with ligands that have been proposed to bind according to 'two-domain' and 'charnière' models. CONCLUSIONS AND IMPLICATIONS: The present findings indicate a familiar experimental situation where bivalency may give rise to observations that could inadvertently be interpreted as allosteric binding. Yet, both mechanisms could be differentiated based on alternative experiments and structural considerations.


Subject(s)
Models, Theoretical , Receptors, Cell Surface/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Binding Sites , Binding, Competitive , Computer Simulation , Dose-Response Relationship, Drug , Ligands , Protein Binding , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/drug effects , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/drug effects
16.
Br J Pharmacol ; 171(23): 5295-312, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25040887

ABSTRACT

BACKGROUND AND PURPOSE: Many GPCRs can be allosterically modulated by small-molecule ligands. This modulation is best understood in terms of the kinetics of the ligand-receptor interaction. However, many current kinetic assays require at least the (radio)labelling of the orthosteric ligand, which is impractical for studying a range of ligands. Here, we describe the application of a so-called competition association assay at the adenosine A1 receptor for this purpose. EXPERIMENTAL APPROACH: We used a competition association assay to examine the binding kinetics of several unlabelled orthosteric agonists of the A1 receptor in the absence or presence of two allosteric modulators. We also tested three bitopic ligands, in which an orthosteric and an allosteric pharmacophore were covalently linked with different spacer lengths. The relevance of the competition association assay for the binding kinetics of the bitopic ligands was also explored by analysing simulated data. KEY RESULTS: The binding kinetics of an unlabelled orthosteric ligand were affected by the addition of an allosteric modulator and such effects were probe- and concentration-dependent. Covalently linking the orthosteric and allosteric pharmacophores into one bitopic molecule had a substantial effect on the overall on- or off-rate. CONCLUSION AND IMPLICATIONS: The competition association assay is a useful tool for exploring the allosteric modulation of the human adenosine A1 receptor. This assay may have general applicability to study allosteric modulation at other GPCRs as well.


Subject(s)
Receptor, Adenosine A1/metabolism , Allosteric Regulation , Animals , Binding, Competitive , CHO Cells , Computer Simulation , Cricetulus , Humans , Kinetics , Ligands , Models, Molecular , Protein Binding
17.
Br J Pharmacol ; 171(14): 3364-75, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24628038

ABSTRACT

BACKGROUND AND PURPOSE: The human CCR5 receptor is a co-receptor for HIV-1 infection and a target for anti-viral therapy. A greater understanding of the binding kinetics of small molecule allosteric ligand interactions with CCR5 will lead to a better understanding of the binding process and may help discover new molecules that avoid resistance. EXPERIMENTAL APPROACH: Using [(3) H] maraviroc as a radioligand, a number of different binding protocols were employed in conjunction with simulations to determine rate constants, kinetic mechanism and mutant kinetic fingerprints for wild-type and mutant human CCR5 with maraviroc, aplaviroc and vicriviroc. KEY RESULTS: Kinetic characterization of maraviroc binding to the wild-type CCR5 was consistent with a two-step kinetic mechanism that involved an initial receptor-ligand complex (RA), which transitioned to a more stable complex, R'A, with at least a 13-fold increase in affinity. The dissociation rate from R'A, k-2 , was 1.2 × 10(-3) min(-1) . The maraviroc time-dependent transition was influenced by F85L, W86A, Y108A, I198A and Y251A mutations of CCR5. CONCLUSIONS AND IMPLICATIONS: The interaction between maraviroc and CCR5 proceeded according to a multi-step kinetic mechanism, whereby initial mass action binding and later reorganizations of the initial maraviroc-receptor complex lead to a complex with longer residence time. Site-directed mutagenesis identified a kinetic fingerprint of residues that affected the binding kinetics, leading to the conclusion that allosteric ligand binding to CCR5 involved the rearrangement of the binding site in a manner specific to each allosteric ligand.


Subject(s)
Allosteric Regulation/drug effects , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/pharmacology , Receptors, CCR5/metabolism , Triazoles/pharmacology , Binding Sites/drug effects , CCR5 Receptor Antagonists/chemistry , Cyclohexanes/chemistry , Dose-Response Relationship, Drug , Humans , Kinetics , Ligands , Maraviroc , Structure-Activity Relationship , Time Factors , Triazoles/chemistry
18.
Fundam Clin Pharmacol ; 28(5): 530-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24118041

ABSTRACT

Bivalent ligands bear two target-binding pharmacophores. Their simultaneous binding increases their affinity (avidity) and residence time. They become 'bitopic' when the binding sites at the target permit the pharmacophores the exert allosteric modulation of each other's affinity and/or activity. Present simulations reveal that positive cooperativity exacerbates these phenomena, whereas negative cooperativity curtails them, irrespective of whether the association or dissociation rates of the individual pharmacophores are affected. Positive cooperativity delays the attainment of equilibrium binding, yielding 'hemi-equilibrium' conditions and only apparent affinity constants under usual experimental conditions. Monovalent ligands that bind to one of the target sites decrease the bitopic ligand's residence time concentration-wise; their potency depends on their association rate and thereon acting cooperativity rather than on affinity. This stems from the repetitive, very fast reformation of fully bound bitopic ligand-target complexes by rebinding of freshly dissociated pharmacophores. These studies deal with kinetic binding properties (of increasing interest in pharmacology) of bitopic ligands (a promising avenue in medicinal chemistry).


Subject(s)
Ligands , Protein Binding , Allosteric Regulation , Humans , Kinetics , Molecular Dynamics Simulation
19.
Naunyn Schmiedebergs Arch Pharmacol ; 386(11): 949-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23812645

ABSTRACT

Bivalent ligands often display high affinity/avidity for and long residence time at their target. Thereto responsible is the synergy that emanates from the simultaneous binding of their two pharmacophores to their respective target sites. Thermodynamic cycle models permit the most complete description of the binding process, and thereto, corresponding differential equation-based simulations link the "microscopic" rate constants that govern the individual binding steps to the "macroscopic" bivalent ligand's binding properties. Present simulations of heterobivalent ligand binding led to an appreciably simpler description thereof. The thermodynamic cycle model can be split into two pathways/lanes that the bivalent ligand can solicit to reach fully bound state. Since the first binding event prompts the still free pharmacophore to stay into "forced proximity" of its target site, such lanes can be looked into by the equations that also apply to induced fit binding mechanisms. Interestingly, the simplest equations apply when bivalency goes along with a large gain in avidity. The overall bivalent ligand association and dissociation will be swifter than via each lane apart, but it is the lane that allows the fastest bidirectional "transit" between the free and the fully bound target that is chiefly solicited. The bivalent ligand's residence time is governed not only by the stability of the fully bound complex but also by the ability of freshly dissociated pharmacophores to successfully rebind. Hence, the presence of a slow-associating pharmacophore could be counterproductive. Yet, a long residence time is unfortunately also responsible for the slow attainment of binding equilibrium.


Subject(s)
Ligands , Models, Biological , Computer Simulation , Pharmaceutical Preparations/metabolism
20.
Eur J Pharmacol ; 702(1-3): 93-102, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23376157

ABSTRACT

The hexapeptide angiotensin IV (Ang IV) induces diverse biological effects such as memory enhancement and protection against ischemic stroke. Studies on the mechanism of Ang IV however are hampered by its instability and its lack of selectivity. The high-affinity binding site for Ang IV is the insulin-regulated aminopeptidase (IRAP, EC 3.4.11.3), but Ang IV also acts as a weak agonist for the Ang II-receptor (AT1), implying the need for stable and highly selective Ang IV-analogues. Here we present the screening of novel Ang IV-analogues, selected on basis of high affinity for IRAP, high selectivity (compared to aminopeptidase N and the AT1 receptor) and resistance against proteases. The selected compound IVDE77 possesses a number of advantages compared to Ang IV: (i) it has a 40 times higher affinity for IRAP (Ki 1.71 nM), (ii) it does not activate the AT1 receptor, (iii) it is easily radiolabeled with tritium and (iv) it is resistant to proteolysis, even in human plasma. In addition, pre-treatment of intact CHO-K1 cells with IVDE77 led to a virtually complete inhibition of subsequent intracellular accumulation of [(3)H]IVDE77-IRAP complexes. IVDE77 thus represents the first Ang IV-analogue able to abolish IRAP-availability completely at the cell surface in vitro. In summary, IVDE77 is a useful tool for the detection of IRAP under physiological conditions, and may contribute to elucidating the mechanism of Ang IV to ascertain which functions are IRAP-dependent.


Subject(s)
Angiotensin II/analogs & derivatives , Azepines/pharmacology , Cystinyl Aminopeptidase/metabolism , Oligopeptides/pharmacology , Angiotensin II/pharmacology , Animals , Binding, Competitive , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Ligands
SELECTION OF CITATIONS
SEARCH DETAIL
...