Abstract
Background and Purpose
Drug‐target residence time is an important, yet often overlooked, parameter in drug discovery. Multiple studies have proposed an increased residence time to be beneficial for improved drug efficacy and/or longer duration of action. Currently, there are many drugs on the market targeting the gonadotropin‐releasing hormone (GnRH) receptor for the treatment of hormone‐dependent diseases. Surprisingly, the kinetic receptor‐binding parameters of these analogues have not yet been reported. Therefore, this project focused on determining the receptor‐binding kinetics of 12 GnRH peptide agonists, including many marketed drugs.
Experimental Approach
A novel radioligand‐binding competition association assay was developed and optimized for the human GnRH receptor with the use of a radiolabelled peptide agonist, [125I]‐triptorelin. In addition to radioligand‐binding studies, a homogeneous time‐resolved FRET Tag‐lite™ method was developed as an alternative assay for the same purpose.
Key Results
Two novel competition association assays were successfully developed and applied to determine the kinetic receptor‐binding characteristics of 12 high‐affinity GnRH peptide agonists. Results obtained from both methods were highly correlated. Interestingly, the binding kinetics of the peptide agonists were more divergent than their affinities with residence times ranging from 5.6 min (goserelin) to 125 min (deslorelin).
Conclusions and Implications
Our research provides new insights by incorporating kinetic, next to equilibrium, binding parameters in current research and development that can potentially improve future drug discovery targeting the GnRH receptor.
Abbreviations
- CHOhGnRH
CHO cells stably expressing the human GnRH receptor
- GnRH
gonadotropin‐releasing hormone
- k1
the association rate constant of the radioligand
- k2
the dissociation rate constant of the radioligand
- k3
the association rate constant of the unlabelled ligand
- k4
the dissociation rate constant of the unlabelled ligand
- RT
residence time
- TR‐FRET
time‐resolved FRET
Tables of Links
| TARGETS |
|---|
| GnRH receptors |
These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (Alexander et al., 2013).
Introduction
Drug‐target residence time is emerging as an important parameter in the drug discovery process. Multiple studies provide evidence that the binding kinetics of drug‐target interactions rather than the typical equilibrium binding parameters are important for in vivo efficacy (Swinney, 2004; Copeland et al., 2006; Tummino and Copeland, 2008; Zhang and Monsma, 2009). Several marketed drugs in the field of GPCRs have retrospectively been shown to display slow receptor dissociation rates or, in other words, long receptor residence times (Guo et al., 2014). For instance, the histamine H1 receptor antagonist desloratidine was found to have a long residence time, which could explain its high potency and 24 h duration of action observed in clinical studies (Anthes et al., 2002). Another example is the insurmountable antagonist for the angiotensin AT1 receptor, telmisartan. The authors deemed the insurmountability and therefore improved efficacy of telmisartan to be partly due to its very slow dissociation from AT1 receptors (Maillard et al., 2002).
The hypothalamic neuropeptide gonadotropin‐releasing hormone (GnRH) is a central mediator of reproductive functions. This decapeptide binds to a class A GPCR, namely, the GnRH receptor located mainly on pituitary gonadotrophs. Along with the pituitary, GnRH receptors are expressed in reproductive tissues, both normal and malignant, such as those of the prostate and mammary gland (Kakar et al., 1994; von Alten et al., 2006; Angelucci et al., 2009; Aguilar‐Rojas et al., 2012). Upon receptor activation, the gonadotropins luteinizing hormone (LH) and follicle‐stimulating hormone (FSH) are synthesized and secreted from gonadotrophic cells. LH and FSH consecutively induce follicle stimulation and ovulation in women and promote steroidogenesis in both men and women (Stojilkovic et al., 1994).
The pulsatile release of GnRH from the hypothalamus is essential for the maintenance of ovarian function. Sustained exposure of GnRH receptors to GnRH or GnRH analogues leads to activation, commonly named ‘flare’, followed by desensitization of GnRH receptor‐mediated gonadotropin secretion. Accordingly, blockade by antagonists and desensitization of GnRH receptor‐mediated gonadotropin secretion both ultimately reduce circulating levels of gonadotropins and gonadal steroids (Belchetz et al., 1978; Lahlou et al., 1987). This so‐called chemical castration underlies the therapeutic use of GnRH analogues to treat sex hormone‐dependent diseases (Depalo et al., 2012; Labrie, 2014; Leone Roberti Maggiore et al., 2014).
Consequently, considerable efforts have been put towards the development of agonists and antagonists targeting the GnRH receptor (Karten and Rivier, 1986; Sealfon et al., 1997; Millar et al., 2004; Heitman and IJzerman, 2008; Millar and Newton, 2013). Only a few studies have examined the receptor‐binding kinetics of GnRH ligands. A paper of Heise et al. (2007) described a scintillation proximity assay to qualitatively distinguish between fast and slowly dissociating antagonists for the GnRH receptor. The authors demonstrated that slow dissociation rates were responsible for large discrepancies between a ligand's Ki value determined at 30 min versus 10 h, and they suggested using the Ki ratio as a screening method to select slowly dissociating compounds. Two other studies focused on a quantitative determination of receptor‐binding kinetics of small molecule GnRH antagonists. Here, a direct correlation between the insurmountability and slow dissociation rates of these antagonists was shown (Sullivan et al., 2006; Kohout et al., 2007).
Currently, multiple peptide GnRH analogues have been approved for the treatment of advanced prostatic cancer, endometriosis, in vitro fertilization and more (Depalo et al., 2012; Romero et al., 2012; Aydiner et al., 2013; Goericke‐Pesch et al., 2013; Lewis et al., 2013; Labrie, 2014; Leone Roberti Maggiore et al., 2014). Remarkably, the receptor‐binding kinetics of peptide GnRH receptor ligands have never been reported. Therefore, we developed a novel radioligand‐binding competition association assay that allowed us to determine the kinetic binding parameters and focused on 12 GnRH peptide agonists, including many marketed drugs (Table 1). In addition, we compared these kinetic parameters with those from a newly developed homogeneous time‐resolved fluorescent assay. Both assays may improve future drug discovery targeting the GnRH receptors by incorporating kinetic receptor‐binding parameters into current research and development trajectories.
Table 1.
Amino acid sequences of the 12 GnRH peptide agonists examined
| 1 | 2 | 3 | 4 | 5 | 6 | 7 | 8 | 9 | 10 | |
|---|---|---|---|---|---|---|---|---|---|---|
| GnRH | pGlu* | His | Trp | Ser | Tyr | Gly | Leu | Arg | Pro | Gly–NH2 |
| Triptorelin | pGlu* | His | Trp | Ser | Tyr | d‐Trp | Leu | Arg | Pro | Gly–NH2 |
| [d‐Ala6]‐GnRH | pGlu* | His | Trp | Ser | Tyr | d‐Ala | Leu | Arg | Pro | Gly–NH2 |
| [d‐Lys6]‐GnRH | pGlu* | His | Trp | Ser | Tyr | d‐Lys | Leu | Arg | Pro | Gly‐NH2 |
| Fertirelin | pGlu* | His | Trp | Ser | Tyr | Gly | Leu | Arg | Pro | NHEt * |
| Alarelin | pGlu* | His | Trp | Ser | Tyr | d‐Ala | Leu | Arg | Pro | NHEt * |
| Deslorelin | pGlu* | His | Trp | Ser | Tyr | d‐Trp | Leu | Arg | Pro | NHEt * |
| Leuprorelin | pGlu* | His | Trp | Ser | Tyr | d‐Leu | Leu | Arg | Pro | NHEt * |
| Nafarelin | pGlu* | His | Trp | Ser | Tyr | D2Nal | Leu | Arg | Pro | Gly–NH2 |
| Buserelin | pGlu* | His | Trp | Ser | Tyr | Ser‐tBu * | Leu | Arg | Pro | NHEt * |
| Goserelin | pGlu* | His | Trp | Ser | Tyr | Ser‐tBu * | Leu | Arg | Pro | azaGly–NH2 * |
| Histerelin | pGlu* | His | Trp | Ser | Tyr | His(Bzl) * | Leu | Arg | Pro | NHEt * |
The differences between the peptides are expressed in bold
pGlu, pyroglutamic acid; D2Nal, (2‐naphthyl)‐d‐alanine; Ser‐tBu, serine‐tert‐butyl; His(Bzl), N‐benzyl‐l‐histidine; azaGly–NH2, aza‐glycine amine; NHEt, ethylamide
Methods
Cell culture
For radioligand‐binding assays, CHOhGnRH cells were grown in Ham's F12/DMEM (1:1) medium supplemented with 10% normal bovine serum, 2 mM glutamine, penicillin (100 IU·mL−1), streptomycin (100 µg·mL−1) and G418 (200 µg·mL−1) at 37°C in 5% CO2.
For time‐resolved FRET (TR‐FRET) experiments, 1 mL (7 * 106 cells·mL−1) of Tag‐lite GnRH cells were thawed, washed with 15 mL ice‐cold Tag‐lite buffer (TLB), resuspended in 5 mL TLB and immediately used.
Membrane preparation
CHOhGnRH cells were scraped from the plates in 5 mL PBS, collected and centrifuged at 700× gfor 5 min. Derived pellets were pooled and resuspended in 50 mM Tris HCl buffer pH 7.4 at 25°C supplemented with 2 mM MgCl2 and subsequently homogenized with an UltraThurrax (Heidolph Instruments, Schwabach, Germany). The cytosolic fraction and membranes were separated by centrifugation at 100 000× g in an Optima LE‐80 K ultracentrifuge (Beckman Coulter, Fullterton, CA, USA) for 20 min at 4°C. The pellet was resuspended, and centrifugation was repeated. The obtained pellet was resuspended; membranes were aliquoted and stored at −80°C. Membrane protein concentrations were determined using the bicinchoninic acid method with BSA as a standard (Smith et al., 1985).
Radioligand equilibrium assays
Displacement experiments were performed as previously reported (Heitman et al., 2008). In short, membrane aliquots containing 15–20 µg protein were incubated in a total volume of 100 μL assay buffer (25 mM Tris HCl, pH 7.4 at 25°C, supplemented with 2 mM MgCl2, 0.1% (w v‐1) BSA) at 25°C for 2 h. Ten concentrations of competing ligand were used in the presence of 30.000 c.p.m. (~0.1 nM) [125I]‐triptorelin. At this concentration, total radioligand binding did not exceed 10% of that added to prevent ligand depletion. Non‐specific binding was determined in the presence of an excess amount of GnRH (1 μM). The reaction was terminated by the addition of 1 mL ice‐cold wash buffer (25 mM Tris HCl, pH 7.4 at 25°C, supplemented with 2 mM MgCl2 and 0.05% (w v‐1) BSA). Separation of bound from free radioligand was performed by rapid filtration through Whatman GF/B filters saturated with 0.25% polyethylene imine using a Brandel harvester. Filters were subsequently washed three times with 2 mL ice‐cold wash buffer. Filter bound radioactivity was determined using a γ‐counter (Wizard 1470, PerkinElmer).
Radioligand kinetic association and dissociation assays
Association experiments were carried out by incubating membrane aliquots containing 15–20 µg protein in a total volume of 100 μL assay buffer at 25°C with 30.000 c.p.m. (~0.1 nM) [125I]‐triptorelin. The amount of radioligand bound to the receptor was determined at different time intervals for a total incubation time of 120 min.
Dissociation experiments were performed by pre‐incubating membrane aliquots containing 15–20 µg protein in a total volume of 100 μL assay buffer at 25°C for 45 min with 30.000 c.p.m. (~0.1 nM) [125I]‐triptorelin. After pre‐incubation, dissociation was initiated by addition of an excess amount of GnRH (1 μM) in a total volume of 2.5 μL. The amount of radioligand still bound to the receptor was measured at various time intervals for a total incubation time of 120 min. The reaction was stopped, and samples were harvested as described under Radioligand Equilibrium Assays.
Radioligand kinetic competition association assays
The binding kinetics of unlabelled ligands were quantified using the competition association assay based on the method by Motulsky and Mahan (1984). During optimization, three different concentrations of unlabelled triptorelin were tested; 0.3‐fold, 1‐fold and 3‐fold its Ki value. The kinetic parameters of all other unlabelled ligands were determined at a concentration of onefold their Ki, unless stated otherwise. The competition association assay was initiated by adding membrane aliquots containing 15–20 µg protein in a total volume of 100 μL assay buffer at 25°C with 50.000 c.p.m. (~0.15 nM) [125I]‐triptorelin in the absence or presence of competing ligand. Of note, total radioligand binding did not exceed 10% of that added at this concentration to prevent ligand depletion. The amount of radioligand bound to the receptor was determined at different time intervals for a total incubation time of 120 min. The reaction was stopped, and samples were harvested as described under Radioligand Equilibrium Assays.
TR‐FRET probe equilibrium assays
Unless otherwise specified, TR‐FRET measurements were carried out using the conditions described in Schiele et al. (2014). To determine the equilibrium affinity of the fluorescent probe, 5 μL Tag‐lite GnRH cells (1400 cells·μL−1) were incubated for 1 h, to ensure signal stability, with increasing probe concentrations (ranging from 0 to 100 nM: Supporting Information Fig. S1) in a final volume of 10 μL. In parallel, a non‐specific binding control was carried out in the presence of an excess amount of buserelin (100 μM). Binding signals were measured in a PHERAstar FS plate reader BMG Labtech (Offenburg, Germany) by exciting the Tb donor with five laser flashes at a wavelength of 337 nm and recording acceptor and donor emission fluorescence channels (A and B channels), at wavelengths of 520 and 490 nm respectively.
TR‐FRET equilibrium probe competition assays
‘Ready‐to‐use’ assay plates containing serial dilutions of the test agonists were prepared as described in Schiele et al. (2014). Subsequently, 5 μL Tag‐lite GnRH cells (1400 cells·μL−1) and 50 nM probe were added to the competitors and incubated at room temperature for 1 h, to ensure signal stability, in a final volume of 10 μL. Non‐specific binding (‘low signal’) controls contained an excess amount of buserelin (100 μM), whereas in ‘high signal’ controls, the test compounds were replaced by DMSO. Binding signals were recorded as described under TR‐FRET Probe Equilibrium Assays.
TR‐FRET kinetic probe association and dissociation assays
Measurements were carried out in quadruplicate and in a final volume of 15 μL per well. First, a 5‐point, twofold serial dilution of fluorescent probe (Supporting Information Fig. 2) was pre‐dispensed on black 384‐well low volume plates (Greiner Bio‐one (Frickenhausen, Germany)), and the PHERAstar FS injection system's syringes (previously washed with NaOH/H2O) were primed either with 1500 μL solution of Tag‐lite GnRH cells (1000 cells·μL−1) or with 200 μM buserelin. Then, 4 μL of cells were quickly added to the probe with the first syringe, and the association traces were recorded as described under TR‐FRET Probe Equilibrium Assays, with kinetic intervals of 26 s. After 30 min, fluorescent probe dissociation was initiated by addition of 5 μL of an excess of unlabelled buserelin (final concentration 67 μM) with the second syringe, and the traces were recorded with kinetic intervals of 300 s in the same fashion. Alternatively, a 1‐point measurement was performed with 5 μL of probe (final concentration 25 nM) and 5 μL of Tag‐lite GnRH cells (1400 cells·μL−1), and association was recorded with kinetic intervals of 120 s. After 24 min, dissociation was initiated and recorded with the same kinetic interval.
TR‐FRET kinetic probe titration assays
First, 5 μL of increasing concentrations of fluorescent probe were dispensed into 384‐well plates; the injection system of the PHERAstar FS plate reader was primed with Tag‐lite GnRH cells as described under TR‐FRET Probe Equilibrium Assays. Then, 5 μL of cell solution was added with the syringe, and the TR‐FRET signals corresponding to probe association were recorded as described under TR‐FRET Probe Equilibrium Assays.
TR‐FRET kinetic probe competition assays
The basic principle of this assay is explained in Schiele et al. (2014). Before each experiment, 6 μL of fluorescent probe (final concentration 15 nM) was dispensed to the ‘assay‐ready’ plates containing 100 nL of compound dilutions using a Multidrop Combi Thermo Fischer Scientific (Dreieich, Germany), and the injection system of the PHERAstar FS plate reader was washed with NaOH/H20 and primed with Tag‐lite GnRH cells. Finally, the assay plates were introduced into the instrument, 4 μL of cells (1000 cells·μL−1) were rapidly dispensed with the syringe to each well and the TR‐FRET signals corresponding to the competitive binding of probe and test compounds were recorded as described under TR‐FRET Probe Equilibrium Assays with kinetic intervals of 78 s.
Data analysis
All experimental data were analysed using the nonlinear regression curve‐fitting programme GraphPad Prism v. 5.00 (GraphPad Software Inc., San Diego, CA, USA). Further details on the handling of TR‐FRET data are available in Schiele et al. (2014).
For radioligand‐binding assays, the previously reported KD value of 0.35 nM for [125I]‐triptorelin (Heitman et al., 2008) was used to convert IC50 values obtained from competition curve analysis into Ki values with the help of the Cheng–Prusoff equation (Cheng and Prusoff, 1973):
Likewise, a KD value of 0.8 nM obtained for the ‘red’‐labelled buserelin by fitting the data from the TR‐FRET probe equilibrium binding assay (Supporting Information Fig. S1) to the model ‘One site – Specific binding’ was used to convert IC50 values from TR‐FRET experiments to Ki values.
The observed association rates (k obs) derived from both assays were obtained by fitting association data using one‐phase exponential association. The dissociation rates were obtained by fitting dissociation data to a one‐phase exponential decay model. The k obs values were converted into association rate (k on) values using the following equation:
The association and dissociation rates were used to calculate the kinetic KD using the following equation:
To further validate probe affinity and kinetic rate constants, association data from kinetic probe titration experiments were fitted to the ‘Association kinetics – two or more concentrations of hot’ model (Supporting Information Fig. S2). The k on obtained from these experiments and KD from equilibrium binding were used to calculate k off as described previously.
Association and dissociation rates for unlabelled ligands were calculated by fitting the data of the competition association assay using kinetics of competitive binding (Motulsky and Mahan, 1984):
where k 1 is the k on of the radioligand (M−1·min−1), k 2 is the k off of the radioligand (min−1), L is the radioligand concentration (nM), I is the concentration of the unlabelled competitor (nM), X is the time (min) and Y is the specific binding of the radioligand (DPM). During a competition association, these parameters are set, obtaining k 1 from the control curve without competitor and k 2 from previously performed dissociation assays described under Radioligand Association and Dissociation Assays. With that, the k 3, k 4 and B max can be calculated, where k 3 represents the k on (M−1·min−1) of the unlabelled ligand, k 4 stands for the k off of the unlabelled ligand and B max equals the total binding (DPM). All competition association data were globally fitted.
In case of kinetic probe competition assays (kPCA), the kinetics of the competitive binding model was enhanced with a mathematical term describing a mono‐exponential decay that accounts for signal drift (Schiele et al., 2014). As stated for the radioligand‐binding assays, the kinetic rate constants of the fluorescent probe (k 1 and k 2) were determined in separate experiments and set constant in kPCA data analysis.
The RT was calculated as in the following equation:
All values obtained from radioligand‐binding assays are means of at least three independent experiments performed in duplicate, unless stated otherwise. Values obtained from TR‐FRET assays are means of two independent experiments performed in quadruplicate, unless stated otherwise.
Reagents and peptides
Deslorelin and fertirelin (Table 1) were obtained from Genway Biotech Inc. (San Diego, CA, USA) and American Peptide Company (Sunnyvale, CA, USA) respectively. All other peptide analogues (Table 1) and BSA were purchased from Sigma‐Aldrich Chemie B.V. (Zwijndrecht, The Netherlands). Bicinchoninic acid protein assay reagent was obtained from Pierce Chemical Company (Rockford, IL, USA). [125I]‐triptorelin (specific activity 2200 Ci·mmol−1) was purchased from PerkinElmer (Groningen, The Netherlands). CHO cells stably expressing the human GnRH receptor (from now on CHOhGnRH cells) were kindly provided by MSD (Oss, The Netherlands). Tag‐lite™ HEK293 cells containing a stably overexpressed human GnRH receptor labelled with Tb (from now on Tag‐lite GnRH cells) were obtained as frozen stocks from Cisbio (Codolet, France). A buserelin‐derived tracer, labelled at the sixth position with a red emitting fluorophore, and (TLB) were also purchased from Cisbio. All other chemicals and cell culture materials were obtained from standard commercial sources.
Results
Determination of the association and dissociation rate constants of [125I]‐triptorelin
The binding properties of [125I]‐triptorelin to CHOhGnRH membranes were quantified with traditional kinetic radioligand‐binding assays. Association and dissociation experiments provided k on and k off values of 0.4 ± 0.1 nM−1·min−1 and 0.05 ± 0.0004 min−1 respectively (Figure 1A and Table 2). From these data, the equilibrium dissociation constant (kinetic KD) was calculated, which had a value of 0.2 nM.
Figure 1.

Association and dissociation kinetics of [125I]‐triptorelin (A,B) or fluorescent probe (C) at the hGnRH receptor. Representative graphs from one experiment performed in duplicate (see Tables 2 and 3 for kinetic parameters).
Table 2.
Comparison of the affinity, dissociation constants and kinetic parameters of reference agonist triptorelin obtained with different radioligand‐binding assays
| Assay | pKD b and (KD (nM)) | pKi and (Ki (nM)) | k on (nM−1·min−1) | k off (min−1) |
|---|---|---|---|---|
| Displacement | NA | 9.6 ± 0.09 (0.27) | NA | NA |
| Association and dissociation | 9.9 ± 0.11 (0.13) | NA | 0.40 ± 0.12 | 0.050 ± 0.0004 |
| Competition associationa | 9.7 ± 0.12 (0.22) | NA | 0.12 ± 0.014 | 0.026 ± 0.008 |
Values are means ± SEM of three separate experiments performed in duplicate
NA, not applicable
The binding kinetics of unlabelled triptorelin were determined by addition of 0.3‐fold, 1‐fold and 3‐fold its Ki value
KD = k off/k on
Determination of the association and dissociation rate constants of the fluorescently labelled buserelin derivative probe
A fluorescently labelled buserelin derivative was used as a probe in all TR‐FRET assays. The kinetic parameters of the fluorescent tracer were determined by performing association and dissociation experiments. Experiments yielded a k on and k off of 0.008 ± 0.001 nM−1·min−1 and 0.01 ± 0.001 min−1 respectively (Figure 1B, Table 3 and Supporting Information Fig. 2). The kinetic KD value calculated from these experiments was 1.2 nM.
Table 3.
Comparison of the affinity, dissociation constants and kinetic parameters of the fluorescent buserelin probe obtained with different TR‐FRET assays
| Assay | pKD and (KD (nM)) | pKi and (Ki (nM)) | k on (nM−1·min−1) | k off (min−1)b |
|---|---|---|---|---|
| Equilibrium association | 9.1 ± 0.8 (0.8) | NA | NA | NA |
| Association and dissociationa | 8.9 ± 0.9 (1.2) | NA | 0.008 ± 0.001 | 0.010 ± 0.001 |
| Multiple association and dissociation | 8.7 ± 0.06 (2.1) | NA | 0.008 ± 0.001 | 0.016 ± 0.002 |
Values are means ± SEM of three separate experiments
NA, not applicable
The dissociation kinetics of fluorescently labelled buserelin derivative were determined by addition of 10 μM buserelin
k off = KD(equilibrium)/k on
Determination of the binding affinity of hGnRH receptor agonists with [125I]‐triptorelin
Equilibrium radioligand‐binding assays were performed to assess the ability of 12 GnRH analogues to displace [125I]‐triptorelin from CHOhGnRH cell membranes. All ligands were able to fully displace [125I]‐triptorelin in a concentration‐dependent manner (Figure 2A and Table 4). All peptides had a Hill coefficient close to unity in the [125I]‐triptorelin displacement assay (data not shown), which indicated a competitive mode of inhibition with regard to the radioligand. Of all tested ligands, nafarelin had the highest affinity for the hGnRH receptor with a Ki value of 0.06 nM, and GnRH had the lowest affinity of 13 nM. All other ligands had affinities in the low to sub‐nanomolar range.
Figure 2.

Displacement of [125I]‐triptorelin (A) or fluorescent probe (B) from the hGnRH receptor by the 12 peptide agonists. Representative graphs from one experiment performed in duplicate (see Table 4 for affinity values).
Table 4.
Binding parameters of GnRH peptide agonists derived from radioligand binding and TR‐FRET experiments
| Radioligand binding | TR‐FRET | |||
|---|---|---|---|---|
| Agonist | pKi and (Ki (nM)) | pKD and (KD (nM)) | pKi and (Ki (nM)) | pKD and (KD (nM)) |
| GnRH | 7.9 ± 0.05 (13) | 8.5 ± 0.08 (2.9) | 8.4 ± 0.6 (4.0) | 7.7 ± 0.03 (22) |
| Triptorelin | 9.6 ± 0.09 (0.3) | 9.7 ± 0.1 (0.2) | 9.5 ± 0.2 (0.4) | 9.5 ± 0.03 (0.4) |
| [d‐Ala6]‐GnRH | 9.0 ± 0.05 (0.8) | 9.1 ± 0.09 (0.8) | 8.6 ± 0.4 (2.3) | 8.9 ± 0.02 (1.3) |
| [d‐Lys6]‐GnRH | 8.3 ± 0.1 (5.2) | 8.4 ± 0.2 (3.7)# | 7.8 ± 0.3 (16) | 7.8 ± 0.01 (15) |
| Fertirelin | 9.2 ± 0.05 (0.7) | 9.1 ± 0.08 (0.8)# | 9.0 ± 0.3 (1.0) | 9.0 ± 0.02 (0.9) |
| Alarelin | 9.4 ± 0.1 (0.5) | 9.8 ± 0.1 (0.2) | 9.0 ± 0.3 (0.9) | 9.4 ± 0.03 (0.4) |
| Deslorelin | 10 ± 0.1 (0.1) | 9.9 ± 0.1 (0.1)# | 8.6 ± 0.6 (0.8) | 9.4 ± 0.04 (0.4) |
| Leuprorelin | 9.5 ± 0.09 (0.3) | 9.8 ± 0.1 (0.2) | 9.7 ± 0.3 (0.2) | 8.9 ± 0.03 (1.2) |
| Nafarelin | 10 ± 0.06 (0.06) | 10.6 ± 0.1 (0.03) | 9.8 ± 0.3 (0.2) | 9.7 ± 0.06 (0.2) |
| Buserelin | 9.9 ± 0.05 (0.1) | 10.4 ± 0.2 (0.04) | 9.4 ± 0.2 (0.4) | 9.5 ± 0.04 (0.3) |
| Goserelin | 8.8 ± 0.06 (1.6) | 9.0 ± 0.08 (1.1) | 9.1 ± 0.3 (0.9) | 8.6 ± 0.02 (2.7) |
| Histerelin | 9.8 ± 0.2 (0.2) | 10 ± 0.08 (0.04) | 8.7 ± 0.5 (1.9) | 9.0 ± 0.04 (1.0) |
Values are means ± SEM of at least three separate experiments performed in duplicate
Values are means ± SEM of two separate experiments performed in duplicate
KD = k off/k on
Determination of the binding affinity of hGnRH receptor agonists with TR‐FRET
The binding affinity of 12 agonists was also determined using a fluorescently labelled buserelin derivative as a tracer and Tag‐lite GnRH cells in a TR‐FRET assay. In accordance with the radioligand‐binding results, all agonists were able to fully displace the fluorescent tracer from the hGnRH receptor in a concentration‐dependent manner (Figure 2B and Table 4). The data were in fair agreement with the affinities determined in the radioligand displacement assay despite the inherent differences between the two assays (r 2 = 0.5 and P < 0.05) (Figure 4C).
Figure 4.

Correlation between affinities (pKi) and dissociation constants (pKD) derived from (A) radioligand binding (r 2 = 0.9, P < 0.0001) and (B) TR‐FRET experiments (r 2 = 0.5, P < 0.05). (C) Correlation between affinities (pKi) derived from radioligand binding and homogeneous time‐resolved fluorescent experiments (r 2 = 0.5, P < 0.05). (D) Correlation between dissociation constants (pKD) derived from radioligand binding and TR‐FRET experiments (r 2 = 0.8, P < 0.001). In all cases, pKi values were obtained from equilibrium displacement studies, and pKD values were determined with competition association experiments.
Validation and optimization of the competition association assay with [125I]‐triptorelin
With the k on and k off values of [125I]‐triptorelin obtained from traditional association and dissociation experiments, the k on (k 3) and k off (k 4) values of unlabelled triptorelin could be determined by fitting the kinetic parameters into the model of ‘kinetics of competitive binding’ as described under Methods. Three different concentrations of unlabelled triptorelin were tested and presented a shared k on (k 3) and k off (k 4) value of 0.1 ± 0.01 nM−1·min−1 and 0.03 ± 0.008 min−1 respectively (Figure 3A). These values were in good agreement with the association and dissociation rates obtained with traditional kinetic experiments (Table 2). Additionally, a comparison of the affinity (0.3 nM) and dissociation constants (0.1 and 0.2 nM), acquired from equilibrium and kinetic experiments, respectively, further confirmed the competition association assay as a valid tool to determine the binding kinetics of unlabelled ligands at the hGnRH receptor (Table 2).
Figure 3.

Competition association experiment with [125I]‐triptorelin in the absence or presence of 0.3, 1 or 3 * Ki value of unlabelled triptorelin (A) or 1 * Ki value of buserelin, leuprorelin or GnRH (B). Representative graphs from one experiment performed in duplicate (See Table 2 for kinetic parameters).
To improve the throughput of this assay, one concentration of competitor was selected that yielded an assay window discernable from both the baseline and control curve (i.e. specific binding approximately 40–60%). In this case, a concentration of competitor equal to onefold its Ki value presented the best assay window. Analysis of this single concentration showed similar kinetic rates for triptorelin in comparison with the three‐concentration method, which were statistically indifferent (data not shown; P > 0.05). Thus, this one‐concentration method was used for subsequent determination of the binding kinetics of other unlabelled hGnRH receptor peptide agonists.
Determination of the receptor‐binding kinetics of unlabelled hGnRH receptor agonists with [125I]‐triptorelin
By use of the one‐concentration competition association assay, the binding kinetics of 11 other unlabelled hGnRH receptor agonists were quantified (Figure 3B and Table 5). Juxtaposing affinities (Ki values) and dissociation constants (KD values) acquired from equilibrium and kinetic experiments resulted in a high correlation (r 2 = 0.9, P < 0.0001). Firstly, this further confirmed that the competition association assay was a valid tool to determine the binding kinetics of unlabelled ligands for the hGnRH receptor (Figure 4A) and, secondly, proved that equilibrium was reached for all agonists in the displacement experiments. The dissociation rates ranged from 0.2 ± 0.03 min−1 for goserelin to 0.01 ± 0.003 min−1 for buserelin, a variance of roughly 20‐fold. Interestingly, three distinctive association patterns were obtained from the competition association assays (Figure 3B). Firstly, an ‘overshoot’ in [125I]‐triptorelin association was observed for slowly dissociating compounds, such as buserelin. Secondly, we noticed a shallow increase in [125I]‐triptorelin association for rapidly dissociating compounds, such as GnRH, and lastly, no difference was observed in the shape of the [125I]‐triptorelin association curve for equally fast‐dissociating compounds, such as leuprorelin. The observed differences in dissociation kinetics were all in comparison with those of the radioligand [125I]‐triptorelin (Figure 3B). Association rates ranged from 0.8 ± 0.2 nM−1·min−1 for nafarelin to 0.02 ± 0.004 nM−1·min−1 for fertirelin, a span of approximately 35‐fold.
Table 5.
Kinetic receptor‐binding parameters of GnRH peptide agonists derived from radioligand‐binding competition association assays and kPCA TR‐FRET experiments
| Radioligand binding | TR‐FRET | |||||
|---|---|---|---|---|---|---|
| Agonist | k on (nM−1·min−1)a | k off (min−1)a | RT (min)c | k on (nM−1·min−1)b | k off (min−1)b | RT (min)c |
| GnRH | 0.06 ± 0.01 | 0.2 ± 0.02 | 6.3 ± 0.6 | 0.02 ± 0.01 | 0.44 ± 0.3 | 2.3 ± 1.6 |
| Triptorelin | 0.1 ± 0.01 | 0.03 ± 0.008 | 39 ± 12 | 0.1 ± 0.02 | 0.05 ± 0.008 | 21 ± 3.7 |
| [d‐Ala6]‐GnRH | 0.08 ± 0.01 | 0.07 ± 0.01 | 15 ± 3.1 | 0.05 ± 0.007 | 0.07 ± 0.01 | 14 ± 2.3 |
| [d‐Lys6]‐GnRH | 0.04 ± 0.02# | 0.1 ± 0.04# | 7.7 ± 2.3# | 0.02 ± 0.01 | 0.25 ± 0.17 | 4 ± 2.6 |
| Fertirelin | 0.02 ± 0.004# | 0.02 ± 0.001# | 56 ± 3.1# | 0.07 ± 0.009 | 0.06 ± 0.01 | 15 ± 2.4 |
| Alarelin | 0.09 ± 0.02 | 0.01 ± 0.002 | 77 ± 12 | 0.09 ± 0.009 | 0.03 ± 0.005 | 31 ± 4.8 |
| Deslorelin | 0.07 ± 0.01# | 0.01 ± 0.002# | 100 ± 20# | 0.05 ± 0.005 | 0.02 ±0.004 | 44 ± 6.9 |
| Leuprorelin | 0.2 ± 0.04 | 0.03± 0.005 | 36 ± 6.4 | 0.03 ± 0.004 | 0.04 ± 0.006 | 26 ± 4.4 |
| Nafarelin | 0.8 ± 0.2 | 0.02 ± 0.003 | 50 ± 7.5 | 0.1 ± 0.02 | 0.03 ± 0.006 | 39 ± 9.8 |
| Buserelin | 0.2 ± 0.06 | 0.009 ± 0.003 | 111 ± 37 | 0.05 ± 0.004 | 0.02 ± 0.003 | 61 ± 10 |
| Goserelin | 0.2 ± 0.002 | 0.2 ± 0.03 | 5.6 ± 0.8 | 0.03 ± 0.005 | 0.08 ± 0.01 | 13 ± 2.4 |
| Histerelin | 0.3 ± 0.04 | 0.01 ± 0.002 | 83 ± 14 | 0.02 ± 0.002 | 0.02 ± 0.004 | 50 ± 8.8 |
Values are means ± SEM of at least three separate experiments performed in duplicate
Values are means ± SEM of two separate experiments performed in duplicate
k on and k off of unlabelled GnRH agonists were determined at onefold Ki concentrations
k on and k off of unlabelled GnRH agonists were determined at 0.5, 5, 50 and 500 nM
RT = 1/k off
Determination of the receptor‐binding kinetics of unlabelled hGnRH receptor agonists with a fluorescently labelled buserelin derivative
The kinetic parameters of the 12 GnRH agonists were also determined with TR‐FRET experiments (Figure 5, Table 5 and Supporting Information Fig. 3). Association rates ranged from 0.1 ± 0.02 nM−1·min−1 for triptorelin to 0.02 ± 0.002 nM−1·min−1 for histerelin. Buserelin was again one of the slowest dissociating agonists with a dissociation rate of 0.02 ± 0.003 min−1, while GnRH had the fastest dissociation rate of 0.4 ± 0.03 min−1. The dissociation constants (KD) calculated from k on and k off values were consistent with the affinities determined in homogeneous time‐resolved fluorescent displacement assays (Figure 4B) as well as with the KD values obtained from the radioligand‐binding studies (Figure 4D). Dissociation rate constants (k off) were in good agreement with the data obtained from radioligand‐binding experiments (r 2 = 0.7, P < 0.0005) (Figure 6A), while the association rates (k on) presented no correlation (r 2 = 0.03, P = 0.6) (Figure 6B).
Figure 5.

Competition association experiment with fluorescent probe in the absence or presence of increasing concentrations of buserelin (A) or one‐concentration of unlabelled agonist that showed around 50% displacement (B). Representative graphs from one experiment performed in quadruplicate.
Figure 6.

Correlation between the kinetic parameters obtained from radioligand‐binding assays and kPCA TR‐FRET experiments. (A) Dissociation rate (k off) (r 2 = 0.7, P < 0.0005); (B) association rate (k on) (r 2 = 0.03, P = 0.6).
Discussion and conclusions
Over the years, several studies have indicated that long duration of action is an important feature contributing to improved efficacy of drugs designed to treat chronic illness. Moreover, increased target residence time offers the potential for a once‐daily dosage form that increases patient compliance, which is crucial for the management of diseases (Smith et al., 1996; Tashkin, 2005; Copeland et al., 2006; Dowling and Charlton, 2006; Vauquelin and Van Liefde, 2006; Swinney, 2009; Zhang and Monsma, 2009; Guo et al., 2014).
The GnRH receptor is the target of multiple marketed peptide agonists, classified as functional antagonists, used to treat hormone‐dependent diseases. Available patient information for the most commonly prescribed GnRH analogues suggests that the pharmacokinetic/pharmacodynamics profiles are very similar. Hence, knowledge of the in vitro binding kinetics could give extra insights into these well‐known drugs. However, the potential impact of variable‐binding kinetics of these GnRH peptide derivatives on clinical efficacy has not been investigated. Aside from agonists, a few studies have detailed the effect of slow dissociation kinetics of antagonists for the GnRH receptor to decrease the maximal response of an agonist (insurmountability) in vitro and to improve and prolong efficacy in vivo. A study of Kohout and coworkers (2007) addressed the insurmountability of a small molecule GnRH antagonist, TAK‐013. The authors examined the differences in antagonistic and kinetic properties of TAK‐013 for hGnRH receptors, mouse GnRH receptors and mutated mouse GnRH receptors and found a good correlation between the degree of insurmountability in in vitro functional assays and the dissociation rate from the receptor. Therefore, they proposed slow receptor dissociation kinetics to be accountable for the mechanism of insurmountability of TAK‐013. Similar findings were published (Sullivan et al., 2006) for another series of small molecule antagonists, that is, uracils. Slowly dissociating ligands displayed insurmountable antagonism, whereas faster dissociating ligands proved to be surmountable antagonists. To determine the dissociation rates of these uracil series of antagonists, the competition association method (Motulsky and Mahan, 1984) was used with a proprietary small molecule radioligand as a tracer. Such a competition association assay has recently been applied to determine the receptor kinetics of ligands for several different GPCRs such as the adenosine A2A receptor (Guo et al., 2012), the muscarinic M3 receptor (Sykes et al., 2009), the chemokine receptor CCR2 (Zweemer et al., 2013) and the histamine H1 and H3 receptor (Slack et al., 2011). We were able, for the first time, to determine the kinetic parameters of 12 GnRH peptide agonists, including many marketed drugs.
Two different techniques were applied, namely, radioligand‐binding studies and kPCA with a TR‐FRET read‐out. For the former, a comparison of the radioligand's kinetic parameters obtained from traditional radioligand‐binding experiments showed a good consistency with the kinetic parameters for triptorelin derived from the competition association assay (Table 2). Moreover, the kinetics of the 11 remaining GnRH agonists presented a good correlation between the kinetically derived KD and the affinity obtained from equilibrium radioligand‐binding studies (Figure 4). Secondly, we also conducted these experiments with a fluorescently labelled buserelin probe in a TR‐FRET assay. This technology has already been used for examining equilibrium GPCR ligand binding (Degorce et al., 2009; Zhang and Xie, 2012), and more recently, it was used to characterize the binding kinetics of the histamine H1 receptor (Schiele et al., 2014).
Comparing affinities and kinetic KD values from both the radioligand‐binding and TR‐FRET assays yielded significant correlations demonstrating a good reproducibility between both techniques. The two distinct assays also proved to be very amenable to the determination of the kinetic receptor‐binding parameters of (peptide) GnRH agonists. Dissociation rates, and thus residence times, between assays were in good accordance with P‐values of <0.0005, while the association rates were in less agreement between techniques. It should be noted that the experimental differences between both assays are considerable, which may have consequences for the kinetic parameters derived in the two assays. For example, the radioligand‐binding studies were manually dispensed while the TR‐FRET assays were performed using automated dispensing devices. It has been reported that compound handling can be an important source of assay variability (Gubler et al., 2013). In addition, the kinetic binding parameters were determined using a one‐concentration method for the radioligand‐binding experiments, whereas the kPCA studies used five different concentrations. Another notable difference is that in the radioligand‐binding studies, CHOhGnRH membranes were used, whereas the TR‐FRET assays were performed with Tag‐lite HEK293 GnRH cells. Packeu et al. (2008) discussed the differences in membrane interactions of membrane preparations and whole cells and their effects on binding kinetics for the D2L‐dopamine receptors. Moreover, the authors found slower dissociation rates from intact cells in comparison with membrane preparations, and they proposed that an intact cellular environment could play a role in stabilizing the D2L‐dopamine receptors in a particular conformation. A similar reasoning might be applicable to the GnRH receptors, although in our case, the receptor appears in a way that slows down the association rates of the peptides (Table 5). It may also be that the peptides simply have more difficulty in reaching the receptor on intact cells than on membrane fragments.
It might be argued that the assay temperature of 25°C is not representative for binding kinetics observed in vivo. For example, Sakai (1991) examined the effect of temperature on the dissociation of [125I]‐prolactin from the rabbit mammary gland prolactin receptor. They found a linear relationship between the dissociation rate and temperature with an increased dissociation rate at higher temperatures. Another study (Treherne and Young, 1988) also showed that the dissociation of [3H]‐QMDP from the histamine H1 receptor was temperature‐dependent, which was also true for the association rate but to a lesser extent. Arrhenius plots for both the association rate and dissociation rate of [3H]‐QMDP were linear between 6 and 37°C. It should be noted that, although these studies show a linear increase in dissociation rates with higher temperatures, the slope of this increase could be very different between targets and their ligands. Taken together, this indicates that the kinetic ranking of ligands for the same receptor can be expected to stay the same over different temperatures. Therefore, even though all our experiments were performed at 25°C, the results are still of great value for translation to in vivo outcomes.
Numerous peptide GnRH derivatives have been synthesized and studied for their so‐called structure–affinity relationships, with the aim to improve their affinity, potency and/or metabolic stability (Fujino et al., 1972; Monahan et al., 1973; Karten and Rivier, 1986; Sealfon et al., 1997; Hovelmann et al., 2002; Millar et al., 2004) In summary, it was established that the NH2‐terminal domain (pGlu–His–Trp–Ser) of GnRH is important for receptor binding and activation with Trp3 as a critical residue. In addition, the COOH‐terminal domain (Pro–Gly–NH2) is crucial for receptor binding where substitution of Pro9 or removal of NH2 results in very low affinity unless the COOH‐terminal tail is substituted for an ethylamide, which also improves metabolic stability. In contrast, the central domain of the peptide is less conserved, and studies show that exchange of Tyr5, Leu7 or Arg8 is mostly well tolerated. The most beneficial substitution is that of Gly6 with a d‐amino acid, which provides a more favourable conformation and in turn results in increased potency. d‐amino acids at the sixth position of the peptide are therefore incorporated in all the GnRH analogues marketed. The amino acid sequences of the 12 GnRH peptides tested in this study are identical with the exception of the sixth amino acid and the carboxylic tail (Table 1). A tentative structure–kinetic relationship could be established for the carboxylic tail (i.e. substitution of the glycine amide for an ethylamide). For instance, a comparison of triptorelin and deslorelin showed 2/3‐fold changes in affinity (Table 4), which was also observed in residence time. This ethylamide‐induced improvement in residence time was also true to a bigger extent for goserelin and buserelin, where the affinity was improved 2‐fold or 16‐fold (TR‐FRET and radioligand binding, respectively), while the residence time was more significantly affected, witnessed by a fivefold increase in the kPCA TR‐FRET experiments and a 20‐fold increase in the radioligand‐binding studies. This shows that shortening the carboxylic tail of the peptide slightly increases the affinity but results in a more significant improvement in residence time. Interestingly, three decades ago, it was already speculated that buserelin has a longer residence time. In these studies, the authors proposed that the high potency and long duration of action of buserelin in vivo was a result of prolonged GnRH receptors binding (Yeo et al., 1981; Koiter et al., 1984; Koiter et al., 1986). Along similar lines, Flanagan and coworkers (1998) discussed slower dissociation rates of GnRH agonists with a more hydrophobic amino acid at position 6. However, no mechanism or kinetic binding data were reported at that time.
Previously published mutagenesis studies further strengthen our hypothesis indicating the importance of the ethylamide at the carboxylic tail. Davidson and coworkers (1995) showed that the Asn2.65(102) residue located near the extracellular end of TM2 plays a role in ligand binding, specifically with the carboxylic tail of GnRH analogues. Mutations to alanine at this position significantly decreased the potency of GnRH analogues with Gly10–NH2 but had a lesser effect on GnRH analogues with an ethylamide tail (Davidson et al., 1996; Hoffmann et al., 2000; Millar et al., 2004). It may be hypothesized that substitution of Gly10–NH2 with an ethylamide moiety creates less steric hindrance and increases hydrophobicity, thereby improving the fit of the agonist and thus elongating its residence time on the receptor.
In conclusion, two novel competition association assays were successfully developed and applied to determine the kinetic binding characteristics of 12 peptide agonists, including many marketed drugs targeting the GnRH receptor. All agonists proved to have high affinity for the GnRH receptor, whereas significant differences were observed in their binding kinetics. These findings provide new insights and tools for the development of improved drugs targeting the GnRH receptor by incorporating optimized kinetic binding parameters. They also suggest that bringing this knowledge on kinetics to the clinic may help in improving or adjusting treatment protocols with better patient outcomes.
Author contributions
I. N., F. S., V. G., K. N‐R., A. E. F‐M., A. P. I. and L. H. H. participated in research design. I. N., F. S. and V. G. conducted experiments. I. N., F. S. and V. G. performed data analysis. I. N., L. H. H., A.P. I., F. S., V. G. and A. E. F‐M. wrote or contributed to the writing of the manuscript.
Conflict of interest
None.
Supporting information
Figure 1. Saturation equilibrium binding of fluorescent buserelin probe to Tag‐lite™ GnRH cells (IC50 = 5.9 nM, r2 = 0.99). Representative graph from one experiment performed in duplicate.
Figure 2. Association and dissociation kinetics of five concentrations of fluorescent buserelin probe to Tag‐lite™ GnRH cells. Representative graph from one experiment performed in duplicate.
Figure 3. kPCA traces of the GnRH peptide agonists analysed. Four concentrations (0.5, 5, 50 and 500 nM) were examined. Representative graphs from one experiment performed in duplicate.
Supporting info item
Supporting info item
Supporting info item
Acknowledgements
This study was partly undertaken within the framework of the ‘Kinetics for Drug Discovery (K4DD)’ consortium. The K4DD project is supported by the Innovative Medicines Initiative Joint Undertaking (IMI JU) under grant agreement no. 115366, resources of which are composed of financial contribution from the European Union's Seventh Framework Programme (FP7/2007‐2013) and EFPIA companies in kind contribution. The authors would like to acknowledge Delphine Jaga (Cisbio Bioassays) for excellent technical support.
Nederpelt, I. , Georgi, V. , Schiele, F. , Nowak‐Reppel, K. , Fernández‐Montalván, A. E. , IJzerman, A. P. , and Heitman, L. H. (2016) Characterization of 12 GnRH peptide agonists – a kinetic perspective. British Journal of Pharmacology, 173: 128–141. doi: 10.1111/bph.13342.
References
- Aguilar‐Rojas A, Huerta‐Reyes M, Maya‐Nunez G, Arechavaleta‐Velasco F, Conn PM, Ulloa‐Aguirre A et al. (2012). Gonadotropin‐releasing hormone receptor activates GTPase RhoA and inhibits cell invasion in the breast cancer cell line MDA‐MB‐231. BMC Cancer 12: 550–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M et al. (2013). The Concise Guide to Pharmacology 2013/14: G protein‐coupled receptors. Br J Pharmacol 170: 1459–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Angelucci C, Lama G, Iacopino F, Ferracuti S, Bono AV, Millar RP et al. (2009). GnRH receptor expression in human prostate cancer cells is affected by hormones and growth factors. Endocrine 36: 87–97. [DOI] [PubMed] [Google Scholar]
- Anthes JC, Gilchrest H, Richard C, Eckel S, Hesk D, West RE Jr et al. (2002). Biochemical characterization of desloratadine, a potent antagonist of the human histamine H(1) receptor. Eur J Pharmacol 449: 229–237. [DOI] [PubMed] [Google Scholar]
- Aydiner A, Kilic L, Yildiz I, Keskin S, Sen F, Kucucuk S et al. (2013). Two different formulations with equivalent effect? Comparison of serum estradiol suppression with monthly goserelin and trimonthly leuprolide in breast cancer patients. Med Oncol 30: 354–362. [DOI] [PubMed] [Google Scholar]
- Belchetz PE, Plant TM, Nakai Y, Keogh EJ, Knobil E (1978). Hypophysial responses to continuous and intermittent delivery of hypopthalamic gonadotropin‐releasing hormone. Science 202: 631–633. [DOI] [PubMed] [Google Scholar]
- Cheng Y, Prusoff WH (1973). Relationship between inhibition constant (K1) and concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic‐reaction. Biochem Pharmacol 22: 3099–3108. [DOI] [PubMed] [Google Scholar]
- Copeland RA, Pompliano DL, Meek TD (2006). Drug‐target residence time and its implications for lead optimization. Nat Rev Drug Discov 5: 730–739. [DOI] [PubMed] [Google Scholar]
- Davidson JS, Flanagan CA, Zhou W, Becker II, Elario R, Emeran W et al. (1995). Identification of N‐glycosylation sites in the gonadotropin‐releasing hormone receptor: role in receptor expression but not ligand binding. Mol Cell Endocrinol 107: 241–245. [DOI] [PubMed] [Google Scholar]
- Davidson JS, McArdle CA, Davies P, Elario R, Flanagan CA, Millar RP (1996). Asn102 of the gonadotropin‐releasing hormone receptor is a critical determinant of potency for agonists containing C‐terminal glycinamide. J Biol Chem 271: 15510–15514. [DOI] [PubMed] [Google Scholar]
- Degorce F, Card A, Soh S, Trinquet E, Knapik GP, Xie B (2009). HTRF: a technology tailored for drug discovery – a review of theoretical aspects and recent applications. Curr Chem Genomics 3: 22–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Depalo R, Jayakrishan K, Garruti G, Totaro I, Panzarino M, Giorgino F et al. (2012). GnRH agonist versus GnRH antagonist in in vitro fertilization and embryo transfer (IVF/ET). Reprod Biol Endocrinol 10: 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowling MR, Charlton SJ (2006). Quantifying the association and dissociation rates of unlabelled antagonists at the muscarinic M3 receptor. Br J Pharmacol 148: 927–937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flanagan CA, Fromme BJ, Davidson JS, Millar RP (1998). A high affinity gonadotropin‐releasing hormone (GnRH) tracer, radioiodinated at position 6, facilitates analysis of mutant GnRH receptors. Endocrinology 139: 4115–4119. [DOI] [PubMed] [Google Scholar]
- Fujino M, Fukuda T, Shinagaw S, White WF, Yamazaki I, Kobayasi S et al. (1972). Syntheses and biological‐activities of analogs of luteinizing‐hormone releasing hormone (Lh‐Rh). Biochem Biophys Res Commun 49: 698–705. [DOI] [PubMed] [Google Scholar]
- Goericke‐Pesch S, Georgiev P, Atanasov A, Albouy M, Navarro C, Wehrend A (2013). Treatment of queens in estrus and after estrus with a GnRH‐agonist implant containing 4.7 mg deslorelin; hormonal response, duration of efficacy, and reversibility. Theriogenology 79: 640–646. [DOI] [PubMed] [Google Scholar]
- Gubler H, Schopfer U, Jacoby E (2013). Theoretical and experimental relationships between percent inhibition and IC50 data observed in high‐throughput screening. J Biomol Screen 18: 1–13. [DOI] [PubMed] [Google Scholar]
- Guo D, Hillger JM, IJzerman AP, Heitman LH (2014). Drug‐target residence time – a case for G protein‐coupled receptors. Med Res Rev 34: 856–892. [DOI] [PubMed] [Google Scholar]
- Guo D, Mulder‐Krieger T, IJzerman AP, Heitman LH (2012). Functional efficacy of adenosine A2A receptor agonists is positively correlated to their receptor residence time. Br J Pharmacol 166: 1846–1859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heise CE, Sullivan SK, Crowe PD (2007). Scintillation proximity assay as a high‐throughput method to identify slowly dissociating nonpeptide ligand binding to the GnRH receptor. J Biomol Screen 12: 235–239. [DOI] [PubMed] [Google Scholar]
- Heitman LH, IJzerman AP (2008). G protein‐coupled receptors of the hypothalamic‐pituitary‐gonadal axis: a case for Gnrh, LH, FSH, and GPR54 receptor ligands. Med Res Rev 28: 975–1011. [DOI] [PubMed] [Google Scholar]
- Heitman LH, Ye K, Oosterom J, Ijzerman AP (2008). Amiloride derivatives and a nonpeptidic antagonist bind at two distinct allosteric sites in the human gonadotropin‐releasing hormone receptor. Mol Pharmacol 73: 1808–1815. [DOI] [PubMed] [Google Scholar]
- Hoffmann SH, ter Laak T, Kuhne R, Reilander H, Beckers T (2000). Residues within transmembrane helices 2 and 5 of the human gonadotropin‐releasing hormone receptor contribute to agonist and antagonist binding. Mol Endocrinol 14: 1099–1115. [DOI] [PubMed] [Google Scholar]
- Hovelmann S, Hoffmann SH, Kuhne R, ter Laak T, Reilander H, Beckers T (2002). Impact of aromatic residues within transmembrane helix 6 of the human gonadotropin‐releasing hormone receptor upon agonist and antagonist binding. Biochemistry 41: 1129–1136. [DOI] [PubMed] [Google Scholar]
- Kakar SS, Grizzle WE, Neill JD (1994). The nucleotide‐sequences of human GnRH receptors in breast and ovarian‐tumors are identical with that found in pituitary. Mol Cell Endocrinol 106: 145–149. [DOI] [PubMed] [Google Scholar]
- Karten MJ, Rivier JE (1986). Gonadotropin‐releasing‐hormone analog design – structure‐function studies toward the development of agonists and antagonists – rationale and perspective. Endocr Rev 7: 44–66. [DOI] [PubMed] [Google Scholar]
- Kohout TA, Xie Q, Reijmers S, Finn KJ, Guo Z, Zhu YF et al. (2007). Trapping of a nonpeptide ligand by the extracellular domains of the gonadotropin‐releasing hormone receptor results in insurmountable antagonism. Mol Pharmacol 72: 238–247. [DOI] [PubMed] [Google Scholar]
- Koiter TR, Denef C, Andries M, Moes H, Schuiling GA (1986). The prolonged action of the LHRH agonist buserelin (HOE 766) may be due to prolonged binding to the LHRH receptor. Life Sci 39: 443–452. [DOI] [PubMed] [Google Scholar]
- Koiter TR, van der Schaaf‐Verdonk GC, Kuiper H, Pols‐Valkhof N, Schuiling GA (1984). A comparison of the LH‐releasing activities of LH‐RH and its agonistic analogue buserelin in the ovariectomized rat. Life Sci 34: 1597–1604. [DOI] [PubMed] [Google Scholar]
- Labrie F (2014). GnRH agonists and the rapidly increasing use of combined androgen blockade in prostate cancer. Endocr Relat Cancer 4: 301–317. [DOI] [PubMed] [Google Scholar]
- Lahlou N, Roger M, Chaussain JL, Feinstein MC, Sultan C, Toublanc JE et al. (1987). Gonadotropin and alpha‐subunit secretion during long term pituitary suppression by D‐Trp6‐luteinizing hormone‐releasing hormone microcapsules as treatment of precocious puberty. J Clin Endocrinol Metab 65: 946–953. [DOI] [PubMed] [Google Scholar]
- Leone Roberti Maggiore U, Scala C, Remorgida V, Venturini PL, Del Deo F, Torella M et al. (2014). Triptorelin for the treatment of endometriosis. Expert Opin Pharmacother 15: 1153–1179. [DOI] [PubMed] [Google Scholar]
- Lewis KA, Goldyn AK, West KW, Eugster EA (2013). A single histrelin implant is effective for 2 years for treatment of central precocious puberty. J Pediatr 163: 1214–1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maillard MP, Perregaux C, Centeno C, Stangier J, Wienen W, Brunner HR et al. (2002). In vitro and in vivo characterization of the activity of telmisartan: an insurmountable angiotensin II receptor antagonist. J Pharmacol Exp Ther 302: 1089–1095. [DOI] [PubMed] [Google Scholar]
- Millar RP, Lu ZL, Pawson AJ, Flanagan CA, Morgan K, Maudsley SR (2004). Gonadotropin‐releasing hormone receptors. Endocr Rev 25: 235–275. [DOI] [PubMed] [Google Scholar]
- Millar RP, Newton CL (2013). Current and future applications of GnRH, kisspeptin and neurokinin B analogues. Nat Rev Endocrinol 9: 451–466. [DOI] [PubMed] [Google Scholar]
- Monahan MW, Amoss MS, Anderson HA, Vale W (1973). Synthetic analogs of hypothalamic luteinizing‐hormone releasing factor with increased agonist or antagonist properties. Biochemistry 12: 4616–4620. [DOI] [PubMed] [Google Scholar]
- Motulsky HJ, Mahan LC (1984). The kinetics of competitive radioligand binding predicted by the law of mass action. Mol Pharmacol 25: 1–9. [PubMed] [Google Scholar]
- Packeu A, De Backer JP, Van Liefde I, Vanderheyden PM, Vauquelin G (2008). Antagonist‐radioligand binding to D2L‐receptors in intact cells. Biochem Pharmacol 75: 2192–2203. [DOI] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP et al. (2014). The IUPHAR/BPS Guide to PHARMACOLOGY: an expert‐driven knowledgebase of drug targets and their ligands. Nucl Acids Res 42 (Database Issue): D1098–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Romero E, Velez de Mendizabal N, Cendros JM, Peraire C, Bascompta E, Obach R et al. (2012). Pharmacokinetic/pharmacodynamic model of the testosterone effects of triptorelin administered in sustained release formulations in patients with prostate cancer. J Pharmacol Exp Ther 342: 788–798. [DOI] [PubMed] [Google Scholar]
- Sakai S (1991). Effect of hormones on dissociation of prolactin from the rabbit mammary gland prolactin receptor. Biochem J 279 (Pt 2): 461–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiele F, Ayaz P, Fernandez‐Montalvan A (2014). A universal, homogenous assay for high throughput determination of binding kinetics. Anal Biochem 486C: 42–49. [DOI] [PubMed] [Google Scholar]
- Sealfon SC, Weinstein H, Millar RP (1997). Molecular mechanisms of ligand interaction with the gonadotropin‐releasing hormone receptor. Endocr Rev 18: 180–205. [DOI] [PubMed] [Google Scholar]
- Slack RJ, Russell LJ, Hall DA, Luttmann MA, Ford AJ, Saunders KA et al. (2011). Pharmacological characterization of GSK1004723, a novel, long‐acting antagonist at histamine H(1) and H(3) receptors. Br J Pharmacol 164: 1627–1641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith DA, Jones BC, Walker DK (1996). Design of drugs involving the concepts and theories of drug metabolism and pharmacokinetics. Med Res Rev 16: 243–266. [DOI] [PubMed] [Google Scholar]
- Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, Provenzano MD et al. (1985). Measurement of protein using bicinchoninic acid. Anal Biochem 150: 76–85. [DOI] [PubMed] [Google Scholar]
- Stojilkovic SS, Reinhart J, Catt KJ (1994). Gonadotropin‐releasing hormone receptors: structure and signal transduction pathways. Endocr Rev 15: 462–499. [DOI] [PubMed] [Google Scholar]
- Sullivan SK, Hoare SR, Fleck BA, Zhu YF, Heise CE, Struthers RS et al. (2006). Kinetics of nonpeptide antagonist binding to the human gonadotropin‐releasing hormone receptor: implications for structure‐activity relationships and insurmountable antagonism. Biochem Pharmacol 72: 838–849. [DOI] [PubMed] [Google Scholar]
- Swinney DC (2004). Biochemical mechanisms of drug action: what does it take for success? Nat Rev Drug Discov 3: 801–808. [DOI] [PubMed] [Google Scholar]
- Swinney DC (2009). The role of binding kinetics in therapeutically useful drug action. Curr Opin Drug Discov Devel 12: 31–39. [PubMed] [Google Scholar]
- Sykes DA, Dowling MR, Charlton SJ (2009). Exploring the mechanism of agonist efficacy: a relationship between efficacy and agonist dissociation rate at the muscarinic M3 receptor. Mol Pharmacol 76: 543–551. [DOI] [PubMed] [Google Scholar]
- Tashkin DP (2005). Is a long‐acting inhaled bronchodilator the first agent to use in stable chronic obstructive pulmonary disease? Curr Opin Pulm Med 11: 121–128. [DOI] [PubMed] [Google Scholar]
- Treherne JM, Young JM (1988). Temperature‐dependence of the kinetics of the binding of [3H]‐(+)‐N‐methyl‐4‐methyldiphenhydramine to the histamine H1‐receptor: comparison with the kinetics of [3H]‐mepyramine. Br J Pharmacol 94: 811–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tummino PJ, Copeland RA (2008). Residence time of receptor‐ligand complexes and its effect on biological function. Biochemistry 47: 5481–5492. [DOI] [PubMed] [Google Scholar]
- Vauquelin G, Van Liefde I (2006). Slow antagonist dissociation and long‐lasting in vivo receptor protection. Trends Pharmacol Sci 27: 356–359. [DOI] [PubMed] [Google Scholar]
- von Alten J, Fister S, Schulz H, Viereck V, Frosch KH, Emons G et al. (2006). GnRH analogs reduce invasiveness of human breast cancer cells. Breast Cancer Res Tr 100: 13–21. [DOI] [PubMed] [Google Scholar]
- Yeo T, Grossman A, Belchetz P, Besser GM (1981). Response of luteinizing hormone from columns of dispersed rat pituitary cells to a highly potent analogue of luteinizing hormone releasing hormone. J Endocrinol 91: 33–41. [DOI] [PubMed] [Google Scholar]
- Zhang R, Monsma F (2009). The importance of drug‐target residence time. Curr Opin Drug Discov Devel 12: 488–496. [PubMed] [Google Scholar]
- Zhang R, Xie X (2012). Tools for GPCR drug discovery. Acta Pharmacol Sin 33: 372–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zweemer AJ, Nederpelt I, Vrieling H, Hafith S, Doornbos ML, de Vries H et al. (2013). Multiple binding sites for small‐molecule antagonists at the CC chemokine receptor 2. Mol Pharmacol 84: 551–561. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure 1. Saturation equilibrium binding of fluorescent buserelin probe to Tag‐lite™ GnRH cells (IC50 = 5.9 nM, r2 = 0.99). Representative graph from one experiment performed in duplicate.
Figure 2. Association and dissociation kinetics of five concentrations of fluorescent buserelin probe to Tag‐lite™ GnRH cells. Representative graph from one experiment performed in duplicate.
Figure 3. kPCA traces of the GnRH peptide agonists analysed. Four concentrations (0.5, 5, 50 and 500 nM) were examined. Representative graphs from one experiment performed in duplicate.
Supporting info item
Supporting info item
Supporting info item
