Abstract
The human adenosine A3 (hA3) receptor has been suggested as a viable drug target in inflammatory diseases and in cancer. So far, a number of selective hA3 receptor agonists (e.g. IB-MECA and 2-Cl-IB-MECA) inducing anti-inflammatory or anticancer effects are under clinical investigation. Drug-target binding kinetics is increasingly recognized as another pharmacological parameter, next to affinity, for compound triage in the early phases of drug discovery. However, such a kinetics-driven analysis has not yet been performed for the hA3 receptor. In this study, we first validated a competition association assay for adenosine A3 receptor agonists to determine the target interaction kinetics. Affinities and Kinetic Rate Index (KRI) values of 11 ribofurano and 10 methanocarba nucleosides were determined in radioligand binding assays. Afterwards, 15 analogues were further selected (KRI <0.70 or KRI >1.35) for full kinetics characterization. The structure-kinetics relationships (SKR) were derived and longer residence times were associated with methanocarba and enlarged adenine N6 and C2 substitutions. In addition, from a kon-koff-KD kinetic map we divided the agonists into three subgroups. A residence time “cliff” was observed, which might be relevant to (N)-methanocarba derivatives' rigid C2-arylalkynyl substitutions. Our findings provide substantial evidence that, next to affinity, additional knowledge of binding kinetics is useful for developing and selecting new hA3R agonists in the early phase of the drug discovery process.
Keywords: Residence time, Structure-kinetics relationships, Adenosine A3 receptor, Binding Kinetics, Nucleosides
Graphical abstract

1. Introduction
The adenosine A3 receptor is the youngest member discovered in the family of adenosine receptors (A1, A2A, A2B and A3), all of which belong to class A G protein-coupled receptors (GPCRs)[1]. Unlike the other subtypes, the adenosine A3 receptor is endogenously activated by both adenosine and inosine[2]. Following agonist activation, the receptor causes a decrease in cAMP levels as it primarily couples to Gi protein. The adenosine A3 receptor is widely distributed throughout the body, albeit at low expression levels, and its activation can affect numerous organs, tissues and systems. However, this receptor is overexpressed in inflammatory and cancer cells, reflecting its importance as a therapeutic target and biological marker in these tissues [3, 4]. Moreover, the adenosine A3 receptor is rapidly internalized within 30 min after agonist exposure [5, 6]. It is unresolved whether receptor desensitization terminates agonist-induced signaling or if this signaling persists after internalization[7].
Since agonists cause effects of anti-inflammation, cardioprotection and neuroprotection, numerous potent and selective agonists have been extensively studied and developed[8-13]. Most of these agonists are derivatives of adenosine, with substitutions on the adenine nucleobase or the ribose moiety. In particular, IB-MECA and 2-Cl-IB-MECA are two potent clinical candidates with excellent bioavailability and safety profiles. Selective hA3 receptor agonists are being evaluated for the treatment of chronic inflammatory diseases (e.g., rheumatoid arthritis or psoriasis), neuropathic pain and cancer (i.e. hepatocellular carcinoma)[14-17].
Traditional drug discovery approaches mainly focus on affinity, a parameter that is defined under equilibrium conditions. However, it is emerging that selecting ligands based on their affinity alone, does not necessarily predict in vivo efficacy very well. This may be due to the dynamic conditions in vivo, that often are in contrast to the equilibrium conditions applied in in vitro assays[18]. In fact a ligand's kinetic properties may provide a better indication of how a ligand will perform in vivo[19]. Notably, the parameter of residence time (RT) has been proposed as a more relevant selection criterion. The RT reflects the lifetime of the ligand-receptor complex and can be calculated as the reciprocal of the ligand's dissociation constant [20, 21].
Although the binding kinetics of hA3 receptor agonists are occasionally reported [22, 23], a systematic kinetics analysis yielding structure-kinetics relationships (SKRs) has not been conducted. Therefore, we firstly validated the binding kinetics of the prototypical hA3 receptor agonists (i.e. IB-MECA and 2-Cl-IB-MECA), using radioligand displacement and competition association assays. Then, two series of ribofurano and methanocarba ([3.1.0]bicyclohexane) adenosine derivatives were evaluated for both their affinity (Ki) and kinetics (kinetic rate index values, kon, koff, and RTs). This allowed a complete SKR analysis next to a more traditional SAR study. Afterwards, a retrospective evaluation linking residence times and in vivo efficacies was discussed. Last but not least, from a kon-koff-KD kinetic map we divided the agonists into three subgroups, providing a possible direction for the further development of hA3R agonists.
2. Materials & Methods
2.1. Chemicals and reagents
[3H]8-Ethyl-4-methyl-2-phenyl-(8R)-4,5,7,8-tetrahydro-1H-imidazo[2,1-i]-purin-5-one ([3H]PSB-11, specific activity 56 Ci·mmol-1) was obtained with the kind help of Prof. C.E. Müller (University of Bonn, Germany). 5′-N-methylcarboxamidoadenosine (MECA) was provided by one of the authors (KAJ). Unlabeled PSB-11, 1-deoxy-1-[6-[((3-Iodophenyl)methyl)amino]-9H-purin-9-yl]-N-methyl-β-D-ribofuranuronamide (IB-MECA), and 2-chloro-N6-(3-iodobenzyl)-adenosine-5′-N-methyluronamide (2-Cl-IB-MECA) were purchased from Tocris Ltd. (Abingdon, UK). 5′-N-ethylcarboxamidoadenosine (NECA) was purchased from Sigma-Aldrich (Steinheim, Germany). LUF5501, LUF5505, LUF5500, LUF5506 and LUF5521 have been synthesized and described previously by de Zwart et al.[24]; LUF5595, LUF5586 and LUF5589 have been described by van Tilburg et al.[25]; MRS7140, MRS5980, MRS7154, MRS7549, MRS5655, MRS5679, MRS5698, MRS5644, MRS5667 and MRS7294 have been described by Tosh et al.[26-29], and MRS3558 by Tchilibon et al.[30]. Adenosine deaminase (ADA) was purchased from Boehringer Mannheim (Mannheim, Germany). Bicinchoninic acid (BCA) and BCA protein assay reagents were purchased from Pierce Chemical Company (Rockford, IL, USA). Chinese hamster ovary (CHO) cells stably expressing the human adenosine A3 receptor (CHOhA3) were a gift from Dr. K-N Klotz (University of Würzburg, Germany). All other chemicals were obtained from standard commercial sources and were of analytical grade.
2.2 Cell culture and membrane preparation
CHOhA3 cells were cultured and membranes were prepared and stored as previously described [22]. Protein determination was done through use of the bicinchoninic acid (BCA) method [31].
2.3 Radioligand equilibrium displacement assays
Membrane aliquots containing ∼15 μg of CHOhA3 protein were incubated in a total volume of 100 μL assay buffer (50 mM Tris-HCl, 5 mM MgCl2, supplemented with 0.01% CHAPS and 1 mM EDTA, pH 7.4) at 10 °C for 240 min. Displacement experiments were performed using 6 concentrations of competing agonist in the presence of a final concentration of ∼10 nM [3H]PSB-11. At this concentration, total radioligand binding did not exceed 10% of that added to prevent ligand depletion. Nonspecific binding (NSB) was determined in the presence of 100 μM NECA. Incubation was terminated by rapid filtration performed on 96-well GF/B filter plates (Perkin Elmer, Groningen, the Netherlands), using a PerkinElmer Filtermate-harvester (Perkin Elmer, Groningen, the Netherlands). After drying the filter plate at 50 °C for 30 min, the filter-bound radioactivity was determined by scintillation spectrometry using the 2450 MicroBeta2 Plate Counter (Perkin Elmer, Boston, MA).
2.4 Radioligand competition association assays
The binding kinetics of unlabeled ligands were quantified using the competition association assay based on the theoretical framework by Motulsky and Mahan [32]. The competition association assay was initiated by adding membrane aliquots (15 μg/well) at different time points for a total of 240 min to a total volume of 100 μl of assay buffer at 10 °C with ∼10 nM [3H]PSB-11 in the absence or presence of a single concentration of competing hA3 receptor agonists (i.e. at their IC50 value), and, in some experiments indicated, in the simultaneous presence of 1 mM GTP. Incubations were terminated and samples were obtained as described under Radioligand Displacement Assay. The “dual-point” competition association assays were designed as described previously [33], where in this case the two time points were selected at 20 min (t1) and 240 min (t2).
2.5 Data analysis
All experimental data were analyzed using the nonlinear regression curve fitting program GraphPad Prism 7.0 (GraphPad Software, Inc., San Diego, CA). From displacement assays, IC50 values were obtained by non-linear regression analysis of the displacement curves. The obtained IC50 values were converted into Ki values using the Cheng-Prusoff equation to determine the affinity of the ligands [34], using a KD value for the radioligand of 1.04 nM at 10 °C as determined from radioligand association and dissociation assays, as previously reported [35]. The residence time (RT, in min) was calculated using the equation RT = 1/(60 * koff) ), as koff is in s-1. Association and dissociation rate constants for unlabeled compounds were calculated by fitting the data into the competition association model using “kinetics of competitive binding”:
where k1 is the kon of the radioligand (M-1s-1), k2 is the koff of the radioligand (s-1), L is the radioligand concentration (nM), I is the concentration of the unlabeled competitor (nM), X is the time (s) and Y is the specific binding of the radioligand (DPM). The control curve (without competitor) from competition association assays generates the k1 value, and the k2 value was obtained from previous radioligand association and dissociation assays [35]. With that the k3, k4 and Bmax were calculated, where k3 represents the kon (M-1s-1) of the unlabeled ligand, k4 stands for the koff (s-1) of the unlabeled ligand and Bmax equals the total binding (DPM). All competition association data were globally fitted.
3. Results
3.1 The effects of GTP on affinities and kinetics of the reference hA3 receptor agonists IB-MECA and 2-Cl-IB-MECA
Firstly, we determined the effects of GTP (1 mM) on both the affinities and binding kinetics of the reference hA3 receptor agonists IB-MECA and 2-Cl-IB-MECA. Under the two different assay conditions (i.e. in the absence or presence of GTP) the affinities of IB-MECA and 2-Cl-IB-MECA were significantly different (Table 1). We observed a rightward shift of the displacement curves in the presence of GTP (Fig. 1A, closed symbols), representing an approx. 3–fold decrease in affinity for both compounds. Of note, under both experimental conditions, monophasic displacement curves were obtained with pseudo-Hill coefficients close to unity (Table 1).
Table 1.
The effects of GTP (1 mM) on the affinities and kinetics of hA3 receptor agonists IB-MECA and 2-Cl-IB-MECA.
| Agonists | pKi (mean Ki in nM)a | Hill Slope a | KRIb | kon (M-1 s-1)c | koff (s-1)d | RT (min)e | Kinetic KD (mean pKD)f |
|---|---|---|---|---|---|---|---|
|
| |||||||
| no GTP | |||||||
|
| |||||||
| IB-MECA | 8.5 ± 0.07 *** (2.9) | -0.94 ± 0.05 ns | 1.55 ± 0.07 ** | (5.9 ± 0.9) × 105 ns | (1.8 ± 0.2) × 10-4 ns | 95 ± 13 * | 0.30 ± 0.06 (9.5) |
| 2-Cl-IB-MECA | 8.5 ± 0.1* (3.5) | -0.85 ± 0.21 ns | 2.02 ± 0.06 *** | (4.0 ± 0.5) × 105 ns | (7.2 ± 1.1) × 10-5 * | 231 ± 34 * | 0.18 ± 0.04 (9.7) |
|
| |||||||
| + 1 mM GTP | |||||||
|
| |||||||
| IB-MECA | 8.0 ± 0.02 (11) | -0.99 ± 0.07 | 0.99 ± 0.08 | (4.5 ± 2.5) × 105 | (8.5 ± 3.0) × 10-4 | 20 ± 7 | 1.9 ± 1.2 (8.7) |
|
| |||||||
| 2-Cl-IB-MECA | 8.0 ± 0.08 (10) | -1.1 ± 0.1 | 1.07 ± 0.07 | (4.0 ± 2.3) × 105 | (5.7 ± 1.5) × 10-4 | 29 ± 8 | 1.4 ± 0.9 (8.9) |
pKi ± SEM (n ≥ 3, mean Ki value in nM), obtained from radioligand binding assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
KRI ± SEM (n = 3) obtained from dual-point competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
kon ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
koff ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
RT = 1/(60*koff); RT is expressed in min, whereas koff is expressed in s-1.
KD = koff/kon
Student's t-test was applied for the comparison of values of pKi, KRI, kon, koff, RT obtained in the absence vs presence of GTP (1 mM) in the assay conditions,
p < 0.0005,
p < 0.005,
p < 0.05, ns for not significant.
Figure 1.
Displacement of specific [3H]PSB-11 binding from the recombinant hA3 receptor stably expressed on CHO cell membranes by IB-MECA and 2-Cl-IB-MECA in the absence (closed symbols) or presence of 1 mM GTP (open symbols) (A); [3H]PSB-11 competition association experiments in the absence (B) or presence (C) of 1 mM GTP with IB-MECA (blue) or 2-Cl-IB-MECA (red). Combined graphs are shown from at least three experiments performed in duplicate (see Table 1 for pKi values and kinetic parameters). Note, t1, t2 are indicated, which are the two time points used in KRI determinations.
Furthermore, there were significant differences between the kinetics of hA3 receptor agonists tested in the absence or presence of 1 mM GTP. In the competition association assays without GTP, both IB-MECA and 2-Cl-IB-MECA produced a typical “overshoot”, representative of a slower dissociation from the hA3 receptor than the radioligand [3H]PSB-11 (Fig. 1B). This overshoot, however, disappeared in the experiments with GTP (Fig. 1C). Thus, their KRI values were decreased from well-above-unity in the absence of GTP to close-to-unity in the presence of GTP. The koff values of IB-MECA and 2-Cl-IB-MECA determined by the Motulsky-Mahan model[32] were (1.8 ± 0.2) × 10-4 s-1 and (7.2 ± 1.1) × 10-5 s-1 in the absence of GTP, respectively (Table 1), which were smaller than the corresponding values determined in the presence of GTP (IB-MECA: (8.5 ± 3.0) × 10-4 s-1; 2-Cl-IB-MECA: (5.7 ± 1.5) × 10-4 s-1, Table 1). Consequently, the calculated RTs in the absence of GTP were longer than the values derived when GTP was present (IB-MECA: 95 min vs 20 min; 2-Cl-IB-MECA 231 min vs 29 min, Table 1). Last but not least, the kon values of these two agonists determined under the two assay conditions were similar. Therefore, in order to define a more agonist-relevant receptor interaction, we decided to use the competition association assay in the absence of GTP for the determination of the affinities and the binding kinetics of other unlabeled hA3 receptor agonists in the remainder of the study.
3.2 Binding affinity (Ki) of hA3 receptor agonists
The binding affinities of 22 human A3 receptor agonists in total were determined in equilibrium radioligand displacement studies. All agonists were able to concentration-dependently inhibit specific [3H]PSB-11 binding to the human A3 receptor and their affinities are listed in Tables 2 and 3. The agonists displayed moderate to high binding affinities, ranging from 82 nM for agonist MRS5679 to 0.72 nM for agonist MRS5980, while the reference agonists IB-MECA and 2-Cl-IB-MECA had an affinity of 2.9 and 3.5 nM, respectively.
Table 2.
Binding Affinities and Kinetic Parameters (KRI, kon, koff, RT and KD) for ribofurano derivatives as hA3 receptor agonists.
| ||||||||||
|---|---|---|---|---|---|---|---|---|---|---|
|
| ||||||||||
| Agonists | R1 | R2 | R3 | pKi (mean Ki in nM) a | KRI b | kon (M-1 s-1)c | koff (s-1) d | RT (min) e | Kinetic KD (mean pKD)f | |
| LUF5501 | H | -Ph-3-Cl | -CH2OH | 7.5 ± 0.02 (31) | 0.64 (0.53; 0.75) | (1.7 ± 0.2) × 105 | (1.6 ± 0.2) × 10-3 | 10 ± 1.3 | 9.2 ± 1.4 (8.0) | |
| LUF5505 | H | -Ph-3-Br | -CH2OH | 7.1 ± 0.1 (78) | 0.65 (0.63; 0.66) | (8.8 ± 1.4) × 104 | (1.1 ± 0.04) × 10-3 | 16 ± 0.6 | 12 ± 1.9 (7.9) | |
| LUF5500 | H | -Ph-4-Cl | -CH2OH | 7.3 ± 0.03 (51) | 0.98 (0.95; 1.01) | N.D. | N.D. | N.D. | N.D. | |
| LUF5506 | H | -Ph-4-I | -CH2OH | 7.6 ± 0.08 (29) | 0.99 (0.98; 1.00) | N.D. | N.D. | N.D. | N.D. | |
| LUF5521 | H | -Ph-4-I | -CONHCH2CH3 | 7.9 ± 0.06 (14) | 2.06 ± 0.82 | (1.9 ± 0.3) × 105 | (1.4 ± 0.1) × 10-4 | 117 ± 12 | 0.76 ± 0.13 (9.1) | |
| MECA | H | -CH2Ph | -CONHCH3 | 7.4 ± 0.04 (42) | 1.24 ± 0.09 | N.D.h | N.D. | N.D. | N.D. | |
| IB-MECAg | H | -CH2Ph-3-I | -CONHCH3 | 8.5 ± 0.07 (2.9) | 1.55 ± 0.07 | (5.9 ± 0.9) × 105 | (1.8 ± 0.2) × 10-4 | 95 ± 13 | 0.30 ± 0.06 (9.5) | |
| 2-Cl-IB-MECAg | -Cl | -CH2Ph-3-I | -CONHCH3 | 8.5 ± 0.1 (3.5) | 2.02 ± 0.06 | (4.0 ± 0.5) × 105 | (7.2 ± 1.1) × 10-5 | 231 ± 34 | 0.18 ± 0.04 (9.7) | |
| LUF5595 | -Cl | -CH2Ph-3-I | -CH2OC3H5 | 8.5 ±0.09 (3.2) | 1.37 (1.39; 1.35) | (9.6 ± 3.1) × 105 | (2.3 ± 1.0) × 10-4 | 72 ± 30 | 0.24 ± 0.13 (9.6) | |
| LUF5586 | H | -CH2Ph-3-I | -CH2OCH2CH3 | 8.6 ± 0.04 (2.8) | 1.30 (1.30; 1.29) | N.D. | N.D. | N.D. | N.D. | |
| LUF5589 | -Cl | -CH2Ph-3-I | -CH2OCH2CH3 | 8.7 ± 0.08 (2.2) | 1.66 ± 0.21 | (8.4 ± 0.7) × 105 | (1.2 ± 0.3) × 10-4 | 138 ± 37 | 0.14 ± 0.04 (9.8) | |
| MRS7294 | -C≡C-C4H2S-5-Cl | -CH3 | -CH2OH | 8.4 ± 0.05 (3.7) | 1.63 (1.50; 1.75) | (8.2 ± 3.8) × 105 | (1.3 ± 0.2) × 10-4 | 125 ± 14 | 0.16 ± 0.02 (9.8) | |
pKi ± SEM (n ≥ 3, mean Ki value in nM), obtained from radioligand binding assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
KRI ± SEM (n = 3) or KRI (n1,n2) (n = 2), obtained from dual-point competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
kon ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
koff ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
RT = 1/(60 * koff); RT is expressed in min, whereas koff is expressed in s-1.
KD = koff/kon, expressed in nM
values taken from Table 1
N.D. = not determined
Table 3.
Binding Affinities and Kinetic Parameters (KRI, kon, koff, RT and KD) for (N)-methanocarba derivatives as hA3 receptor agonists.
| |||||||||
|---|---|---|---|---|---|---|---|---|---|
|
| |||||||||
| Agonists | X | R1 | R2 | pKi (mean Ki value in nM) a | KRI b | kon (M-1 s-1)c | koff (s-1) d | RT (min) e | Kinetic KD (mean pKD)f |
| MRS7294 g | N | -C≡C-C4H2S-5-Cl | -CH3 | 8.4 ± 0.05 (3.7) | 1.63 (1.50; 1.75) | (8.2 ± 3.8) × 105 | (1.3 ± 0.2) × 10-4 | 125 ± 14 | 0.16 ± 0.02 (9.8) |
| MRS5980 | N | -C≡C-C4H2S-5-Cl | -CH3 | 9.2 ± 0.09 (0.72) | 1.97 (1.87; 2.06) | (9.0 ± 2.3) × 105 | (4.0 ± 1.0) × 10-5 | 417 ± 104 | 0.044 ± 0.016 (10.4) |
| MRS7140 | C | -C≡C-C4H2S-5-Cl | -CH3 | 8.3 ± 0.08 (5.2) | 1.94 (1.82; 2.07) | (3.2 ± 1.2) × 105 | (8.5 ± 0.3) × 10-5 | 196 ± 8 | 0.27 ± 0.10 (9.6) |
| MRS7154 | N | -C≡C-C4H2S-5-Cl | -CH2CH2CH3 | 8.7 ± 0.06 (2.2) | 1.67 (1.75; 1.59) | (4.0 ± 0.7) × 105 | (6.2 ± 1.2) × 10-5 | 270 ± 51 | 0.15 ± 0.04 (9.8) |
| MRS3558 | N | -Cl | -CH2Ph-3-Cl | 9.1 ± 0.2 (1.0) | 1.49 (1.33; 1.64) | (5.7 ± 0.8) × 105 | (1.0 ± 0.2) × 10-4 | 167 ± 39 | 0.18 ± 0.05 (9.8) |
| MRS5655 | N | -C≡C-Ph | -CH2Ph-3-Cl | 8.5 ± 0.09 (3.6) | 2.58 (2.05; 3.12) | (4.0 ± 0.8) × 105 | (1.8 ± 0.3) × 10-5 | 909 ± 165 | 0.046 ± 0.013 (10.3) |
| MRS5644 | N | -C≡C-Ph | -CH3 | 8.6 ± 0.01 (2.4) | 2.42 (2.28; 2.56) | (4.5 ± 1.8) × 105 | (1.4 ± 0.2) × 10-5 | 1205 ± 174 | 0.031 ± 0.013 (10.5) |
| MRS5698 | N | -C≡C-(3,4-di-F-Ph) | -CH2Ph-3-Cl | 8.1 ± 0.2 (9.4) | 4.21 (3.75; 4.67) | (1.3 ± 0.4) × 105 | (8.5 ± 0.7) × 10-6 | 1961 ± 154 | 0.068 ± 0.020 (10.2) |
| MRS5679 | N | -C≡C-(4-biphenyl) | -CH2Ph-3-Cl | 7.1 ± 0.05 (82) | 5.56 (6.00; 5.11) | undefined | undefined | undefined | undefined |
| MRS5667 | N | -C≡C-(4-biphenyl) | -CH3 | 8.3 ± 0.05 (4.6) | 3.34 (3.14; 3.55) | (1.8 ± 0.3) × 105 | (1.1 ± 0.3) × 10-5 | 1563 ± 464 | 0.058 ± 0.020 (10.2) |
pKi ± SEM (n ≥ 3, mean Ki value in nM), obtained from radioligand binding assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
KRI ± SEM (n = 3) or KRI (n1,n2) (n = 2), obtained from dual-point competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
kon ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
koff ± SEM (n ≥ 3), obtained from competition association assays with [3H]PSB-11 on the hA3 receptor stably expressed on CHO cell membranes.
RT = 1/(60 * koff); RT is expressed in min, whereas koff is expressed in s-1.
KD = koff/kon, expressed in nM
MRS7294 has a ribofurano ring (see also Table 2).
3.3 Kinetic Rate Index (KRI) values of hA3 receptor agonists
Subsequently, these hA3 receptor agonists were screened in what is termed a “dual-point” competition association assay. The specific binding of [3H]PSB-11 was measured after 20 and 240 minutes in the absence and presence of a single concentration (i.e. IC50) of unlabeled hA3 receptor agonists, which yielded their Kinetic Rate Index (KRI). The KRI values of the hA3 receptor agonists ranged from 0.64 (LUF5501) to 5.56 (MRS5679) (Tables 2 and 3). Agonists with a KRI value larger than unity are considered to have a slower dissociation rate, and thus a longer RT, than the radioligand used, i.e. [3H]PSB-11, and vice versa. Agonists having KRI values below 0.70 (LUF5501 and LUF5505) or above 1.35 (LUF5521, IBMECA to LUF5595, LUF5589, MRS7140, MRS5980, and MRS7154 to MRS5667) were selected for further kinetic profiling.
3.4 Binding kinetics of selected hA3 receptor agonists using the competition association assay
Next, the kinetic binding parameters of selected agonists that had either low or high KRI values were determined using the competition association assay with [3H]PSB-11. Association rate constants varied by only 11-fold, ranging from (8.8 ± 1.4) × 104 M-1s-1 for LUF5505 to (9.6 ± 3.1) × 105 M-1s-1 for LUF5595 (Tables 2 and 3). There was a more pronounced 188-fold difference in dissociation rate constants, in line with the divergent KRI values (LUF5501: (1.6 ± 0.2) × 10-3 s-1 vs MRS5698: (8.5 ± 0.7) × 10-6 s-1). The shortest RT agonist, LUF5501, presented a gradually ascending curve in the competition association assays (Fig. 2A). Of note, the dissociation of MRS5679, with the largest KRI value (5.56), was determined as very slow and not readily quantitated under the current method. The very prominent “overshoot” in the competition association curve was already indicative of a much slower dissociation rate than [3H]PSB-11 (Fig. 2B). Notably, a significant correlation between the negative logarithm of the dissociation rate constants for the selected agonists and their KRI values derived from the kinetic assays was obtained (Fig. 3A), which confirmed that a compound's KRI value is a good predictor for its dissociation rate constant. Besides, a significant correlation was also observed between the agonist affinities (pKi values) determined in equilibrium displacement experiments and their pKD values (“kinetic KD values”) derived from competition association experiments, although the pKD values were on average one log unit higher than the pKi values (Fig. 3B). As to the kinetic rate constants (kon or koff) of the hA3 receptor agonists, their association rate constants showed a better correlation with affinity than their dissociation rate constants (Fig. 3C and 3D). Furthermore, a kon-koff-KD “kinetic map” (Fig. 4) was constructed based on the compounds' divergent affinities (expressed as kinetic KD values) and rate constants, yielding a division of these agonists into three different sub-categories: agonists that show koff values in a lower range but due to divergent kon values have various KD values (Group A); agonists that show koff values in a median range but due to divergent kon values (although smaller than in Group A) have different KD values (Group B); and two agonists that have relatively poor affinity due to larger koff values and smaller kon values (Group C).
Figure 2.
Competition association experiments with [3H]PSB-11 binding to the recombinant hA3 receptor stably expressed on CHO cell membranes (10 °C) in the absence or presence of unlabeled short-residence-time agonist LUF5501 (A), or long-residence-time agonist MRS5679 (B). Representative graphs are shown from one experiment performed in duplicate. Note, t1, t2 are indicated, which are the two time points used in KRI determinations.
Figure 3.
The correlations between the negative logarithm of each hA3 receptor agonist dissociation rate constants (pkoff) and the KRI values (A), the A3 receptor agonist affinity (pKi) and the “kinetic KD” (pKD) (B), logarithm of association rate constants (logkon) (C) and negative logarithm of dissociation rate constants (pkoff) (D). Data used in these plots are detailed in Tables 2 and 3.
Figure 4.
Kinetic map (y axis: kon in M-1·s-1, x axis: koff in s-1) of all compounds that were kinetically characterized in this study. kon and koff values were obtained through competition association assays. The kinetically derived affinity (KD= koff/kon) is represented through diagonal parallel lines. The ribofurano derivatives are colored in blue, and the methanocarba derivatives are colored in red.
3.5 Structure–Affinity Relationships (SAR) and Structure–Kinetics Relationships (SKR) of hA3 receptor agonists
First, a series of hA3R agonists with either a natural or 5′-modified ribose moiety was investigated (Table 2). LUF5501-LUF5506 have an intact ribofurano moiety and a halogen substitution at the N6 phenyl group of the core scaffold. 3-Chlorophenyl substitution (LUF5501) provided somewhat better affinity than 3-bromophenyl (LUF5505) or 4-chlorophenyl (LUF5500) substitution (31 nM vs 78 nM; 31 nM vs 51 nM, respectively). The affinity of the 4-iodophenyl substituted derivative (LUF5506) was similar to LUF5501 (31 nM vs 29 nM). Meanwhile, their KRI values showed that halogen substitution at meta-position led to lower KRI values than those at para-position of the phenyl ring. The KRI values of LUF5501 and LUF5505 were at the lower end (< 0.7) with 0.64 and 0.65, respectively. The full-curve competition association experiments confirmed their similar fast dissociation rates (LUF5501: (1.6 ± 0.2) × 10-3 s-1; LUF5505: (1.1 ± 0.04) × 10-3 s-1). Worthy of note, LUF5501 proved to have the shortest RT (10 min) among the entire selection of compounds.
When the 5′-hydroxymethylene group (LUF5506) was substituted by an ethylcarboxamide (-CONHCH2CH3, LUF5521), affinity (14 nM vs 29 nM) and particularly the KRI increased (2.06 vs 0.99). The association and dissociation rates of LUF5521 were (1.9 ± 0.3) × 105 M-1s-1 and (1.4 ± 0.1) × 10-4 s-1, respectively, and its RT was calculated as 117 min.
We then examined benzyl substitutions at the N6 position of the core scaffold, with a methylcarboxamide (-CONHCH3) modification of the 5′-position on the ribofuran moiety (MECA, IB-MECA and 2-Cl-IBMECA). MECA had an affinity of 42 nM with a KRI value of 1.24. When iodine was introduced at the 3-position of the benzyl ring, yielding IB-MECA, affinity was strongly increased (2.9 nM vs 42 nM) as well as the KRI value (1.55 vs 1.24); with a further chlorine atom substitution at the C-2 position on the core scaffold (2-Cl-IBMECA) there was no further increase in affinity (3.5 nM vs 2.9 nM), but its KRI value increased significantly (2.02 vs 1.55), which is the largest value in this series (Table 2). For IB-MECA and 2-Cl-IBMECA, the association rate constants were similar ((5.9 ± 0.9) × 105 M-1s-1 vs (4.0 ± 0.5) × 105 M-1s-1), however 2-Cl-IBMECA had a slower dissociation rate ((7.2 ± 1.1) × 10-5 s-1 vs (1.8 ± 0.2) × 10-4 s-1) as had been immanent already from their KRI values. The RTs of IB-MECA and 2-Cl-IBMECA were calculated from their dissociation rate constants as 95 min and 231 min, respectively.
Interestingly, when the amide at the 5′-position on the ribofurano ring of 2-Cl-IBMECA was changed to an ether function (-CH2OC3H5, LUF5595), both association ((9.6 ± 3.1) × 105 M-1s-1 vs (4.0 ± 0.5) × 105 M-1s-1) and dissociation (2.3 ± 1.0) × 10-4 s-1 vs (7.2 ± 1.1) × 10-5 s-1) became faster, even though the compound was still a slowly dissociating agonist compared with the radioligand (KRI: 1.37).
Finally, the effect of the 2-Cl substitution was further confirmed in the comparison between LUF5586 and LUF5589. Their affinities were similar (2.8 nM vs 2.2 nM), but with 2-Cl, LUF5589 had a higher KRI than LUF5586 (1.66 vs 1.30), and the RT of LUF5589 was determined as 138 min. In addition, a larger substituent at the C-2 position (MRS7294) was tolerated with respect to both affinity and kinetics, at least in combination with a small N6-methyl substituent.
Table 3 summarizes our findings on the series of ring-rigidified methanocarba derivatives that maintain a receptor-preferred North (N) conformation, with the flexible ribose-substituted MRS7294 included once more to allow a comparison. MRS7294, MRS5980, MRS7140, MRS7158 and MRS7154 share a common fragment ((5-chlorothiophen-2-yl)ethynyl) at the C-2-position (R1 group). MRS7294, the agonist with unmodified ribofuran moiety, served as the starting of the analysis, which had both high affinity (3.7 nM) and a large KRI value (1.63). By competition association experiments its association and dissociation rates were determined as (8.2 ± 3.8) × 105 (M-1s-1) and (1.3 ± 0.2) × 10-4 s-1, respectively; its RT was calculated as 125 min. When the ribofurano ring was changed by the methanocarba moiety (MRS5980), the affinity increased to the sub-nanomolar range (0.72 nM vs 3.7 nM) and its KRI value also increased largely (1.97 vs 1.63). The full-curve competition association experiments showed that MRS5980's association rate was slightly faster ((9.0 ± 2.3) × 105 M-1s-1 vs (8.2 ± 3.8) × 105 M-1s-1 for the respective rate constants), but its dissociation rate was much slower ((4.0 ± 1.0) × 10-5 s-1 vs (1.3 ± 0.2) × 10-4 s-1) than its ribose-equivalent. The RT of MRS5980 was 417 min. When the nitrogen atom at the N-1-position was replaced by a carbon atom (MRS7140), the affinity remained high (5.2 nM), and its kinetics profile was not much altered compared with MRS5980 (kon: (3.2 ± 1.2) × 105 M-1s-1 vs (9.0 ± 2.3) × 105 M-1s-1; koff: (8.5 ± 0.3) × 10-5 s-1 vs (4.0 ± 1.0) × 10-5 s-1). Changing the mono-methyl substitution at the N6 position (MRS5980) for di-methyl substitution (MRS7158), severely compromised both affinity and KRI value (618 nM vs 0.72 nM; 0.93 vs 1.97). When mono-substitution at the N6 position (MRS7140) was extended to n-propyl (MRS7154), affinity remained high (2.2 nM), like the KRI value (1.67); both association and dissociation rates of MRS7154 were quite similar to MRS5980 (kon: (4.0 ± 0.7) × 105 M-1s-1 vs (9.0 ± 2.3) × 105 M-1s-1; koff: (6.2 ± 1.2) × 10-5 s-1 vs (4.0 ± 1.0) × 10-5 s-1), with RTs of 270 min and 417 min, respectively.
The methanocarba derivatives MRS3558, MRS5655, MRS5679 and MRS5698 share their R2 groups (3-chlorobenzyl substitution). When the 2-Cl substituent of MRS3558 was replaced by the bulky and rigid phenylethynyl (MRS5655), the affinity decreased from 1.0 nM (MRS3558) to 3.6 nM (MRS5655), while the KRI value increased, however (1.49 vs 2.58). In the competition association assays the association rate constants of MRS3558 and MRS5655 were similar ((5.7 ± 0.8) × 105 M-1s-1 vs (4.0 ± 0.8) × 105 M-1s-1), while the dissociation rate constants were 5.6-fold different ((1.0 ± 0.2) × 10-4 s-1 vs (1.8 ± 0.3) × 10-5 s-1), leading to a RT for MRS5655 of 909 min. When the phenylethynyl of MRS5655 was 3,4-di-fluorinated as in MRS5698, the affinity was reduced from 3.6 nM (MRS5655) to 9.4 nM (MRS5698), while the KRI value increased significantly from 2.58 (MRS5655) to 4.21 (MRS5698), with both slower association and dissociation rate constants (kon: (4.0 ± 0.8) × 105 M-1s-1 vs (1.3 ± 0.4) × 105 M-1s-1; koff: (1.8 ± 0.3) × 10-5 s-1 vs (8.5 ± 0.7) × 10-6 s-1), thus extending RT from 909 min to 1961 min. Furthermore, when the phenylethynyl fragment of MRS5655 was expanded to a 4-biphenylethynyl substitution on the R1 group (MRS5679), the affinity suffered from 3.6 nM (MRS5655) to 82 nM (MRS5679); however, its KRI value increased dramatically from 2.58 to 5.56. This high value rendered the determination of the kinetics profile of MRS5679 impossible. Obviously, from all these compounds we learned there can be a clear dichotomy between affinity and residence time.
When the benzyl substitutions at the N6 position (R2 in Table 3) were replaced by a simple monomethyl (i.e. MRS5655 to MRS5644; MRS5679 to MRS5667), their binding affinities to hA3 receptors were not compromised (MRS5655/MRS5644: 3.6 nM vs 2.4 nM) or even increased (MRS5679/MRS5667: 82 nM vs 4.6 nM), and so were the kinetics profiles of MRS5655 and MRS5644. The KRI values of MRS5679 and MRS5667 were both at the higher end (5.56 and 3.34).
4. Discussion
4.1 Effects of GTP on agonist binding to the human adenosine A3 receptor
In the current study, the binding interactions of unlabeled hA3 receptor agonists were determined in radioligand binding experiments with the reference tritiated antagonist [3H]PSB-11. The favorable binding characteristics of this radioligand served us particularly well in the competition association experiments where robust binding over time could still be measured in the presence of unlabeled ligands, essential for the calculation of the association and dissociation rate constants. It is well known that antagonists occupy a GPCR irrespective of whether it is coupled to a G protein, whereas agonists prefer the G protein-coupled state of the receptor [36]. Addition of GTP to the incubation mixture induces an uncoupling between receptor and available G protein, which leads to a lower apparent receptor affinity for agonists. We confirmed this behavior in our equilibrium displacement experiments, in which the two reference agonists IB-MECA and 2-Cl-IB-MECA had a 3-4-fold lower affinity in the presence of GTP (Fig. 1A, Table 1). Interestingly, all displacement curves had pseudo-Hill coefficients of approx. unity (Table 1), suggesting that the agonists recognized one receptor state in both the presence and absence of GTP, most likely a G protein-coupled state (no GTP) or an uncoupled state (with GTP). We then determined the agonist binding kinetics for the two states in subsequent competition association assays by measuring these in the absence or presence of GTP (Fig. 1B). Interestingly, the association rate constants were identical for the two states (Table 1). Apparently, the ligand, when approaching the receptor from the extracellular side, is insensitive to the coupling status at the intracellular G protein interface. However, the dissociation kinetics proved to be (significantly) different between the states, up to 8-fold for 2-Cl-IB-MECA (Table 1), suggesting that dissociating from the receptor is influenced by the absence or presence of the G protein. We then performed further experiments in the absence of GTP, as we felt that for the case of the hA3 receptor this experimental condition provides us information on the more relevant high-affinity agonist binding state of the receptor. The mathematical modeling provided by Motulsky and Mahan [32] does not allow the calculation of two receptor states with corresponding kinetic parameters. Hence, in a number of cases similar to ours it was decided to include GTP in the assays. For the adenosine A1 receptor, we included GTP in our assays, forcing the receptor to be in one lower affinity, G protein-uncoupled state only; with this restriction, the kinetic parameters of agonists were determined as conveniently as antagonists [37]. Likewise, the team of Charlton included GTP in kinetic assays on the muscarinic M3 receptor [38]. On the other hand, GTP was excluded in assays on the adenosine A2A receptor, as agonist affinity at this protein is insensitive to GTP in both equilibrium and kinetics assays [39].
4.2 Methodological aspects of the radioligand binding assays
To ensure an accurate and robust kinetic quantification, experiments were performed at 10 °C, as reported and documented previously [35]. Initially, a so-called dual-point competition association assay yielding KRI values for the hA3 receptor was performed as a “kinetic screening campaign” to increase throughput in comparison to the traditional, more elaborate, competition association experiments [35]. Although fast kinetics of agonist binding (e.g. LUF5501, Fig. 2A) was determined accurately at a relatively low temperature, the kinetics of one agonist with obviously very slow dissociation characteristics (MRS5679, Fig. 2B) could not be reliably determined with the Motulsky-Mahan equations. Meanwhile, its large KRI value (5.56) undoubtedly reflects its slow dissociation.
Besides the significant correlation between the agonists' KRI values and dissociation rate constants (Fig. 3A), the equilibrium Ki and kinetic KD values were also well correlated (Fig. 3B). Despite this correlation there are large discrepancies in values between the equilibrium affinities (Ki) and kinetic KD values. In general, the kinetic pKD values are higher than pKi values, with differences from 0.5 to 2.1 log-unit (LUF5501 and MRS5698), while the short RT agonists have smaller differences than long RT agonists. This phenomenon suggests that the affinities determined in the radioligand displacement studies may not reflect the true affinity of the compounds as many of them have residence times much longer than the (already long) incubation time of 240 min. In fact, this points to a general caveat in end-point assays in which the equilibrium characteristics of the probe, e.g., a radiolabeled or fluorescently tagged compound, determine the assay protocol without further consideration of the same characteristics of the unlabeled ligands to be tested.
Furthermore, there was a significant correlation between kon (k3) and affinity values (Ki) of the agonists, but no relationship was found between koff (k4) and Ki (Fig. 3C and 3D). Such correlation between affinity and on-rates has been reported in the case of other GPCRs [40] and ion channels [41, 42]. However, in the current study, variations in association rate constants were approximately 10-fold (logkon: 5.0 to 6.0), much smaller than at the other targets mentioned above. In addition, all the association rates were well below the diffusion limit of around 107 M-1s-1, which had been observed previously for membrane-bound proteins [43].
4.3 Ligand optimization based on Structure-Kinetics Relationships (SKRs)
All the agonists examined in the current study have been reported, and the riboside MRS7294 was most recently disclosed as a hA3 receptor agonist [28]. Most ribofurano derivatives (Table 2) had been synthesized and tested as early as 1994 [44]. The (N)-methanocarba derivatives (Table 3) were explored more recently, showing generally improved affinities over the earlier compounds [26-29]. Although most of these compounds have been described in the context of structure-affinity relationships (SARs), we reasoned that an extensive structure–kinetics relationships (SKRs) analysis is warranted since kinetic profiles are emerging indicators of in vivo functional efficacy[18, 20, 21, 45].
In the series of ribofurano derivatives (Table 2), the most outstanding agonist with respect to residence time was 2-Cl-IB-MECA (231 min). The underlying chemical features that prolonged the residence time in the ribofurano series are: i) a chlorine atom at the C2 position (R1 in Table 2, IB-MECA vs 2-Cl-IB-MECA, 95 min vs 231 min; LUF5586 vs LUF5589 in KRI: 1.30 vs 1.66), ii) a bulky meta-iodobenzyl substitution at the N6 position of the adenine nucleobase (R2 in Table 2, MECA vs IB-MECA in KRI: 1.24 vs 1.55), and iii) an amide modification at the 5′ position (R3 in Table 2, 2-Cl-IB-MECA vs LUF5595: 231 min vs 72 min; LUF5506 vs LUF5521 in KRI: 0.99 vs 2.06). Interestingly, slow dissociation kinetics of agonists can be maintained by introducing an extended linear side chain at the C2 position and removing the bulky substituent at the N6 position (i.e. N6-Me analogue MRS7294 in RT: 125 min). The kinetic behavior of another known reference agonist, 2-(1-hexynyl)-N-methyladenosine (HEMADO), also confirmed this observation from comparison with other research [45, 46].
More impressive kinetics were observed in the methanocarba series. From a previous study it was concluded that a possible H-bond interaction between the amide modification at the original 5′ position (-CONHCH3) and residues in the binding pocket of the hA3 receptor was a key feature of agonist kinetics [47]. Comparing the kinetics of ribofurano MRS7294 and its methanocarba equivalent MRS5980 provides evidence the conformational-constrained (N)-methanocarba moiety as a ribofurano ring substitute adds to the slow dissociation of MRS5980, suggesting this rigid ring system fits even better in the ligand binding pocket. Additionally, the N1 atom on the adenine core is contributing to slow dissociation (1-deza MRS7140 vs MRS5980 in RT: 196 min vs 417 min). The N6 position in the methanocarba series (R2 substituent) accepted a small methyl group as avidly as the larger n-propyl (MRS5980 vs MRS7154) and 3-chlorobenzyl substituent (MRS5644 vs MRS5655), with respect to both affinity and kinetics. Apparently, differences in lipophilicity between the N6-substituents did not seem to matter either. Data for the R1 (C2 position) substituent were already commented on in the Results section. It is noteworthy that the 3,4-difluorophenyl fragment contributed to a long residence time of MRS5698, quite similar to the 2,4-difluorophenyl fragment we encountered in A2A receptor antagonists displaying long residence time [48].
So far the human A3 receptor has not been crystallized yet, although its most homologous relative, the adenosine A1 receptor, has [49]. Homology models of the A3 receptor have been constructed, and their relevance has been recently reviewed and discussed [50]. It appeared that the interaction of the extended and rigid 2-arylethynyl groups at the C-2 position (as in e.g., MRS5679 and MRS5644) with the A3 receptor required an outward movement of TM2. Although the agonists' path of binding to or dissociation from the A3 receptor is unknown, one could speculate that such reorganization of TM2 “locks” the C2-substituted agonist. This reasoning stems from a molecular dynamics simulation and site-directed mutation study of the ligand dissociation pathway from the human adenosine A2A receptor [51], for which we used a high resolution crystal structure [52]. It taught us there may be a lid adjacent to the ligand binding pocket retaining the ligand for some time before the compound leaves the receptor. A follow-up study revealed that ligand dissociation correlated with the strength of the salt-bridge between His264 in EL3 and Glu169 in EL2 [53]. It would be interesting to learn whether a similar mechanism exists for the A3 receptor.
4.4 Retrospective evaluation of selected agonists with complete kinetic profiles
IB-MECA (RT = 95 min) and 2-Cl-IB-MECA (RT = 231 min) are two reference full agonists for the hA3 receptor. Despite numerous reports about their controversial cardioprotection [54, 55], they have been taken into a series of clinical trials: for IB-MECA, in total 12 completed or planned trials have been reported related to inflammatory conditions (e.g., keratoconjunctivitis sicca, rheumatoid arthritis, psoriasis, uveitis); for 2-Cl-IB-MECA, four trials have been registered for liver diseases (e.g., hepatocellular carcinoma, hepatitis C) [56, 57]. Intuitively one would expect that long residence time hA3 receptor agonists could be beneficial in chronic diseases, for instance to allow once daily dosing.
More recently, the neuroprotective effect provided by hA3 receptor agonists has been receiving attention [11, 58]. A number of agonists have been designed and evaluated both in in vitro and in vivo functional experiments, especially for the treatment of chronic neuropathic pain [26, 27, 29]. MRS5698 in particular, having the longest residence time (1961 min) from our research, has been studied extensively in various pre-clinical animal models of neuropathic pain [59-61]. We, however, hesitate to link the binding kinetics profile of MRS5698 with its in vivo effects, also because issues such as pharmacokinetics and species differences (human/rodent) may play an important but yet unknown role.
The adenosine A3 receptor has a peculiar and rapid desensitization/internalization profile in cultured cells [6]. However, the fast desensitization after A3 agonist exposure under in vitro conditions does not generalize to in vivo pain models. MRS5698 maintained full efficacy over a five-day period, with the drug administered either by daily injection or by an implanted mini-pump to provide a steady state plasma concentration [55]. Prolonged MRS5698 exposure in the rat did not reduce its efficacy, as would be expected from agonist-induced desensitization alone. Thus, additional studies of agonist occupancy of the receptor and subsequent receptor processing are warranted to further explore this interesting target for combatting neuropathic pain.
4.5 Kinetic Map
Using the association (kon) and dissociation (koff) rate constants obtained from competition association experiments (Tables 2 and 3), a kinetic map (Fig. 4) was constructed by plotting these values on the y-axis and x-axis, respectively. The dashed diagonal parallel lines represent the kinetically derived KD values (KD = koff/kon). Out of this map three subgroups emerged, which were divided according to koff values: Group A < Group B < Group C. Obviously, Group A and C are solely composed of methanocarba derivatives (Fig. 4, red) and ribofurano derivatives (Fig. 4, blue), respectively. Group B is a mixture of these two compound classes, with the methanocarba derivatives mainly displaying smaller koff values. Thus, rigidifying the ribose ring with the bicyclic group consistently prolonged the dissociation time. This general division indicates a different mode of receptor-ligand interaction during the binding and unbinding process of the two ligand groups. It also seems there is a clear residence-time “cliff” between Groups A (having one rigid C2-phenylethynyl or 4-biphenylethynyl group with or without one enlarged N6-benzyl group) and B (having multiple enlarged adenine substituents) in the kinetic map.
In each subgroup, agonists exhibit koff values in a similar range, but have different kon values. As a consequence, variation in KD values in each group was observed (Group A: ∼10-fold; Group B: ∼100-fold; Group C: ∼3-fold). Previous SKR studies have primarily focused on optimizing dissociation rates and residence times for designing a kinetically favorable ligand. Yet recently, there has been increasing acknowledgement of the important role association rate constants may play in determining the efficacy of a drug as the result of increased rebinding or increased drug-target selectivity [21, 62]. A kinetic map would thus allow for the selection of compounds with appropriate residence times while exploring the role of association rate constants.
In summary, an agonist-related competition association assay at the hA3 receptor was validated, and a series of ribofurano and methanocarba derivatives were kinetically profiled for the first time. A kon-koff-KD kinetic map was constructed and subsequently the agonists with complete kinetic profile were divided into three sub-groups based on their residence times. Longer residence times were associated with methanocarba (vs. ribose) and enlarged adenine N6 and C-2 (rigid arylalkynyl) substitutions. We identified agonists with very long residence times, but these may not be the most therapeutically favorable for treating various conditions in view of the fast desensitization of the receptor. Although far from definitive, this study suggests that proper residence times, not necessarily longer than target turnover time, may be vital parameters in the development of hA3R agonists for therapeutic use.
Acknowledgments
The research in this study has been performed in 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 further acknowledge support from the NIDDK Intramural Research Program and also thank Dr. Anna Junker for the fruitful discussions.
Footnotes
Compound list: IB-MECA (PubChem CID: 123683)
2-Cl-IB-MECA (PubChem CID: 3035850)
PSB-11 (PubChem CID: 9882625)
MRS5698 (PubChem CID: 57523213)
MRS3558 (PubChem CID: 11248240)
Conflict of interest: None.
List of author contributions: Participated in research design; Xia, IJzerman, Heitman.
Conducted experiments; Xia, Kyrizaki, van Duijl, Roorda.
Performed data analysis; Xia, Kyrizaki, van Duijl.
Wrote or contributed to the writing of the manuscript; Xia, Tosh, Jacobson, IJzerman.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Meyerhof W, Müller-Brechlin R, Richter D. Molecular cloning of a novel putative G-protein coupled receptor expressed during rat spermiogenesis. FEBS Lett. 1991;284(2):155–160. doi: 10.1016/0014-5793(91)80674-r. [DOI] [PubMed] [Google Scholar]
- 2.Cortés D, Guinzberg R, Villalobos-Molina R, Piña E. Evidence that endogenous inosine and adenosine-mediated hyperglycaemia during ischaemia–reperfusion through A3 adenosine receptors. Auton Autacoid Pharmacol. 2009;29(4):157–164. doi: 10.1111/j.1474-8665.2009.00443.x. [DOI] [PubMed] [Google Scholar]
- 3.Madi L, Ochaion A, Rath-Wolfson L, Bar-Yehuda S, Erlanger A, Ohana G, Harish A, Merimski O, Barer F, Fishman P. The A3 adenosine receptor is highly expressed in tumor versus normal cells: potential target for tumor growth inhibition. Clin Cancer Res. 2004;10(13):4472–4479. doi: 10.1158/1078-0432.CCR-03-0651. [DOI] [PubMed] [Google Scholar]
- 4.Ochaion A, Bar-Yehuda S, Cohen S, Barer F, Patoka R, Amital H, Reitblat T, Reitblat A, Ophir J, Konfino I, Chowers Y, Ben-Horin S, Fishman P. The anti-inflammatory target A3 adenosine receptor is over-expressed in rheumatoid arthritis, psoriasis and Crohn's disease. Cell Immunol. 2009;258(2):115–122. doi: 10.1016/j.cellimm.2009.03.020. [DOI] [PubMed] [Google Scholar]
- 5.Palmer TM, Stiles GL. Identification of threonine residues controlling the agonist-dependent phosphorylation and desensitization of the rat A3 adenosine receptor. Mol Pharmacol. 2000;57(3):539–545. [PubMed] [Google Scholar]
- 6.Klaasse EC, IJzerman AP, de Grip WJ, Beukers MW. Internalization and desensitization of adenosine receptors. Purinergic Signalling. 2008;4(1):21–37. doi: 10.1007/s11302-007-9086-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Gessi S, Merighi S, Varani K, Leung E, Mac Lennan S, Borea PA. The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacol Ther. 2008;117(1):123–140. doi: 10.1016/j.pharmthera.2007.09.002. [DOI] [PubMed] [Google Scholar]
- 8.Shamama N, Luqman AK, Zafar MA, Seemi FB. Adenosine A3 receptor: a promising therapeutic target in cardiovascular disease. Curr Cardiol Rev. 2016;12(1):18–26. doi: 10.2174/1573403X12666160111125116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Borea PA, Varani K, Vincenzi F, Baraldi PG, Tabrizi MA, Merighi S, Gessi S. The A3 adenosine receptor: history and perspectives. Pharmacol Rev. 2015;67(1):74–102. doi: 10.1124/pr.113.008540. [DOI] [PubMed] [Google Scholar]
- 10.Antonioli L, Csóka B, Fornai M, Colucci R, Kókai E, Blandizzi C, Haskó G. Adenosine and inflammation: what's new on the horizon? Drug Discovery Today. 2014;19(8):1051–1068. doi: 10.1016/j.drudis.2014.02.010. [DOI] [PubMed] [Google Scholar]
- 11.Fishman P, Bar-Yehuda S, Liang BT, Jacobson KA. Pharmacological and therapeutic effects of A3 adenosine receptor (A3AR) agonists. Drug Discovery Today. 2012;17(7-8):359–366. doi: 10.1016/j.drudis.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Müller CE, Jacobson KA. Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochim Biophys Acta, Biomembr. 2011;1808(5):1290–1308. doi: 10.1016/j.bbamem.2010.12.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Koscsó B, Csóka B, Pacher P, Haskó G. Investigational A3 adenosine receptor targeting agents. Expert Opin Invest Drugs. 2011;20(6):757–768. doi: 10.1517/13543784.2011.573785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Salvemini D, Jacobson KA. Highly selective A3 adenosine receptor agonists relieve chronic neuropathic pain. Expert Opin Ther Pat. 2017;27(8):967. doi: 10.1080/13543776.2017.1341018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fishman P, Cohen S. The A3 adenosine receptor (A3AR): therapeutic target and predictive biological marker in rheumatoid arthritis. Clin Rheumatol. 2016;35(9):2359–2362. doi: 10.1007/s10067-016-3202-4. [DOI] [PubMed] [Google Scholar]
- 16.Cohen S, Stemmer SM, Zozulya G, Ochaion A, Patoka R, Barer F, Bar-Yehuda S, Rath-Wolfson L, Jacobson KA, Fishman P. CF102 an A3 adenosine receptor agonist mediates anti-tumor and anti-inflammatory effects in the liver. J Cell Physiol. 2011;226(9):2438–2447. doi: 10.1002/jcp.22593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Jacobson KA, Merighi S, Varani K, Borea PA, Baraldi S, Aghazadeh Tabrizi M, Romagnoli R, Baraldi PG, Ciancetta A, Tosh DK, Gao ZG, Gessi S. A3 adenosine receptors as modulators of inflammation: from medicinal chemistry to therapy. Med Res Rev. doi: 10.1002/med.21456. 10.1002/med.21456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Guo D, Hillger JM, IJzerman AP, Heitman LH. Drug-target residence time—a case for G protein-coupled receptors. Med Res Rev. 2014;34(4):856–892. doi: 10.1002/med.21307. [DOI] [PubMed] [Google Scholar]
- 19.Cusack KP, Wang Y, Hoemann MZ, Marjanovic J, Heym RG, Vasudevan A. Design strategies to address kinetics of drug binding and residence time. Bioorg Med Chem Lett. 2015;25(10):2019–2027. doi: 10.1016/j.bmcl.2015.02.027. [DOI] [PubMed] [Google Scholar]
- 20.Copeland RA, Pompliano DL, Meek TD. Drug-target residence time and its implications for lead optimization. Nat Rev Drug Discovery. 2006;5(9):730–739. doi: 10.1038/nrd2082. [DOI] [PubMed] [Google Scholar]
- 21.Copeland RA. The drug-target residence time model: a 10-year retrospective. Nat Rev Drug Discovery. 2016;15(2):87–95. doi: 10.1038/nrd.2015.18. [DOI] [PubMed] [Google Scholar]
- 22.Heitman LH, Göblyös A, Zweemer AM, Bakker R, Mulder-Krieger T, van Veldhoven JPD, de Vries H, Brussee J, IJzerman AP. A series of 2,4-disubstituted quinolines as a new class of allosteric enhancers of the adenosine A3 receptor. J Med Chem. 2009;52(4):926–931. doi: 10.1021/jm8014052. [DOI] [PubMed] [Google Scholar]
- 23.Gessi S, Varani K, Merighi S, Morelli A, Ferrari D, Leung E, Baraldi PG, Spalluto G, Borea PA. Pharmacological and biochemical characterization of A3 adenosine receptors in Jurkat T cells. Br J Pharmacol. 2001;134(1):116–126. doi: 10.1038/sj.bjp.0704254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.de Zwart M, de Groote M, van der Klein PAM, van Dun S, Bronsing R, von Frijtag Drabbe Künzel JK, IJzerman AP. Phenyl-substituted N6-phenyladenosines and N6-phenyl-5 ′ -N-ethylcarboxamidoadenosines with high activity at human adenosine A2B receptors. Drug Dev Res. 2000;49(2):85–93. [Google Scholar]
- 25.van Tilburg EW, van der Klein PAM, von Frijtag Drabbe Künzel J, de Groote M, Stannek C, Lorenzen A, IJzerman AP. 5′-O-alkyl ethers of N,2-substituted adenosine derivatives: partial agonists for the adenosine A1 and A3 receptors. J Med Chem. 2001;44(18):2966–2975. doi: 10.1021/jm001114o. [DOI] [PubMed] [Google Scholar]
- 26.Tosh DK, Finley A, Paoletta S, Moss SM, Gao ZG, Gizewski ET, Auchampach JA, Salvemini D, Jacobson KA. In vivo phenotypic screening for treating chronic neuropathic pain: modification of C2-arylethynyl group of conformationally constrained A3 adenosine receptor agonists. J Med Chem. 2014;57(23):9901–9914. doi: 10.1021/jm501021n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tosh DK, Crane S, Chen Z, Paoletta S, Gao ZG, Gizewski E, Auchampach JA, Salvemini D, Jacobson KA. Rigidified A3 adenosine receptor agonists: 1-deazaadenine modification maintains high in vivo efficacy. ACS Med Chem Lett. 2015;6(7):804–808. doi: 10.1021/acsmedchemlett.5b00150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tosh DK, Janowsky A, Eshleman AJ, Warnick E, Gao ZG, Chen Z, Gizewski E, Auchampach JA, Salvemini D, Jacobson KA. Scaffold repurposing of nucleosides (adenosine receptor agonists): enhanced activity at the human dopamine and norepinephrine sodium symporters. J Med Chem. 2017;60(7):3109–3123. doi: 10.1021/acs.jmedchem.7b00141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tosh DK, Deflorian F, Phan K, Gao ZG, Wan TC, Gizewski E, Auchampach JA, Jacobson KA. Structure-guided design of A3 adenosine receptor-selective nucleosides: combination of 2-arylethynyl and bicyclo[3.1.0]hexane substitutions. J Med Chem. 2012;55(10):4847–4860. doi: 10.1021/jm300396n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tchilibon S, Joshi BV, Kim SK, Duong HT, Gao ZG, Jacobson KA. (N)-methanocarba 2,N6-disubstituted adenine nucleosides as highly potent and selective A3 adenosine receptor agonists. J Med Chem. 2005;48(6):1745–1758. doi: 10.1021/jm049580r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Smith PK, Krohn RI, Hermanson GT, Mallia AK, Gartner FH, Provenzano MD, Fujimoto EK, Goeke NM, Olson BJ, Klenk DC. Measurement of protein using bicinchoninic acid. Anal Biochem. 1985;150(1):76–85. doi: 10.1016/0003-2697(85)90442-7. [DOI] [PubMed] [Google Scholar]
- 32.Motulsky HJ, Mahan LC. The kinetics of competitive radioligand binding predicted by the law of mass action. Mol Pharmacol. 1984;25(1):1–9. [PubMed] [Google Scholar]
- 33.Guo D, van Dorp EJH, Mulder-Krieger T, van Veldhoven JPD, Brussee J, IJzerman AP, Heitman LH. Dual-point competition association assay: a fast and high-throughput kinetic screening method for assessing ligand-receptor binding kinetics. J Biomol Screening. 2013;18(3):309–320. doi: 10.1177/1087057112464776. [DOI] [PubMed] [Google Scholar]
- 34.Cheng YC, Prusoff WH. Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50 per cent inhibition (IC50) of an enzymatic reaction. Biochem Pharmacol. 1973;22(23):3099–3108. doi: 10.1016/0006-2952(73)90196-2. [DOI] [PubMed] [Google Scholar]
- 35.Xia L, Burger WAC, van Veldhoven JPD, Kuiper BJ, van Duijl TT, Lenselink EB, Paasman E, Heitman LH, IJzerman AP. Structure–affinity relationships and structure–kinetics relationships of pyrido[2,1-f]purine-2,4-dione derivatives as human adenosine A3 receptor antagonists. J Med Chem. 2017;60(17):7555–7568. doi: 10.1021/acs.jmedchem.7b00950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sprang SR. Cell signalling: binding the receptor at both ends. Nature. 2011;469(7329):172–173. doi: 10.1038/469172a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Xia L, de Vries H, IJzerman AP, Heitman LH. Scintillation proximity assay (SPA) as a new approach to determine a ligand's kinetic profile. A case in point for the adenosine A1 receptor. Purinergic Signalling. 2016;12(1):115–126. doi: 10.1007/s11302-015-9485-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sykes DA, Dowling MR, Charlton SJ. Exploring the mechanism of agonist efficacy: a relationship between efficacy and agonist dissociation rate at the muscarinic M3 receptor. Mol Pharmacol. 2009;76(3):543–551. doi: 10.1124/mol.108.054452. [DOI] [PubMed] [Google Scholar]
- 39.Guo D, Mulder-Krieger T, IJzerman AP, Heitman LH. Functional efficacy of adenosine A2A receptor agonists is positively correlated to their receptor residence time. Br J Pharmacol. 2012;166(6):1846–1859. doi: 10.1111/j.1476-5381.2012.01897.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Doornbos MLJ, Cid JM, Haubrich J, Nunes A, van de Sande JW, Vermond SC, Mulder-Krieger T, Trabanco AA, Ahnaou A, Drinkenburg WH, Lavreysen H, Heitman LH, IJzerman AP, Tresadern G. Discovery and kinetic profiling of 7-aryl-1,2,4-triazolo[4,3-a]pyridines: positive allosteric modulators of the metabotropic glutamate receptor 2. J Med Chem. 2017;60(15):6704–6720. doi: 10.1021/acs.jmedchem.7b00669. [DOI] [PubMed] [Google Scholar]
- 41.Yu Z, van Veldhoven JPD, Louvel J, 't Hart IME, Rook MB, van der Heyden MAG, Heitman LH, IJzerman AP. Structure–affinity relationships (SARs) and structure–kinetics relationships (SKRs) of Kv11.1 blockers. J Med Chem. 2015;58(15):5916–5929. doi: 10.1021/acs.jmedchem.5b00518. [DOI] [PubMed] [Google Scholar]
- 42.Yu Z, IJzerman AP, Heitman LH. K(v)11.1 (hERG)-induced cardiotoxicity: a molecular insight from a binding kinetics study of prototypical K(v)11.1 (hERG) inhibitors. Br J Pharmacol. 2015;172(3):940–955. doi: 10.1111/bph.12967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Smith GF. Medicinal chemistry by the numbers: the physicochemistry, thermodynamics and kinetics of modern drug design. In: Lawton G, Witty DR, editors. Progress in Medicinal Chemistry. Elsevier B.V; Burlington: 2009. p. 1. [DOI] [PubMed] [Google Scholar]
- 44.Gallo-Rodriguez C, Ji Xd, Melman N, Siegman BD, Sanders LH, Orlina J, Fischer B, Pu Q, Olah ME, van Galen PJM, Stiles GL, Jacobson KA. Structure – activity relationships of N6-benzyladenosine-5′-uronamides as A3-selective adenosine agonists. J Med Chem. 1994;37(5):636–646. doi: 10.1021/jm00031a014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Guo D, Heitman LH, IJzerman AP. Kinetic aspects of the interaction between ligand and G protein-coupled receptor: the case of the adenosine receptors. Chem Rev. 2017;117(1):38–66. doi: 10.1021/acs.chemrev.6b00025. [DOI] [PubMed] [Google Scholar]
- 46.Klotz KN, Falgner N, Kachler S, Lambertucci C, Vittori S, Volpini R, Cristalli G. [3H]HEMADO— a novel tritiated agonist selective for the human adenosine A3 receptor. Eur J Pharmacol. 2007;556(1):14–18. doi: 10.1016/j.ejphar.2006.10.048. [DOI] [PubMed] [Google Scholar]
- 47.Auchampach JA, Gizewski ET, Wan TC, de Castro S, Brown GG, Jacobson KA. Synthesis and pharmacological characterization of [125I]MRS5127, a high affinity, selective agonist radioligand for the A3 adenosine receptor. Biochem Pharmacol. 2010;79(7):967–973. doi: 10.1016/j.bcp.2009.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Guo D, Xia L, van Veldhoven JPD, Hazeu M, Mocking T, Brussee J, IJzerman AP, Heitman LH. Binding kinetics of ZM241385 derivatives at the human adenosine A2A receptor. ChemMedChem. 2014;9(4):752–761. doi: 10.1002/cmdc.201300474. [DOI] [PubMed] [Google Scholar]
- 49.Glukhova A, Thal DM, Nguyen AT, Vecchio EA, Jörg M, Scammells PJ, May LT, Sexton PM, Christopoulos A. Structure of the adenosine A1 receptor reveals the basis for subtype selectivity. Cell. 2017;168(5):867–877. doi: 10.1016/j.cell.2017.01.042. [DOI] [PubMed] [Google Scholar]
- 50.Ciancetta A, Jacobson K. Structural Probing and Molecular Modeling of the A3 Adenosine Receptor: A Focus on Agonist Binding. Molecules. 2017;22(3):449–466. doi: 10.3390/molecules22030449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Guo D, Pan AC, Dror RO, Mocking T, Liu R, Heitman LH, Shaw DE, IJzerman AP. Molecular basis of ligand dissociation from the adenosine A2A receptor. Mol Pharmacol. 2016;89(5):485–491. doi: 10.1124/mol.115.102657. [DOI] [PubMed] [Google Scholar]
- 52.Liu W, Chun E, Thompson AA, Chubukov P, Xu F, Katritch V, Han GW, Roth CB, Heitman LH, IJzerman AP, Cherezov V, Stevens RC. Structural basis for allosteric regulation of GPCRs by sodium ions. Science. 2012;337(6091):232–236. doi: 10.1126/science.1219218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Segala E, Guo D, Cheng RKY, Bortolato A, Deflorian F, Doré AS, Errey JC, Heitman LH, Ijzerman AP, Marshall FH, Cooke RM. Controlling the Dissociation of Ligands from the Adenosine A2A Receptor through Modulation of Salt Bridge Strength. J Med Chem. 2016;59(13):6470–6479. doi: 10.1021/acs.jmedchem.6b00653. [DOI] [PubMed] [Google Scholar]
- 54.Xu Z, Jang Y, Mueller RA, Norfleet EA. IB-MECA and cardioprotection. Cardiovasc Drug Rev. 2006;24(3-4):227–238. doi: 10.1111/j.1527-3466.2006.00227.x. [DOI] [PubMed] [Google Scholar]
- 55.Ge ZD, Peart JN, Kreckler LM, Wan TC, Jacobson MA, Gross GJ, Auchampach JA. Cl-IB-MECA [2-chloro-N6-(3-iodobenzyl)adenosine-5′-N-methylcarboxamide] reduces ischemia/reperfusion injury in mice by activating the A3 adenosine receptor. J Pharmacol Exp Ther. 2006;319(3):1200–1210. doi: 10.1124/jpet.106.111351. [DOI] [PubMed] [Google Scholar]
- 56.National center for biotechnology information. Pubchem compound database; CID=123683. [accessed June 30, 2017]; https://pubchem.ncbi.nlm.nih.gov/compound/123683.
- 57.National center for biotechnology information. PubChem compound database; CID=3035850. [accessed June 30, 2017]; https://pubchem.ncbi.nlm.nih.gov/compound/3035850.
- 58.Chen Z, Janes K, Chen C, Doyle T, Bryant L, Tosh DK, Jacobson KA, Salvemini D. Controlling murine and rat chronic pain through A3 adenosine receptor activation. FASEB J. 2012;26(5):1855–1865. doi: 10.1096/fj.11-201541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Carlin JL, Tosh DK, Xiao C, Piñol RA, Chen Z, Salvemini D, Gavrilova O, Jacobson KA, Reitman ML. Peripheral adenosine A3 receptor activation causes regulated hypothermia in mice that is dependent on central histamine H1 receptors. J Pharmacol Exp Ther. 2016;356(2):474–482. doi: 10.1124/jpet.115.229872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Little JW, Ford A, Symons-Liguori AM, Chen Z, Janes K, Doyle T, Xie J, Luongo L, Tosh DK, Maione S, Bannister K, Dickenson AH, Vanderah TW, Porreca F, Jacobson KA, Salvemini D. Endogenous adenosine A3 receptor activation selectively alleviates persistent pain states. Brain. 2015;138(1):28–35. doi: 10.1093/brain/awu330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Ford A, Castonguay A, Cottet M, Little JW, Chen Z, Symons-Liguori AM, Doyle T, Egan TM, Vanderah TW, De Konnick Y, Tosh DK, Jacobson KA, Salvemini D. Engagement of the GABA to KCC2 signaling pathway contributes to the analgesic effects of A3AR agonists in neuropathic pain. J Neurosci. 2015;35(15):6057–6067. doi: 10.1523/JNEUROSCI.4495-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.de Witte WEA, Danhof M, van der Graaf PH, de Lange ECM. In vivo target residence time and kinetic selectivity: the association rate constant as determinant. Trends Pharmacol Sci. 2016;37(10):831–842. doi: 10.1016/j.tips.2016.06.008. [DOI] [PubMed] [Google Scholar]






