Next Article in Journal
Mapping miRNA Research in Schizophrenia: A Scientometric Review
Next Article in Special Issue
Aberrant Dopamine System Function in the Ferrous Amyloid Buthionine (FAB) Rat Model of Alzheimer’s Disease
Previous Article in Journal
In Vivo Efficacy of Wound Healing under External (Bio)AgNCs Treatment: Localization Case Study in Liver and Blood Tissue
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Design and Synthesis of Conformationally Flexible Scaffold as Bitopic Ligands for Potent D3-Selective Antagonists

1
Vagelos Laboratories, Department of Radiology, University of Pennsylvania, 1012, 231 S. 34th Street, Philadelphia, PA 19104, USA
2
Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 432; https://doi.org/10.3390/ijms24010432
Submission received: 2 December 2022 / Revised: 22 December 2022 / Accepted: 24 December 2022 / Published: 27 December 2022

Abstract

:
Previous studies have confirmed that the binding of D3 receptor antagonists is competitively inhibited by endogenous dopamine despite excellent binding affinity for D3 receptors. This result urges the development of an alternative scaffold that is capable of competing with dopamine for binding to the D3 receptor. Herein, an SAR study was conducted on metoclopramide that incorporated a flexible scaffold for interaction with the secondary binding site of the D3 receptor. The alteration of benzamide substituents and secondary binding fragments with aryl carboxamides resulted in excellent D3 receptor affinities (Ki = 0.8–13.2 nM) with subtype selectivity to the D2 receptor ranging from 22- to 180-fold. The β-arrestin recruitment assay revealed that 21c with 4-(pyridine-4-yl)benzamide can compete well against dopamine with the highest potency (IC50 = 1.3 nM). Computational studies demonstrated that the high potency of 21c and its analogs was the result of interactions with the secondary binding site of the D3 receptor. These compounds also displayed minimal effects for other GPCRs except moderate affinity for 5-HT3 receptors and TSPO. The results of this study revealed that a new class of selective D3 receptor antagonists should be useful in behavioral pharmacology studies and as lead compounds for PET radiotracer development.

1. Introduction

Targeting D2 and D3 receptors has been studied for the treatment of neuropsychiatric disorders such as schizophrenia, and substance use disorders and addiction [1,2,3,4]. However, preferential localization of D3 receptors in limbic regions of the human brain suggested that D3 receptors may be a suitable target for developing therapeutics for treating neuropsychiatric disorders [5,6]. Other studies have demonstrated that this receptor plays a role in mediating the motivational actions of psychostimulants such as cocaine and amphetamine, and D3 antagonists have shown great promise in blocking cocaine self-administration in rodents and nonhuman primates [7,8]. The recent observation in the treatment of opioid use disorder has accelerated the need for the clinical evaluation of drugs targeting D3 receptors [9,10,11].
The development of dopamine D3-selective ligands continues to be a challenging area of medicinal chemistry research due to the high sequence homology of D2 and D3 receptor within the transmembrane (TM) domains (~79%) [12]. For developing receptor subtype selectivity, a “bitopic ligand” design has proven to be effective in the development of D3-selective compounds [13,14]. In this approach, a protonated basic amine in different scaffolds forms a salt bridge with Asp1103.32 of the D3 receptor in the orthosteric binding site (OBS), which is important for high binding affinity and the potency [15]. A secondary pharmacophore having an aromatic ring and appropriate linker group can result in high selectivity for the D3 receptor by the interaction with the secondary binding site (SBS) [16,17,18].
The first D3-selective scaffold contained an N-aryl piperazine moiety as the orthosteric binding fragment and an aryl carboxamide moiety with an alkyl linker as a secondary binding fragment [16,19,20,21,22,23,24,25]. This scaffold exhibited a sub nM binding affinity and good subtype selectivity for D3 receptors versus D2 receptors. However, these ligands also exhibited high binding affinity for other GPCRs (e.g., 5-HT or adrenergic receptors), which may lead the unwanted side effects [26,27,28]. Moreover, the in vivo properties of radiolabeled versions of this scaffold were not useful as PET radiotracers since they could not compete with endogenous dopamine for binding to the D3 receptor in vivo [21,29]. Since the replacement of substituents on benzamide or secondary binding fragments did not result in a significant change in properties of the N-aryl piperazine congeners, many groups have pursued other scaffolds, including azabicyclo [3.1.0]hexane [30,31,32], azaspiro alkane [33], diazaspiro alkane [34], tranylcypromine [35], or phenylcyclopropylmethylamine (PCPMA) [36]). However, these can be limited for clinical use under certain circumstances due to the poor bioavailability or toxicity [37,38,39] or are still under investigation. Recently, D2/D3 receptor agonist- and antagonist-modified bitopic ligands were developed based on (+)-PD128,907 or PF-592379 for selective agonist [40] and eticlopride for D2/D3 receptor ligands [41]. These compounds had comparatively low selectivity for D3 versus D2 receptors.
In the current study, we designed a new class of D3 receptor antagonist having the conformationally flexible scaffold of metoclopramide and the eticlopride-based benzamides (e.g., [18F]fallypride, Ki D2 = 0.02 nM and D3 = 0.19 nM [42,43], IC50 = 1.7 nM [29]; [11C]FLB457, Ki = 0.02 nM for D2/D3 receptors [44]) as lead compounds. Metoclopramide is largely used as an antiemetic; however, this compound also exhibited the low affinity for mixed D2/D3 receptors with the orthosteric binding fragment [45]. A combination of this flexible scaffold with well-established primary pharmocophore of the eticlopride-based benzamides was expected to achieve the high binding affinity and the potency for D3 receptors. Since the basic amine in this scaffold is structurally flexible without ring strain, the secondary binding fragment can be extended to strongly interact with the SBS while the orhosteric binding fragment remains bound to the OBS. Comprehensive screening was investigated for off-target interactions with other GPCRs; computational studies were also performed to provide the rational for the excellent potency of developed D3 receptor antagonists.

2. Results

2.1. Chemistry

Synthesis of 3-fluoropropyl or bromo analogs which have a dimethyl tert-amine with different length of carbon linker is shown in Scheme 1. 5-(3-Fluoropropyl)-2,3-dimethoxybenzoic acid (1a) or 5-bromo-2,3-dimethoxybenzoic acid (1b) was conjugated with secondary amine Boc-protected tert-butyl (2-aminoethyl)(propyl)carbamate by amide coupling in a quantitative yield. After the removal of the Boc protecting group, free amine 3a or 3b was N-alkylated with 2-(3-bromopropyl) or 2-(4-bromobutyl)-1,3-dioxolane. Dioxolanes 4a to 4d were hydrolyzed using aqueous 4 N HCl at RT to give the aldehyde 5a to 5d which were conjugated with dimethylamine via reductive amination. 5-(3-Fluoropropyl) or 5-bromo-2,3-dimethoxybenzamide analogs having the dimethylamine moiety (6ad) were obtained in 37–47% yield over the two-step synthesis.
The next series focused on preparing analogs having a spacer group with an aromatic ring system for interacting with the SBS. For the aromatic ring moiety, we tested 4-(thiophen-2-yl)benzamide or 4-methyl-5-phenyl-4H-1,2,4-triazole-3-thiol. The 4-(Thiophen-2-yl)benzamide fragment was chosen from our previous results. This aromatic ring system was observed in LS-3-134 and other structural congeners having a high D3 affinity and excellent selectivity versus the D2 receptor [46,47,48]. 3b was N-alkylated with N-(3-bromopropyl) or N-(4-bromobutyl)phthalimide and then the protecting phthalimide 7a or 7b was hydrolyzed using hydrazine hydrate by heating for 3 h to give primary amine 8a or 8b (Scheme 2). 4-(Thiophen-2-yl)benzoic acid was converted to the corresponding acyl chloride using thionyl chloride at RT followed by treatment with 8a or 8b to give 9a or 9b in 50% or 20% yield, respectively. The triazole-thiol ether analogs were prepared by reduction of 5c and 5d to give alcohols 10a and 10b, which were converted to 11a and 11b using Mitsunobu reaction. The desired products 11a or 11b were obtained in 16% and 28% yield, respectively (Scheme 2).
Inspection of the structure of 9a reveals that two different benzamide fragments which share the tert-amine are capable of interacting with the OBS of the D2 and D3 receptors. Therefore, fragments 12 and 14 were synthesized for evaluation in in vitro binding studies (Scheme 3). 12 was synthesized by N-methylation from the secondary amine 3b in 19% yield. For 14, 4-(thiophen-2-yl)benzoic acid was conjugated with 3-bromopropylamine through acyl chlorination followed by N-alkylated with N-methylethanamine.
The next series probed the size of substituents on the tert-amine group. The pendent synthons for allyl (15a) and 4-fluorobenzyl (15b) were prepared from ethylenediamine (Scheme 4). For the synthesis of 15a, one of the primary amines was protected with a trifluoroacetyl group and the other primary amine alkylated with allyl bromide. The secondary amine was protected as a N-Boc and the trifluoroacetyl group was removed. 15b was synthesized in a similar method with 15a except a reductive amination with 4-fluorobenzaldehyde was used. The prepared synthon 15a or 15b was conjugated with 1b, and the N-Boc was removed to give intermediates 17a,b. These intermediates were treated with N-propylphthalimide to give 18a,b. Removal of the phthalimide group with hydrazine hydrate gave corresponding N-propyl intermediate 19a (via reduction of the N-allyl group) and the 4-fluorobenzyl analog 19b. The intermediates 19a and 19b were conjugated with 4-(thiophen-2-yl)benzoic acid to give the desired products 20a and 20c in 71% or 32% yield, respectively. For the N-allyl analog, 17a was directly N-alkylated with 13 to give 20b in 21% yield.
To investigate the nature of the aromatic moiety for binding to the SBS, aryl carboxamides 21bj were synthesized using the same method described for the synthesis of 9a but using different aryl carboxylic acids and the naphthamide 21a was synthesized using 2-naphthoyl chloride using in the basic condition (Scheme 5). The desired benzamide analogs were obtained in yields ranging from 20 to 86%, respectively. The purity of all investigated compounds was confirmed prior to analysis and was greater than 95% on a 2695 Alliance LC-MS (Supplemental Table S1).

2.2. SAR Study

Two different assays were used to evaluate the properties of the analogs described above. The receptor binding affinity was measured by radioligand binding assays using [125I]IABN with D2 or D3 receptors highly expressed HEK293 cells [49]. The functional activity of the analogs was determined using a β-arrestin recruitment assay. The assay was initially conducted in agonist binding mode to confirm that they function as antagonists at the D3 receptor. Once this efficacy was confirmed, the assay was conducted in antagonist mode to determine the ability of the antagonist to compete with dopamine at the D3 receptor. The results of the antagonist mode assay are reported as IC50 values [50,51,52]. Imax values were individually calculated from the assay and reliable with over 50% inhibition.
The first series of compounds evaluated were those synthesized in Scheme 1 and Scheme 2 (Table 1). The dimethyl amino analogs 6ad displayed a relatively low binding affinity for both D2 and D3 receptors. These data suggest that a basic amine moiety in the spacer group reduces affinity at both receptors. The observation that 6d had a 10-fold higher affinity than its structural congener 6b indicates that the Br-substituent is more preferred in the OBS than the corresponding fluoropropyl substituent. Compounds 9a,b and 11a,b, which have aromatic groups in the SBS, displayed a higher affinity at both D2 and D3 receptors. The 4-(thiophen-2-yl)benzamide analogs were more potent at the D3 receptor than the corresponding 4-methyl-5-phenyl-4H-1,2,4-triazole-3-thiol analogs. These data suggest that benzamides are preferred in the SBS of the D3 receptor for this scaffold. It is of interest to note that 9a had ~170-fold higher affinity at the D3 versus the D2 receptor.
It is important to note that 9a has two different modes in which it can bind to the D3 receptor. The first mode has the bromobenzamide moiety binding to the OBS and the 4-(thiophen-2-yl)benzamide binding to the SBS. The second mode has the 4-(thiophen-2-yl)benzamide binding to the OBS and the bromobenzamide moiety binding to the SBS. In vitro binding studies revealed that fragment 12 showed non-selectively high Ki values at both of dopamine receptor subtypes (Ki D2 = 89.2 ± 5.6 nM, D3 = 21.8 ± 5.1 nM), whereas 14 did not show any binding affinity at D2 and D3 receptors (Ki D2 > 1000 nM and D3 > 1000 nM). Moreover, the β-arrestin recruitment assay indicated that compound 18 is very potent for the D3 receptor (IC50 = 4.6 ± 1.2 nM). These data are consistent with the first mode that the bromobenzamide moiety binds to the OBS and the 4-(thiophen-2-yl)benzamide binds to the SBS.
Table 2 shows the effect of the size of the N-alkyl group in the tert-amine on the D2 and D3 receptor binding. Our results indicate that the N-ethyl substituent 9a showed the highest binding affinity and subtype selectivity at the D3 receptor versus the D2 receptor. There was a slight decrease in affinity in going from propyl to allyl groups, whereas the 4-fluorobenzyl group resulted in a large loss in affinity at both D2 and D3 receptors (Table 2). When the size of substituents was increased, the binding affinity and subtype selectivity was decreased. This reduction in affinity also translated to the β-arrestin recruitment assay. That is, there was a trend of decreased potency in the order of 9a (IC50 = 14.0 ± 7.4 nM) > 20a (IC50 = 26.5 ± 12.9 nM) > 20b (IC50 = 51.6 ± 40.8 nM). Based on this SAR study, the N-ethyl group is the preferred alkyl group with respect to binding to the OBS.
A number of compounds were prepared to explore the nature of the interaction between the aromatic ring and the SBS. Previous studies with the N-aryl piperazine analogs revealed that a wide range of aromatic rings are tolerated in the SBS with respect to D3 affinity, but the overall D3 versus D2 selectivity can be influenced by the nature of this interaction. The results of this study are shown in Table 3. All compounds had good affinity at D3 receptors, with Ki values ranging between 0.8 and 13.2 nM. The D2 affinities ranged between 107 and 525 nM, resulting in a D3 selectivity ratio (i.e., D2/D3 ratio) ranging from 22.1- to 180-fold. The effect of the nature of the aromatic ring in the SBS on the ability of the antagonist to compete with dopamine in the β-arrestin assay was somewhat unexpected. For example, both 21a and 21c have ~1 nM affinity for the D3 receptor in the radioligand binding assay, but the potency of 21c in the β-arrestin recruitment assay was 10-fold higher than that of 21a (IC50 = 1.3 vs. 16.4 nM).

2.3. Molecular Docking and Molecular Dynamics Simulations (MDS)

To understand the favorable binding profiles of the metoclopramide analogs, molecular docking and MDS studies were performed using different N-alkyl compounds (9a, 20a, 20b, 20c and 21c) with the D3 receptor (PDB: 3PBL) (Table 4). These compounds were chosen because they are close structural analogs and have a wide range in D3 receptor affinity (1–300 nM). As reported in previous studies [13,29,53], the binding pose that formed a bridge hydrogen bond between the carboxylate of ASP1103.32 and the protonated nitrogen was considered to be critical for high binding affinity for the D3 receptor. The distance between the protonated nitrogen ranged between 2.6 and 2.9 Å, and 9a was found to have the closest interaction (2.6 Å). The estimated binding energies were not significantly different for each compound (−9.74 to −10.22 kcal/mol). Therefore, the difference in D3 affinity of the five compounds cannot be explained by the distance between ASP1103.32 and the protonated nitrogen atom, and the calculated binding energies from docking studies.
In MDS studies, the root mean square distance (RMSD) was calculated over 50–200 ns in five copies of the MDS production (Table 4). The first time frame (0 ns) of the production run was used as the reference position to determine the stability of each compound in the binding site. 21c presented the lowest standard deviation of RMSD (2.45 ± 0.49 Å) indicating the least amount of movement in the binding site. A relatively higher amount of motion (3.00 ± 0.82 Å) with 20c is consistent with the lower binding affinity for D3 receptors. These results indicate the MDS studies correlate better with D3 affinity than the results of docking studies.
The representative binding pose of the MDS production run is displayed in Figure 1. Within the OBS of the D3 receptor, all the selected compounds were engaged in multiple interactions. The hydrogen bond with ASP1103.32 and π staking interactions with PHE3456.52 were observed with all five compounds. However, a halogen bond between VAL1895.39 and the bromine of the 5-bromo-2,3-dimethoxybenzene moiety was observed for 9a, 21c, and 20c (Figure 1a,b,e, respectively). It is of interest to note the 21c, the most potent compound in the β-arrestin recruitment assay, which predicts the ability to compete with endogenous dopamine, had a cation–π interaction between the protonated nitrogen and PHE1063.28 residue (Figure 1b).
The summary of overall frequency of contacts from the MDS studies, including hydrophobic interactions, hydrogen bonds, the salt bridge, halogen bonds, and π-interactions, is shown in Figure 2. All five compounds formed stable interactions (frequency of contact > 0.6) with most of residues in the OBS (i.e., ASP1103.32, VAL1113.33, CYS1143.36, SER1965.46, PHE3456.51, and THR3697.39). The frequency of all interactions in the OBS of 20c, which exhibited the lowest binding affinity for D3 receptors, was lower than the higher-affinity compounds. As mentioned above, 21c showed a high frequency of contacts with PHE1063.28 including approximately 95% of hydrophobic interactions and 10% of cation–π interactions over the MDS production runs.
Consistent with previous modeling studies, the formation of key interactions between ASP1103.32 and the protonated nitrogen of the ligand stabilized the binding pose of 9a, 20a, 20b, and 20c (frequency of contact > 0.998) by 97.8% to 99.4% of the hydrogen bond formation. However, the frequency of contacts between ASP1103.32 and 20c was relatively lower (frequency of contact = 0.990) and formed only 68.6% of hydrogen bonds over the MDS production runs.
In the SBS, 9a, 20a and 21c that exhibited high subtype selectivity, presented a moderate to high probability (frequency of contacts = 0.4–0.9) of interaction with VAL862.61, LEU892.64, GLY93EL1, and GLY94EL1. In addition, the pyridine of 21c formed a hydrophobic interaction with GLU902.65 (frequency of interaction = 0.563). In contrast to our expectations, 90% of the hydrophobic interactions that formed with VAL862.61 were from the 4-fluorobenzyl group whereas 10% of the interactions were from the 4-(thiophen-2-yl)benzamide moiety.
The average frequency of the overall interactions in the binding sites (i.e., OBS and SBS) was correlated with D3 receptor binding affinity (r = −0.8756, and p = 0.0517). In addition, the average frequency of interaction in the OBS was significantly correlated with the IC50 values from the β-arrestin recruitment assay (r = −0.9934, and p = 0.0066).

2.4. Comprehensive Screening for Other GPCRs

Based on the results in the dopamine receptor radioligand binding assays, nine flexible-based compounds were selected for further evaluation for off-target binding with other GPCRs through the Psychoactive Drug Screening Program (PDSP) (Supplemental Table S2) [54]. Previous studies with the N-aryl piperazine analogs showed high binding affinity for serotonin 5-HT1A and 5-HT2B receptors. For example, many of the N-aryl piperazine-based analogs that our group developed in the past for either the D2 or D3 receptor had high affinity for the 5-HT1A receptor [55,56,57,58]. It is of interest to note that none of the panel submitted for evaluation had a high affinity for the 5-HT1A receptor or any of the other GPCRs in the screening assay (Supplemental Table S2). Compounds 21a, 21c, and 21i had modest affinity for the 5-HT3 receptor (Ki values 29–58 nM). Furthermore, a relatively high affinity of compounds 20a, 21a, 21e, 21g, and 21i for the peripheral benzodiazepine receptor (PBR) was observed. This mitochondrial-based protein is typically used as a target for imaging neuroinflammation. The results of the PDSP-binding assays also confirmed the data obtained in our lab for the binding of this panel of nine compounds to D2 and D3 receptors (Supplemental Table S2).

3. Discussion

The goal of the current study was to identify a new scaffold for D3-selective antagonists that must display a high affinity and selectivity for D3 versus D2 receptors in the radioligand binding assays, but also a high potency in a β-arrestin recruitment assay, which measures the ability of a compound to compete with dopamine in binding to the D3 receptor [21,29]. Previous studies have shown that a PET radiotracer developed in our lab having a high affinity for the D3 receptor (Kd~50 pM) and excellent selectivity versus the D2 receptor (>150-fold) was not able to image D3 receptors in vivo without pretreatment with drugs that reduce synaptic levels of dopamine [59].
For the current study, we chose metoclopramide as the lead compound for our SAR studies. Metoclopramide was chosen as the lead compound for this study because it has a modest affinity for both D2 and D3 receptors and it should be possible to make analogs of this compound having an improved D3 binding affinity while minimizing D2 receptor affinity by interacting with the SBS. The results of SAR indicated that 5-bromo-2,3-dimethoxybenzamide, the moiety from FLB457, was more favorable for binding to the OBS, which is important for determining affinity for both D3 and D2 receptors. The size of fragments in 9a,b or 11a,b that interact with the SBP residues of the D3 receptor are important for high selectivity for D3 versus D2 selectivity [60]. It is of interest to note that the appropriate length of linker between the basic amine and the secondary binding fragment was one carbon shorter than other known D3 receptor antagonists such as N-arylpiperazine congeners. The D3 receptor binding affinity was also affected by steric hindrance of the substituent on the basic amine.
A number of the compounds reported here exhibited excellent D3 binding affinity (ranging from 0.8 to 13.2 nM) and excellent selectivity (22.1- to 180-fold) for D3 vs. D2 receptors. Although analogs such as 9a, 21a, 21d, and 21g exhibit high binding affinity and subtype selectivity for the D3 receptor, 21c was identified as the best-in-series candidate because of its high D3 affinity and selectivity, and excellent potency in the β-arrestin recruitment assay (IC50 = 1.3 nM). This IC50 value was comparable with fallypride that is widely used as a non-selective PET probe for D2/D3 receptors and can bind to D3 receptors in the presence of endogenous dopamine (fallypride, IC50 = 1.7 nM) [29]. Moreover, the computational modeling studies demonstrated that the high potency of 21c may result from the short distance of the bridge-bond with ASP1103.32 and the high-frequency contacts between 21c and residues in the OBS and SBS in the D3 receptor.
Since metoclopramide was previously used in drug development and led to the identification of compounds having a diverse range of pharmacologic activity including mixed 5-HT3 antagonists/5-HT4 agonists (e.g., zacopride, BRL 24682) and D2 antagonists (e.g., clebopride, BRL 25594) [45], there was a concern that the conformational flexibility of our compounds could result in significant off-target bindings to other G-proteins. By the comprehensive screening from PDSP, these compounds possess minimal affinity for other GPCRs except a moderate affinity for 5-HT3 receptors (29–58 nM). Interestingly, 21d, which has an indole carboxamide as a secondary binding fragment, exhibited nM binding affinity for the histamine H1 receptor (0.95 nM). Other compounds acquired affinity for the translocator protein (TSPO); however, it is not clear if this off-target binding would be problematic for using these compounds in D3 receptor binding assays or behavioral studies. Further studies are ongoing in our lab to prepare radiolabeled versions of 21c for imaging D3 receptors in the brain, and SAR studies are being conducted that aim to improve the properties of this new scaffold as a means of identifying potential D3 receptor selective PET radiotracers.

4. Materials and Methods

4.1. General

5-(3-Fluoropropyl)-2,3-dimethoxybenzoic acid (1a) was prepared from methyl 5-allyl-3-methoxy salicylate via methylation for phenol, oxidation of the allyl group, fluorination, and hydrolysis of methyl ester [61]. 5-Bromo-2,3-dimethoxybenzoic acid (1b) was prepared from 5-bromo-2-hydroxy-3-methoxy benzoic acid via methylation for phenol and oxidation of aldehyde to carboxylic acid using silver catalyst [62]. For the OBS binding, tert-butyl (2-aminoethyl) ethylcarbamate was prepared from N-ethylethylenediamine via the primary amine protection, the secondary amine protection and the primary amine de-protection according to the literature [63]. 2-(2-Bromoethyl)-1,3-dioxolane and 2-(3-bromopropyl)-1,3-dioxolane were prepared via reduction followed by cyclization [64]. The other reagents and solvents were purchased from Sigma-Aldrich, TCI, Matrix Scientific, Advanced chemtech, Fisher chemical, Ambeed, Chembridge corporation, Acros organics, and Decon laboratories and used as received (Supplemental Table S3). Reactions were monitored by thin layer chromatography (TLC) using TLC silica gel 60W F254S plates and the spots were detected under UV light (254 nm) or developed using ninhydrin. Flash column chromatography was carried out on a Biotage Isolera One with a dual wavelength UV-vis detector. 1H and 13C NMR spectra were obtained on a Bruker NEO-400 spectrometer (Bruker, Billerica, MA, USA). Chemical shifts (δ) were recorded in parts per million (ppm) relative to the deuterated solvent as an internal reference. Mass spectra (m/z) were recorded on a 2695 Alliance LC-MS (Waters Corporation, Milford, MA, USA) using positive electrospray ionization (ESI+). High resolution mass spectra (HRMS, m/z) were acquired on a waters LCT premier mass spectrometer (Waters Corporation, Milford, MA, USA). PathHunterTM β-arrestin recruitment assay kit and the Chinese hamster ovary CHO-K1 cell line were purchased from DiscoverX (Fremont, CA, USA).

4.2. Chemistry

tert-Butyl ethyl(2-(5-(3-fluoropropyl)-2,3-dimethoxybenzamido)ethyl)carbamate (2a) In a mixture of 1a (1 g, 4.13 mmol), tert-butyl (2-aminoethyl) ethylcarbamate, (1.55 g, 8.26 mmol) and HBTU (1.55 g, 6.2 mmol) in DMF (20 mL), DIPEA (1.08 mL, 6.2 mmol) was added. The mixture was stirred for 24 h at RT. After completion of the reaction, the mixture was diluted with EtOAc and washed with water and brine. The volatiles were removed under reduced pressure and the crude product was purified by flash chromatography on silica gel (EtOAc/hexane = 1:2) to afford 2a (1.15 g, 68% yield) as a yellow oil. (1H NMR, 400 MHz, CDCl3): δ = 8.12 (br, 1H), 7.45 (s, 1H), 6.81 (s, 1H), 4.43 (t, J = 5.9 Hz, 1H), 4.31 (t, J = 5.9 Hz, 1H), 3.81 (s, 3H), 3.80 (s, 3H), 3.53 (q, J = 6.1 Hz, 2H), 3.37 (t, J = 6.3 Hz, 2H), 3.22 (d, J = 6.2 Hz, 2H), 2.66 (t, J = 7.4 Hz, 2H), 2.00–1.87 (m, 2H), 1.37 (s, 9H), 1.05 (t, J = 7.0 Hz, 3H); (13C NMR, 100 MHz, CDCl3): δ = 165.4, 152.3, 145.8, 137.2, 83.6, 82.0, 79.4, 61.1, 56.0, 45.7, 31.8, 31.6, 31.03, 30.98, 28.2; ESI-MS m/z calculated for C21H34FN2O5+ [M+H]+ 413.5; found 413.6.
tert-Butyl (2-(5-bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)carbamate (2b) 2b was synthesized using 1b (5 g, 19.15 mmol) in the same procedure as 2a and purified by flash chromatography on silica gel (EtOAc/hexane = 1:2) to afford 2b (8 g, 97% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 7.96 (br, 1H), 7.57 (d, J = 2.2 Hz, 1H), 7.24 (d, J = 2.3 Hz, 1H), 3.85 (s, 3H), 3.83 (s, 3H), 3.48 (q, J = 6.0 Hz, 2H), 3.37 (t, J = 6.0 Hz, 2H), 3.23 (q, J = 7.0 Hz, 2H), 1.38 (s, 9H), 1.07 (t, J = 7.0 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 164.8, 154.9, 147.9, 125.1, 119.2, 117.1, 79.8, 62.0, 57.2, 39.4, 28.6; ESI-MS m/z calculated for C18H27BrN2O5+ [M]+ 431.3; found 431.4.
N-(2-(Ethylamino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (3a) In a solution of 2a (1.15 g, 2.79 mmol) in 15 mL of CH2Cl2, TFA (15 mL, 196 mmol) was slowly added at 0 °C. The reaction mixture was warmed to RT and stirred for 1 h. The volatiles were removed followed by the residue was dissolved in CH2Cl2 and the organic layer washed by aq saturated NaHCO3 solution. The inorganic layer was extracted by CH2Cl2 and the combined layer was washed by brine, dried over anhydrous MgSO4, filtered and concentrated in vacuo to afford 3a (850 mg, 98% yield) as a yellow oil. The crude product was used for the next step without further purification. (1H NMR, 400 MHz, CD3CN): δ = 8.26 (br, 1H), 7.36 (d, J = 2.2 Hz, 1H), 7.02 (d, J = 2.1 Hz, 1H), 4.52 (t, J = 6.0 Hz, 1H), 4.40 (t, J = 6.0 Hz, 1H), 3.86 (s, 3H), 3.84 (s, 3H), 3.43 (q, J = 5.8 Hz, 2H), 2.78 (t, J = 6.1 Hz, 2H), 2.71 (t, J = 7.6 Hz, 2H), 2.65 (q, J = 7.1 Hz, 2H), 2.00–1.96 (m, 2H), 1.37 (s, 9H), 1.08 (t, J = 7.1 Hz, 3H); (13C NMR, 100 MHz, CD3CN): δ = 165.9, 153.9, 146.8, 138.7, 128.1, 122.4, 116.7, 85.2, 61.8, 56.8, 49.4, 44.5, 40.2, 32.9, 31.8, 15.7; ESI-MS m/z calculated for C16H26FN2O3+ [M+H]+ 313.4; found 313.5.
Bromo-N-(2-(ethylamino)ethyl)-2,3-dimethoxybenzamide (3b) 3b was synthesized using 2b (8 g, 18.55 mmol) in the same procedure as 3a and purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 20:1) to afford 3b (6 g, 98% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 8.24 (br, 1H), 7.60 (d, J = 2.4 Hz, 1H), 7.26 (d, J = 2.4 Hz, 1H), 3.86 (s, 3H), 3.85 (s, 3H), 3.43 (q, J = 5.7 Hz, 2H), 2.80 (t, J = 6.0 Hz, 2H), 2.66 (q, J = 7.1 Hz, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 164.5, 155.0, 148.0, 129.8, 125.2, 119.3, 117.2, 62.0, 57.2, 49.1, 44.4, 40.1, 15.4; ESI-MS m/z calculated for C13H21BrN2O3+ [M]+ 331.2; found 331.4.
N-(2-((3-(1,3-Dioxolan-2-yl)propyl)(ethyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (4a) In a solution of 3a (300 mg, 0.96 mmol) in 9.6 mL of MeCN, 2-(3-bromopropyl)-1,3-dioxolane (281 mg, 1.44 mmol) and Na2CO3 (254 mg, 2.4 mmol) were added. The reaction mixture was stirred for 24 h at 65 °C and another 1 eq of 2-(3-bromopropyl)-1,3-dioxolane (187 mg, 0.96 mmol) was added. The reaction mixture was stirred for 48 h at 65 °C. After the completion of the reaction which was checked by TLC, aq saturated NaHCO3 solution was added and the crude product was extracted with EtOAc. The organic layer was washed by brine, dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/MeOH = 10:1) to afford 10a (240 mg, 59% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 8.19 (br, 1H), 7.30 (d, J = 2.1 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 4.70 (t, J = 4.4 Hz, 1H), 4.44 (t, J = 6.0 Hz, 1H), 4.32 (t, J = 6.0 Hz, 1H), 3.79–3.75 (m, 8H), 3.69–3.65 (m, 2H), 3.35 (q, J = 5.8 Hz, 2H), 2.63 (t, J = 7.6 Hz, 2H), 2.55 (t, J = 6.1 Hz, 2H), 2.51 (q, J = 7.1 Hz, 2H), 2.44 (t, J = 7.2 Hz, 2H), 1.89–1.86 (m, 2H), 1.561.42 (m, 4H), 0.94 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 165.2, 153.4, 146.4, 138.2, 127.4, 122.0, 116.2, 104.7, 84.7, 83.1, 65.1, 61.4, 56.3, 53.3, 52.6, 47.6, 37.8, 32.4, 32.2, 32.0, 31.4, 31.3, 22.1, 15.6; ESI-MS m/z calculated for C22H36FN2O5+ [M+H]+ 427.5; found 427.6.
N-(2-((4-(1,3-Dioxolan-2-yl)butyl)(ethyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (4b) 4b was synthesized using 3a (346 mg, 1.10 mmol) and 2-(4-bromobutyl)-1,3-dioxolane (577 mg, 2.76 mmol) in the same procedures as 4a and obtained 282 mg (58% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 8.34 (br, 1H), 7.41 (d, J = 2.0 Hz, 1H), 7.06 (d, J = 2.0 Hz, 1H), 4.75 (t, J = 4.4 Hz, 1H), 4.56 (t, J = 6.0 Hz, 1H), 4.44 (t, J = 6.0 Hz, 1H), 3.90–3.88 (m, 8H), 3.79–3.76 (m, 2H), 3.49 (q, J = 5.7 Hz, 2H), 2.77–2.66 (m, 6H), 2.57 (t, J = 6.5 Hz, 2H), 2.10–2.00 (m, 2H), 1.64–1.52 (m, 4H), 1.46–1.38 (m, 2H), 1.09 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 165.3, 153.4, 146.4, 138.2, 127.4, 121.9, 116.2, 104.7, 84.7, 83.1, 65.0, 61.4, 56.3, 53.4, 52.6, 47.8, 37.6, 34.0, 32.4, 32.2, 31.4, 31.3, 22.2; ESI-MS m/z calculated for C23H38FN2O5+ [M+H]+ 441.6; found 441.7.
N-(2-((3-(1,3-Dioxolan-2-yl)propyl)(ethyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (4c) 4c was synthesized using 3b (320 mg, 0.97 mmol) in the same procedures as 4a and obtained 269 mg (62% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 8.23 (br, 1H), 7.62 (d, J = 2.4 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 4.77 (t, J = 4.8 Hz, 1H), 3.85–3.82 (m, 8H), 3.75–3.71 (m, 2H), 3.40 (q, J = 5.7 Hz, 2H), 2.59 (t, J = 6.1 Hz, 2H), 2.55 (q, J = 7.1 Hz, 2H), 2.48 (t, J = 7.2 Hz, 2H), 1.59–1.49 (m, 4H), 0.99 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 164.0, 154.9, 148.0, 129.6, 125.3, 119.3, 117.2, 105.2, 65.5, 62.0, 57.2, 53.7, 52.9, 48.0, 38.4, 32.5, 22.7, 12.2; ESI-MS m/z calculated for C19H29BrN2O5+ [M]+ 445.4; found 445.5.
N-(2-((4-(1,3-Dioxolan-2-yl)butyl)(ethyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (4d) 4d was synthesized using 3b (309 mg, 0.93 mmol) in the same procedures as 4a and obtained 412 mg (96% yield) as a yellow oil. (1H NMR, 400 MHz, CD3CN): δ = 8.25 (br, 1H), 7.62 (d, J = 2.4 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 4.70 (t, J = 4.8 Hz, 1H), 3.85–3.82 (m, 8H), 3.74–3.70 (m, 2H), 3.41 (q, J = 5.8 Hz, 2H), 2.61 (t, J = 6.1 Hz, 2H), 2.57 (q, J = 7.1 Hz, 2H), 2.47 (t, J = 7.1 Hz, 2H), 1.58–1.53 (m, 2H), 1.51–1.44 (m, 2H), 1.40–1.32 (m, 2H), 1.00 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, CD3CN): δ = 164.1, 154.9, 148.0, 129.6, 125.3, 119.3, 117.1, 105.2, 65.5, 62.0, 57.2, 53.8, 52.9, 48.1, 38.3, 34.6, 27.7, 22.8, 12.0; ESI-MS m/z calculated for C20H31BrN2O5+ [M]+ 459.4; found 459.5.
N-(2-(Ethyl(4-oxobutyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (5a) In a solution of 4a (86 mg 0.2 mmol), in 2 mL of THF, 2 mL of aq 4 N HCl was slowly added. The reaction mixture was stirred for 3 h at RT and then, neutralized by 4 mL of aq 2 N NaOH solution. The crude product was extracted with EtOAc and the organic layer was washed by aq saturated NaHCO3 solution, water and brine. The organic layer was dried over MgSO4, filtered and concentrated in vacuo to afford 11a (74 mg, 96% yield) as a colorless oil. 11a was used without further purification for the next step. ESI-MS m/z calculated for C20H32FN2O4+ [M+H]+ 383.5; found 383.5.
N-(2-(Ethyl(5-oxopentyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (5b) 5b was synthesized using 4b (42 mg, 0.1 mmol) in the same procedures as 5a and obtained 33 mg (88% yield) as a colorless oil. ESI-MS m/z calculated for C21H34FN2O4+ [M+H]+ 397.5; found 397.7.
5-Bromo-N-(2-(ethyl(4-oxobutyl)amino)ethyl)-2,3-dimethoxybenzamide (5c) 5c was synthesized using 4c (74 mg, 0.17 mmol) in the same procedures as 5a and obtained 64 mg (96% yield) as a colorless oil. ESI-MS m/z calculated for C17H25BrN2O4+ [M]+ 401.3; found 401.4.
5-Bromo-N-(2-(ethyl(5-oxopentyl)amino)ethyl)-2,3-dimethoxybenzamide (5d) 5d was synthesized using 4d (90 mg, 0.2 mmol) in the same procedures as 5a and obtained 63 mg (78% yield) as a colorless oil. ESI-MS m/z calculated for C18H27BrN2O4+ [M]+ 415.3; found 415.4.
N-(2-((4-(Dimethylamino)butyl)(ethyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (6a) In a mixture of 5a (74 mg, 0.19 mmol) and 2 M solution of dimethylamine in THF (0.97 mL, 1.93 mmol) in 2 mL of dichloroethane, sodium triacetoxyborohydride (205 mg, 0.97 mmol) was added. The mixture was stirred at RT for 16 h. After completion of the reaction, the mixture was diluted with EtOAc and washed by aq saturated NaHCO3 solution and brine. The organic layer was dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 20:1) to afford 6a (33 mg, 42% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.30 (d, J = 1.9 Hz, 1H), 7.06 (d, J = 1.9 Hz, 1H), 4.51 (t, J = 5.9 Hz, 1H), 4.39 (t, J = 5.9 Hz, 1H), 3.90 (s, 3H), 3.88 (s, 3H), 3.51 (d, J = 6.6 Hz, 2H), 2.76–2.69 (m, 4H), 2.65 (q, J = 7.1 Hz, 2H), 2.56 (t, J = 6.9 Hz, 2H), 2.33 (t, J = 7.3 Hz, 2H), 2.22 (s, 6H), 2.07–1.94 (m, 2H), 1.54–1.50 (m, 4H), 1.10 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 168.1, 154.3, 147.3, 139.2, 128.1, 122.3, 117.1, 84.9, 83.2, 62.0, 60.7, 56.7, 54.5, 53.2, 45.5, 38.6, 33.5, 33.3, 32.32, 32.27, 26.3, 26.1, 12.0; ESI-MS m/z calculated for C22H39FN3O3+ [M+H]+ 412.6; found 412.6 HRMS (ESI) for C22H39FN3O3+ [M+H]+ requires 412.2975; found 412.2972.
N-(2-((5-(Dimethylamino)pentyl)(ethyl)amino)ethyl)-5-(3-fluoropropyl)-2,3-dimethoxybenzamide (6b) 6b was synthesized using 5b (33 mg, 0.08 mmol) in the same procedures as 6a and obtained 14 mg (42% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.31 (d, J = 1.9 Hz, 1H), 7.07 (d, J = 1.8 Hz, 1H), 4.51 (t, J = 5.9 Hz, 1H), 4.40 (t, J = 5.9 Hz, 1H), 3.91 (s, 3H), 3.88 (s, 3H), 3.51 (d, J = 6.6 Hz, 2H), 2.77–2.69 (m, 4H), 2.65 (q, J = 7.2 Hz, 2H), 2.54 (t, J = 7.4 Hz, 2H), 2.31 (t, J = 7.7 Hz, 2H), 2.23 (s, 6H), 2.08–1.95 (m, 2H), 1.58–1.48 (m, 4H), 1.38–1.31 (m, 2H), 1.10 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 168.1, 154.3, 147.3, 139.2, 128.1, 122.4, 117.2, 84.9, 83.2, 62.0, 60.7, 56.7, 54.5, 53.2, 45.4, 38.6, 33.5, 33.3, 32.33, 32.27, 28.2, 28.0, 26.5, 22.2, 12.0; ESI-MS m/z calculated for C23H41FN3O3+ [M+H]+ 426.6; found 426.6 HRMS (ESI) for C23H41FN3O3+ [M+H]+ requires 426.3132; found 426.3136.
5-Bromo-N-(2-((4-(dimethylamino)butyl)(ethyl)amino)ethyl)-2,3-dimethoxybenzamide (6c) 6c was synthesized using 5c (64 mg, 0.16 mmol) in the same procedures as 6a and obtained 34 mg (49% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.53 (d, J = 2.3 Hz, 1H), 7.31 (d, J = 2.3 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.48 (d, J = 6.5 Hz, 2H), 2.68 (d, J = 6.5 Hz, 2H), 2.62 (q, J = 7.1 Hz, 2H), 2.53 (t, J = 6.7 Hz, 2H), 2.32 (t, J = 7.2 Hz, 2H), 2.22 (s, 6H), 1.50 (t, J = 3.5 Hz, 4H), 1.07 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.5, 155.4, 148.4, 130.0, 125.3, 119.7, 117.7, 62.1, 60.7, 57.1, 54.5, 53.1, 45.4, 38.7, 26.3, 26.1, 12.0; ESI-MS m/z calculated for C19H32BrN3O3+ [M]+ 430.4; found 430.5 HRMS (ESI) for C19H32BrN3O3+ [M]+ requires 430.1705; found 430.1703.
5-Bromo-N-(2-((5-(dimethylamino)pentyl)(ethyl)amino)ethyl)-2,3-dimethoxybenzamide (6d) 6d was synthesized using 5d (63 mg, 0.15 mmol) in the same procedures as 6a and obtained 31 mg (46% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.54 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.3 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.48 (d, J = 6.5 Hz, 2H), 2.67 (d, J = 6.5 Hz, 2H), 2.62 (q, J = 7.2 Hz, 2H), 2.51 (t, J = 7.4 Hz, 2H), 2.27 (t, J = 7.7 Hz, 2H), 2.21 (s, 6H), 1.55–1.45 (m, 4H), 1.35–1.28 (m, 2H), 1.07 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.5, 155.3, 148.4, 130.0, 125.3, 119.7, 117.7, 62.1, 60.8, 57.1, 54.5, 53.0, 45.5, 38.7, 28.3, 28.1, 26.6, 12.0; ESI-MS m/z calculated for C20H34BrN3O3+ [M]+ 444.4; found 444.5 HRMS (ESI) for C20H34BrN3O3+ [M]+ requires 444.1862; found 444.1872.
5-Bromo-N-(2-((3-(1,3-dioxoisoindolin-2-yl)propyl)(ethyl)amino)ethyl)-2,3-dimethoxybenzamide (7a) In a solution of 3b (2 g, 6.04 mmol) in 20 mL of DMF, N-(3-bromopropyl)phthalimide (3.4 g, 12.08 mmol) and K2CO3 (2.1 g, 15.1 mmol) were added. The mixture was heated to 65 °C and stirred for 16 h. The reaction mixture was cooled to RT and diluted with EtOAc. The mixture was washed by aq saturated NaHCO3 solution, water and brine. The organic layer was dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 40:1) to afford 7a (1.82 g, 57% yield) as a white solid. (1H NMR, 400 MHz, acetone-d6): δ = 8.32 (br, 1H), 7.82 (s, 4H), 7.66 (d, J = 2.4 Hz, 1H), 7.28 (d, J = 2.5 Hz, 1H), 3.92 (s, 3H), 3.91 (s, 3H), 3.73 (t, J = 7.1 Hz, 2H), 3.47 (q, J = 6.0 Hz, 2H), 2.66 (t, J = 6.2 Hz, 2H), 2.62–2.58 (m, 4H), 1.91–1.84 (m, 2H), 1.02 (t, J = 7.2 Hz, 3H) (13C NMR, 100 MHz, acetone-d6): δ = 168.9, 163.7, 154.9, 148.1, 135.0, 133.3, 129.7, 125.5, 123.7, 119.0, 116.9, 61.8, 57.0, 53.2, 51.6, 47.9, 38.4, 36.9, 27.1, 12.0; ESI-MS m/z calculated for C24H28BrN3O5+ [M]+ 518.4; found 518.5.
5-Bromo-N-(2-((4-(1,3-dioxoisoindolin-2-yl)butyl)(ethyl)amino)ethyl)-2,3-dimethoxybenzamide (7b) 7b was synthesized using 3b (180 mg, 0.54 mmol) and N-(4-bromobutyl)phthalimide (305 mg, 1.08 mmol) in the same procedures as 7a and obtained 220 mg (77% yield) as a colorless oil. (1H NMR, 400 MHz, acetone-d6): δ = 8.32 (br, 1H), 7.83 (s, 4H), 7.67 (d, J = 2.4 Hz, 1H), 7.26 (d, J = 2.5 Hz, 1H), 3.90 (s, 3H), 3.87 (s, 3H), 3.73 (t, J = 7.1 Hz, 2H), 3.46 (q, J = 5.8 Hz, 2H), 2.65 (t, J = 6.1 Hz, 2H), 2.62–2.54 (m, 4H), 1.76–1.69 (m, 2H), 1.58–1.51 (m, 2H), 1.04 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, acetone-d6): δ = 168.9, 163.7, 154.9, 148.1, 135.0, 133.2, 129.6, 125.5, 123.7, 119.1, 116.9, 61.8, 57.0, 53.5, 53.1, 48.1, 38.4, 27.2, 25.3, 12.2, 0.1; ESI-MS m/z calculated for C25H30BrN3O5+ [M]+ 532.4; found 532.5.
N-(2-((3-Aminopropyl)(ethyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (8a) In a solution of 7a (1.82 g, 3.42 mmol) in 34 mL of EtOH, hydrazine hydrate (519 µL, 10.25 mmol) was added. The mixture was heated at 75 °C for 3 h and cooled to RT. The mixture was diluted with EtOAc and washed by aq saturated NaHCO3 solution and brine. The organic layer was dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 10:1) to afford 8a (1.03 g, 78% yield) as a yellow oil. (1H NMR, 400 MHz, MeOD): δ = 7.52 (d, J = 2.3 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 3.89 (d, J = 2.7 Hz, 6H), 3.49 (t, J = 6.6 Hz, 2H), 2.68 (t, J = 7.0 Hz, 4H), 2.63 (q, J = 7.2 Hz, 2H), 2.57 (t, J = 7.2 Hz, 2H), 1.70–1.63 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.6, 155.4, 148.3, 130.1, 125.2, 119.8, 117.7, 62.1, 57.1, 53.2, 52.4, 48.6, 41.1, 38.7, 30.9, 12.0; ESI-MS m/z calculated for C16H26BrN3O3+ [M]+ 388.3; found 388.4.
N-(2-((4-Aminobutyl)(ethyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (8b) 8b was synthesized using 7b (220 mg, 0.41 mmol) in the same procedures as 8a and obtained 165 mg (77% yield) as a yellow oil. (1H NMR, 400 MHz, MeOD): δ = 7.53 (d, J = 2.2 Hz, 1H), 7.30 (d, J = 2.2 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.47 (t, J = 6.6 Hz, 2H), 2.69–2.59 (m, 6H), 2.52 (t, J = 6.6 Hz, 2H), 1.56–1.44 (m, 4H), 1.07 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.5, 155.3, 148.3, 130.0, 125.3, 119.7, 117.7, 62.1, 57.1, 54.5, 53.1, 48.7, 42.6, 38.7, 31.8, 25.5, 12.1; ESI-MS m/z calculated for C17H28BrN3O3+ [M]+ 402.3; found 402.4.
5-Bromo-N-(2-(ethyl(3-(4-(thiophen-2-yl)benzamido)propyl)amino)ethyl)-2,3-dimethoxybenzamide (9a) Thionyl chloride (1.38 mL, 18.9 mmol) was added to 4-(thiophen-2-yl)benzoic acid (129 mg, 0.63 mmol) in a vial. The mixture was stirred at RT for 3 h and the volatiles were removed under the reduced pressure. 8a (163 mg, 0.42 mmol) in 4.2 mL of CH2Cl2 and Et3N (0.15 mL, 1.05 mmol) were added and the mixture was stirred for 16 h. After completion of the reaction, the volatiles were removed under the reduced pressure and the crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 40:1) to afford 9a (120 mg, 50% yield) as a yellow oil. (1H NMR, 400 MHz, MeOD): δ = 7.78 (d, J = 8.6 Hz, 2H), 7.67 (d, J = 8.5 Hz, 2H), 7.50 (d, J = 2.4 Hz, 1H), 7.47 (dd, J1 = 3.6 Hz, J2 = 1.0 Hz, 1H), 7.43 (dd, J1 = 5.1 Hz, J2 = 1.0 Hz, 1H), 7.24 (d, J = 2.4 Hz, 1H), 7.11 (dd, J1 = 5.0 Hz, J2 = 3.7 Hz, 1H), 3.85 (s, 3H), 3.83 (s, 3H), 3.50 (t, J = 6.4 Hz, 2H), 3.43 (t, J = 6.8 Hz, 2H), 2.70 (t, J = 6.4 Hz, 2H), 2.672.62 (m, 4H), 1.861.79 (m, 2H), 1.07 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.6, 166.7, 155.3, 148.3, 144.3, 138.9, 134.4, 129.9, 129.5, 129.1, 127.2, 126.6, 125.6, 125.3, 119.8, 117.7, 62.1, 57.0, 53.4, 52.3, 39.7, 38.8, 27.8, 11.9; ESI-MS m/z calculated for C27H33BrN3O4S+ [M+H]+ 575.5; found 575.5 HRMS (ESI) for C27H33BrN3O4S+ [M+H]+ 574.1375; found 574.1381.
5-Bromo-N-(2-(ethyl(4-(4-(thiophen-2-yl)benzamido)butyl)amino)ethyl)-2,3-dimethoxybenzamide (9b) 9b was synthesized using 8b (120 mg, 0.3 mmol) in the same procedures as 9a and obtained 35 mg (20% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.80 (d, J = 8.5 Hz, 2H), 7.68 (d, J = 8.6 Hz, 2H), 7.52 (d, J = 2.4 Hz, 1H), 7.49 (dd, J1 = 3.6 Hz, J2 = 1.1 Hz, 1H), 7.45 (dd, J1 = 5.1 Hz, J2 = 1.1 Hz, 1H), 7.24 (d, J = 2.4 Hz, 1H), 7.12 (dd, J1 = 5.1 Hz, J2 = 3.6 Hz, 1H), 3.83 (s, 3H), 3.82 (s, 3H), 3.49 (t, J = 6.6 Hz, 2H), 3.41 (t, J = 6.6 Hz, 2H), 2.70 (t, J = 6.5 Hz, 2H), 2.64 (q, J = 7.2 Hz, 2H), 2.59 (t, J = 7.4 Hz, 2H), 1.68–1.56 (m, 4H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.7, 166.6, 155.3, 148.3, 144.3, 138.9, 134.5, 130.0, 129.5, 129.2, 127.2, 126.6, 125.6, 125.3, 119.8, 117.7, 62.1, 57.0, 54.2, 53.1, 48.8, 40.8, 38.6, 28.5, 25.5, 11.9; ESI-MS m/z calculated for C28H35BrN3O4S+ [M+H]+ 589.6; found 589.4 HRMS (ESI) for C28H35BrN3O4S+ [M+H]+ 588.1532; found 588.1555.
5-Bromo-N-(2-(ethyl(4-hydroxybutyl)amino)ethyl)-2,3-dimethoxybenzamide (10a) In a solution of 5c (586 mg, 1.46 mmol) in 15 mL of CH2Cl2, sodium triacetoxyborohydride (774 mg, 3.65 mmol) was added. The mixture was stirred at RT for 16 h. After completion of the reaction, the mixture was diluted with EtOAc and washed by aq saturated NaHCO3 solution and brine. The organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 20:1) to afford 10a (300 mg, 51% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.52 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.55 (t, J = 6.0 Hz, 2H), 3.49 (t, J = 6.7 Hz, 2H), 2.70 (t, J = 6.7 Hz, 2H), 2.64 (q, J = 7.2 Hz, 2H), 2.55 (t, J = 7.0 Hz, 2H), 1.61–1.54 (m, 4H), 1.08 (t, J = 7.2 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.7, 155.4, 148.3, 130.1, 125.2, 119.8, 117.7, 63.0, 62.1, 57.1, 54.7, 53.1, 48.7, 38.6, 31.9, 24.8, 11.9; ESI-MS m/z calculated for C17H28BrN2O4+ [M+H]+ 404.3; found 404.3.
5-Bromo-N-(2-(ethyl(5-hydroxypentyl)amino)ethyl)-2,3-dimethoxybenzamide (10b) 10b was synthesized using 5d (51 mg, 0.12 mmol) in the same procedures as 10a and obtained 25 mg (29% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.52 (d, J = 2.3 Hz, 1H), 7.30 (d, J = 2.4 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.53 (t, J = 6.6 Hz, 2H), 3.48 (t, J = 6.6 Hz, 2H), 2.69 (t, J = 6.6 Hz, 2H), 2.63 (q, J = 7.2 Hz, 2H), 2.54 (t, J = 7.5 Hz, 2H), 1.58–1.49 (m, 4H), 1.40–1.34 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.7, 155.4, 148.4, 130.1, 125.2, 119.8, 117.7, 63.0, 62.1, 57.1, 54.7, 53.1, 48.8, 38.7, 33.7, 27.9, 25.0, 12.0; ESI-MS m/z calculated for C18H29BrN2O4+ [M]+ 417.3; found 417.4.
5-Bromo-N-(2-(ethyl(4-((4-methyl-5-phenyl-4H-1,2,4-triazol-3-yl)thio)butyl)amino)ethyl)-2,3-dimethoxybenzamide (11a) In a mixture of 10a (100 mg, 0.25 mmol), 4-methyl-5-phenyl-4H-1,2,4-triazole-3-thiol (57 mg, 0.3 mmol), and PPh3 (98 mg, 0.37 mmol) in 2.5 mL of THF, DIAD (73 µL, 0.37 mmol) was slowly added. The mixture was stirred at RT for 24 h. The mixture was diluted with EtOAc and washed by aq saturated NaHCO3 solution and brine. The organic layer was dried over Na2SO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/MeOH = 15:1) to afford 11a (23 mg, 16% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.67–7.65 (m, 2H), 7.58–7.54 (m, 3H), 7.51 (d, J = 2.4 Hz, 1H), 7.27 (d, J = 2.4 Hz, 1H), 4.28 (t, J = 6.9 Hz, 2H), 3.85 (s, 6H), 3.61 (s, 3H), 3.50 (t, J = 6.4 Hz, 2H), 2.73 (t, J = 6.4 Hz, 2H), 2.72–2.63 (m, 4H), 1.98–1.90 (m, 2H), 1.63–1.56 (m, 2H), 1.09 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 168.5, 166.7, 155.3, 152.4, 148.4, 132.2, 130.3, 129.9, 129.8, 127.3, 125.3, 119.8, 117.7, 62.2, 57.1, 54.0, 53.2, 38.6, 33.7, 27.1, 24.8, 11.8; ESI-MS m/z calculated for C26H35BrN5O3S+ [M+H]+ 577.6; found 577.3 HRMS (ESI) for C26H35BrN5O3S+ [M+H]+ 576.1644; found 576.1639.
5-Bromo-N-(2-(ethyl(5-((4-methyl-5-phenyl-4H-1,2,4-triazol-3-yl)thio)pentyl)amino)ethyl)-2,3-dimethoxybenzamide (11b) 11b was synthesized using 10b (25 mg, 0.06 mmol) in the same procedures as 11a and purified by flash chromatography on silica gel (CH2Cl2/MeOH/7 N NH3 in MeOH = 20:1:0.1). 11b was obtained 10 mg (28% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.69–7.67 (m, 2H), 7.58–7.53 (m, 3H), 7.52 (d, J = 2.4 Hz, 1H), 7.29 (d, J = 2.4 Hz, 1H), 4.23 (t, J = 7.0 Hz, 2H), 3.88 (s, 3H), 3.87 (s, 3H), 3.62 (s, 3H), 3.49 (t, J = 6.5 Hz, 2H), 2.72 (t, J = 6.4 Hz, 2H), 2.66 (q, J = 7.1 Hz, 2H), 2.57 (t, J = 7.4 Hz, 2H), 1.94–1.87 (m, 2H), 1.63–1.56 (m, 2H), 1.44–1.36 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 168.4, 166.7, 155.4, 152.4, 148.4, 132.2, 130.3, 130.0, 129.9, 127.4, 125.3, 119.9, 117.7, 62.2, 57.1, 54.4, 53.2, 48.9, 38.6, 33.7, 29.0, 27.3, 25.3, 11.9; ESI-MS m/z calculated for C27H36BrN5O3S+ [M]+ 590.6; found 590.6 HRMS (ESI) for C27H37BrN5O3S+ [M]+ 590.1800; found 590.1787.
5-Bromo-N-(2-(ethyl(methyl)amino)ethyl)-2,3-dimethoxybenzamide (12) In a solution of 3b (40 mg, 0.12 mmol) in 5 mL of acetone, CH3I (7.5 µL, 0.12 mmol) and K2CO3 (36 mg, 0.26 mmol) were added. The mixture was refluxed for 16 h and cooled to RT. The volatiles were removed under the reduced pressure and the crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 40:1) to afford 12 (7.8 mg, 19% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.51 (d, J = 2.4 Hz, 1H), 7.31 (d, J = 2.4 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.52 (t, J = 6.7 Hz, 2H), 2.63 (t, J = 6.7 Hz, 2H), 2.54 (q, J = 7.2 Hz, 2H), 2.31 (s, 3H), 1.11 (t, J = 7.2 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.8, 155.4, 148.3, 130.3, 125.2, 119.8, 117.7, 62.1, 57.0, 56.6, 52.6, 41.8, 38.4, 12.4; ESI-MS m/z calculated for C14H21BrN2O3+ [M]+ 345.2; found 345.3 HRMS (ESI) for C14H22BrN2O3+ [M+H]+ 345.0814; found 345.0813.
N-(3-Bromopropyl)-4-(thiophen-2-yl)benzamide (13) 13 was synthesized using 4-(thiophen-2-yl)benzoic acid (150 mg, 0.73 mmol) and 3-bromopropylamine hydrobromide (161 mg, 0.73 mmol) in the same procedures as 9a and obtained 35 mg (15% yield) as a white solid. (1H NMR, 400 MHz, DMSO-d6): δ = 8.57 (t, J = 5.5 Hz, 1H), 7.89 (d, J = 8.5 Hz, 2H), 7.75 (d, J = 8.5 Hz, 2H), 7.64–7.61 (m, 2H), 7.17 (dd, J1 = 5.1 Hz, J2 = 3.7 Hz, 1H), 3.59 (t, J = 6.6 Hz, 2H), 3.39 (t, J = 6.6 Hz, 2H), 2.12–2.05 (m, 2H) (13C NMR, 100 MHz, DMSO-d6): δ = 166.2, 142.8, 140.2, 136.7, 129.6, 129.2, 128.6, 127.2, 126.0, 125.5, 38.4, 33.0, 18.9; ESI-MS m/z calculated for C14H14BrNOS+ [M]+ 324.2; found 324.2.
N-(3-(Ethyl(methyl)amino)propyl)-4-(thiophen-2-yl)benzamide (14) 14 was synthesized using 13 (35 mg, 0.11 mmol) and N-methylethanamine (12.8 mg, 0.22 mmol) in the same procedures as 13a and obtained 18 mg (55% yield) as a white solid. (1H NMR, 400 MHz, MeOD): δ = 7.89 (d, J = 8.6 Hz, 2H), 7.75 (d, J = 8.5 Hz, 2H), 7.52 (dd, J1 = 3.6 Hz, J2 = 1.0 Hz, 1H), 7.37 (dd, J1 = 5.1 Hz, J2 = 1.0 Hz, 1H), 7.13 (dd, J1 = 5.1 Hz, J2 = 3.7 Hz, 1H), 3.58–3.45 (m, 2H), 3.36–3.24 (m, 2H), 3.22–3.11 (m, 2H), 2.88 (s, 3H), 2.10–2.01 (m, 2H), 1.37 (t, J = 7.3 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 170.4, 144.1, 139.4, 133.7, 129.6, 129.3, 127.4, 126.7, 125.8, 54.6, 52.8, 39.8, 37.7, 26.1, 9.7; ESI-MS m/z calculated for C17H24N2OS+ [M+2H]+ 304.5; found 304.2 HRMS (ESI) for C17H23N2OS+ [M+H]+ 303.1531; found 303.1516.
tert-Butyl allyl(2-aminoethyl)carbamate (15a) In a solution of ethylenediamine (2.8 mL, 41.6 mmol) in 100 mL of CH2Cl2, ethyl trifluoroacetate (4.9 mL, 41.6 mmol) in 100 mL of CH2Cl2 was added dropwise at 0 °C. The mixture was warmed to RT and stirred for 1 h. The solvent was removed under the reduced pressure and the residue was dissolved with 210 mL of MeOH. Allyl bromide (3.6 mL, 41.6 mmol) and Et3N (6.4 mL, 46 mmol) were added slowly into the mixture and the mixture was stirred for 16 h. Then, (Boc)2O (9.6 mL, 41.6 mmol) was added and the mixture was stirred for another 4 h. The volatiles were removed under the reduced pressure and the residue was dissolved EtOAc. The organic layer was washed by aq 0.5 N HCl solution and brine, dried over MgSO4, filtered and concentrated in vacuo. Deprotection of trifluoroacetyl group was performed according to the reported method [63] and 2.8 g of 15a (34% yield) was obtained as a yellow oil. The crude product was used for the next step without further purification.
tert-Butyl (2-aminoethyl)(4-fluorobenzyl)carbamate (15b) In a solution of N-(2-aminoethyl)-2,2,2-trifluoroacetamide (6.5 g, 41.6 mmol) in 200 mL of CH2Cl2, 4-fluorobenzaldehyde (4.46 mL, 41.6 mmol) and sodium triacetoxyborohydride (17.6 g, 83 mmol) were added. The mixture was stirred for 16 h at RT and washed by aq saturated NaHCO3 solution and brine. The organic layer was dried over MgSO4, filtered and concentrated in vacuo. The crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 20:1) to afford an intermediate (1.7 g, 6.5 mmol) as a colorless oil. Protection of Boc group and deprotection of trifluoroacetyl group were performed according to the reported method [63] and 1.6 g of 15b (15% yield) was obtained as a colorless oil. The crude product was used for the next step without further purification.
tert-Butyl allyl(2-(5-bromo-2,3-dimethoxybenzamido)ethyl)carbamate (16a) 16a was synthesized using 1b (783 mg, 3 mmol) and 15a (1.2 g, 6 mmol) in the same procedures as 2a and obtained 783 mg (60% yield) as a colorless oil. (1H NMR, 400 MHz, CD3CN): δ = 8.04 (br, 1H), 7.69 (d, J = 2.3 Hz, 1H), 7.37 (d, J = 2.3 Hz, 1H), 5.96–5.87 (m, 1H), 5.26–5.19 (m, 2H), 3.97 (s, 3H), 3.95 (s, 3H), 3.94 (br, 2H), 3.59 (q, J = 6.0 Hz, 2H), 3.49 (t, J = 5.9 Hz, 2H), 1.48 (s, 9H) (13C NMR, 100 MHz, CD3CN): δ = 164.8, 154.9, 147.9, 125.1, 119.3, 117.1, 116.7, 80.2, 62.0, 57.3, 39.1, 28.6; ESI-MS m/z calculated for C19H27BrN2O5+ [M]+ 443.3; found 443.3.
tert-Butyl (2-(5-bromo-2,3-dimethoxybenzamido)ethyl)(4-fluorobenzyl)carbamate (16b) 16b was synthesized using 1b (500 mg, 1.92 mmol) and 15b (771 mg, 2.87 mmol) in the same procedures as 2a and obtained 670 mg (68% yield) as a colorless oil. (1H NMR, 400 MHz, CD3CN): δ = 7.90 (br, 1H), 7.57 (d, J = 2.4 Hz, 1H), 7.29–7.25 (m, 3H), 7.05 (t, J = 8.8 Hz, 2H), 4.42 (s, 2H), 3.86 (s, 3H), 3.83 (s, 3H), 3.48 (q, J = 6.0 Hz, 2H), 3.39 (br, 2H), 1.38 (s, 9H) (13C NMR, 100 MHz, CD3CN): δ = 164.9, 164.1, 161.7, 156.8, 155.0, 148.0, 136.1, 130.3, 125.2, 119.4, 117.1, 116.2, 116.0, 80.6, 62.0, 57.3, 46.7, 39.0, 28.6; ESI-MS m/z calculated for C23H28BrFN2O5+ [M]+ 511.4; found 511.3.
N-(2-(Allylamino)ethyl)-5-bromo-2,3-dimethoxybenzamide (17a) 17a was synthesized using 16a (350 mg, 0.79 mmol) in the same procedures as 3a and obtained 250 mg (92% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.45 (d, J = 2.4 Hz, 1H), 7.30 (d, J = 2.3 Hz, 1H), 5.96–5.86 (m, 1H), 5.26–5.12 (m, 2H), 3.89 (s, 3H), 3.87 (s, 3H), 3.51 (t, J = 6.4 Hz, 2H), 3.28 (t, J = 1.2 Hz, 1H), 3.27 (t, J = 1.3 Hz, 1H), 2.81 (t, J = 6.4 Hz, 2H) (13C NMR, 100 MHz, CD3CN): δ = 167.4, 155.4, 148.1, 137.2, 131.0, 124.9, 119.6, 117.4, 117.1, 62.1, 57.1, 52.9, 40.4; ESI-MS m/z calculated for C14H19BrN2O3+ [M]+ 343.2; found 343.5.
5-Bromo-N-(2-((4-fluorobenzyl)amino)ethyl)-2,3-dimethoxybenzamide (17b) 17b was synthesized using 16b (656 mg, 1.28 mmol) in the same procedures as 3a and obtained 480 mg (91% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.45 (d, J = 2.4 Hz, 1H), 7.39–7.35 (m, 2H), 7.29 (d, J = 2.4 Hz, 1H), 7.04 (t, J = 8.8 Hz, 2H), 3.89 (s, 3H), 3.83 (s, 3H), 3.79 (s, 2H), 3.53 (t, J = 6.2 Hz, 2H), 2.83 (t, J = 6.2 Hz, 2H) (13C NMR, 100 MHz, MeOD): δ = 167.4, 164.9, 162.5, 155.3, 148.1, 136.8, 131.6, 131.5, 130.8, 125.0, 119.6, 117.7, 116.3, 116.1, 62.1, 57.1, 53.5, 40.3; ESI-MS m/z calculated for C18H20BrFN2O3+ [M]+ 411.3; found 411.3.
N-(2-(Allyl(3-(1,3-dioxoisoindolin-2-yl)propyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (18a) 18a was synthesized using 17a (240 mg, 0.7 mmol) in the same procedures as 7a and obtained 100 mg (27% yield) as a colorless oil. (1H NMR, 400 MHz, acetone-d6): δ = 8.29 (br, 1H), 7.83 (s, 4H), 7.65 (d, J = 2.4 Hz, 1H), 7.28 (d, J = 2.4 Hz, 1H), 5.92–5.85 (m, 1H), 5.21–5.16 (m, 1H), 5.09–5.06 (m, 1H), 3.92 (s, 3H), 3.91 (s, 3H), 3.72 (t, J = 7.1 Hz, 2H), 3.48 (q, J = 6.0 Hz, 2H), 3.20 (d, J = 1.6 Hz, 2H), 2.68 (t, J = 6.2 Hz, 2H), 2.62 (t, J = 6.9 Hz, 2H), 1.92–1.85 (m, 2H) (13C NMR, 100 MHz, acetone-d6): δ = 168.9, 163.8, 154.9, 148.1, 136.7, 135.0, 133.3, 129.8, 125.5, 123.7, 119.0, 117.8, 116.9, 61.8, 57.4, 57.0, 53.4, 51.7, 38.2, 36.8, 26.9; ESI-MS m/z calculated for C25H29BrN3O5+ [M+H]+ 531.4; found 531.4.
5-Bromo-N-(2-((3-(1,3-dioxoisoindolin-2-yl)propyl)(4-fluorobenzyl)amino)ethyl)-2,3-dimethoxybenzamide (18b) 18b was synthesized using 17b (270 mg, 0.66 mmol) in the same procedures as 7a and obtained 333 mg (84% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.80–7.75 (m, 4H), 7.44 (d, J = 2.4 Hz, 1H), 7.29 (dd, J1 = 8.5 Hz, J2 = 5.5 Hz, 2H), 7.26 (d, J = 2.4 Hz, 1H), 6.85 (t, J = 8.8 Hz, 2H), 3.87 (s, 3H), 3.84 (s, 3H), 3.68 (t, J = 7.0 Hz, 2H), 3.59 (s, 2H), 3.47 (t, J = 6.0 Hz, 2H), 2.66 (t, J = 6.1 Hz, 2H), 2.53 (t, J = 6.8 Hz, 2H), 1.91–1.84 (m, 2H) (13C NMR, 100 MHz, MeOD): δ = 170.0, 166.6, 164.6, 162.2, 155.3, 148.3, 135.4, 133.4, 132.03, 130.95, 130.1, 125.3, 124.2, 119.7, 117.7, 116.0, 115.8, 62.2, 58.6, 57.1, 53.9, 52.0, 38.8, 37.1, 27.0; ESI-MS m/z calculated for C29H30BrFN3O5+ [M+H]+ 599.5; found 599.3.
N-(2-((3-Aminopropyl)(propyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (19a) 19a was synthesized using 18a (100 mg, 0.19 mmol) in the same procedures as 8a and obtained 25 mg (33% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.52 (d, J = 2.3 Hz, 1H), 7.31 (d, J = 2.3 Hz, 1H), 3.89 (s, 3H), 3.88 (s, 3H), 3.48 (t, J = 6.5 Hz, 2H), 2.70–2.66 (m, 4H), 2.56 (t, J = 7.1 Hz, 2H) 2.50–2.46 (m, 2H), 1.70–1.62 (m, 2H), 1.57–1.47 (m, 2H), 0.91 (t, J = 7.4 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.6, 155.3, 148.3, 130.1, 125.2, 119.8, 117.7, 62.1, 57.4, 57.1, 53.9, 53.1, 41.0, 38.8, 30.9, 21.2, 12.3; ESI-MS m/z calculated for C17H28BrN3O3+ [M]+ 402.3; found 402.4.
N-(2-((3-Aminopropyl)(4-fluorobenzyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (19b) 19b was synthesized using 18b (156 mg, 0.26 mmol) in the same procedures as 8a and obtained 61 mg (50% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.50 (d, J = 2.4 Hz, 1H), 7.34 (dd, J1 = 8.5 Hz, J2 = 5.6 Hz, 2H), 7.31 (d, J = 2.4 Hz, 1H), 6.97 (7, J = 8.8 Hz, 2H), 3.90 (s, 3H), 3.85 (s, 3H), 3.60 (s, 2H), 3.49 (t, J = 6.2 Hz, 2H), 2.67–2.61 (m, 4H), 2.53 (t, J = 7.0 Hz, 2H), 1.70–1.63 (m, 2H) (13C NMR, 100 MHz, MeOD): δ = 166.5, 164.7, 162.3, 155.3, 148.3, 136.6, 132.0, 131.9, 130.0, 125.3, 119.7, 117.7, 116.1, 115.9, 62.2, 58.9, 57.8, 52.5, 40.9, 31.1; ESI-MS m/z calculated for C21H27BrFN3O3+ [M]+ 468.4; found 468.5.
5-Bromo-2,3-dimethoxy-N-(2-(propyl(3-(4-(thiophen-2-yl)benzamido)propyl)amino)ethyl)benzamide (20a) 20a was synthesized using 19a (25 mg, 0.06 mmol) in the same procedures as 9a and obtained 25 mg (71% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.79 (d, J = 8.4 Hz, 2H), 7.69 (d, J = 8.4 Hz, 2H), 7.50 (d, J = 2.4 Hz, 1H), 7.49 (d, J = 3.7 Hz, 1H), 7.44 (d, J = 5.0 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 7.12 (dd, J1 = 5.0 Hz, J2 = 3.7 Hz, 1H), 3.86 (s, 3H), 3.84 (s, 3H), 3.50 (t, J = 6.3 Hz, 2H), 3.43 (t, J = 6.8 Hz, 2H), 2.71 (t, J = 6.3 Hz, 2H), 2.64 (t, J = 6.8 Hz, 2H), 2.53–2.49 (m, 2H), 1.86–1.79 (m, 2H), 1.57–1.48 (m, 2H), 0.91 (t, J = 7.3 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.7, 166.7, 155.3, 148.3, 144.3, 138.9, 134.4, 130.0, 129.5, 129.1, 127.2, 126.6, 125.6, 125.3, 119.8, 117.7, 62.1, 57.4, 57.1, 54.1, 52.9, 39.6, 38.9, 27.9, 21.2, 12.3; ESI-MS m/z calculated for C28H35BrN3O4S+ [M+H]+ 589.6; found 589.4 HRMS (ESI) for C28H35BrN3O4S+ [M+H]+ 588.1532; found 588.1527.
N-(2-(Allyl(3-(4-(thiophen-2-yl)benzamido)propyl)amino)ethyl)-5-bromo-2,3-dimethoxybenzamide (20b) 20b was synthesized using 13 (36 mg, 0.11 mmol) and 17a (38 mg, 0.11 mmol) in the same procedures as 7a and obtained 7 mg (21% yield) as a colorless oil. 20 mg of 17a (20 mg, 0.06 mmol) was recovered. (1H NMR, 400 MHz, MeOD): δ = 7.79 (d, J = 8.6 Hz, 2H), 7.69 (d, J = 8.5 Hz, 2H), 7.50–7.49 (m, 2H), 7.45 (dd, J1 = 5.1 Hz, J2 = 1.0 Hz, 1H), 7.26 (d, J = 2.4 Hz, 1H), 7.13 (dd, J1 = 5.1 Hz, J2 = 3.7 Hz, 1H), 5.97–5.86 (m, 1H), 5.26–5.21 (m, 1H), 5.18–5.14 (m, 1H), 3.87 (s, 3H), 3.85 (s, 3H), 3.51 (t, J = 6.3 Hz, 2H), 3.43 (t, J = 6.8 Hz, 2H), 3.22 (d, J = 6.6 Hz, 2H), 2.72 (t, J = 6.3 Hz, 2H), 2.65 (t, J = 6.9 Hz, 2H), 1.87–1.80 (m, 2H) (13C NMR, 100 MHz, MeOD): δ = 169.7, 166.7, 155.3, 148.3, 144.3, 138.9, 136.4, 134.4, 130.0, 129.5, 129.1, 127.2, 126.6, 125.6, 125.3, 119.8, 118.8, 117.7, 62.2, 58.2, 57.1, 53.7, 52.5, 39.6, 38.8, 27.9; ESI-MS m/z calculated for C28H33BrN3O4S+ [M+H]+ 587.6; found 587.3 HRMS (ESI) for C28H33BrN3O4S+ [M+H]+ 586.1375; found 586.1377.
5-Bromo-N-(2-((4-fluorobenzyl)(3-(4-(thiophen-2-yl)benzamido)propyl)amino)ethyl)-2,3-dimethoxybenzamide (20c) 20c was synthesized using 19b (60 mg, 0.13 mmol) in the same procedures as 9a and obtained 40 mg (32% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.75 (d, J = 8.6 Hz, 2H), 7.68 (d, J = 8.6 Hz, 2H), 7.49 (dd, J1 = 3.6 Hz, J2 = 1.1 Hz, 1H), 7.47 (d, J = 2.4 Hz, 1H), 7.44 (dd, J1 = 5.1 Hz, J2 = 1.0 Hz, 1H), 7.33 (dd, J1 = 8.6 Hz, J2 = 5.5 Hz, 2H), 7.27 (d, J = 2.4 Hz, 1H), 7.12 (dd, J1 = 5.1 Hz, J2 = 3.6 Hz, 1H), 6.92 (t, J = 8.8 Hz, 2H), 3.86 (s, 3H), 3.83 (s, 3H), 3.62 (s, 2H), 3.50 (t, J = 6.0 Hz, 2H), 3.41 (t, J = 6.8 Hz, 2H), 2.68 (t, J = 6.1 Hz, 2H), 2.60 (t, J = 6.8 Hz, 2H), 1.88–1.81 (m, 2H) (13C NMR, 100 MHz, MeOD): δ = 169.7, 166.7, 164.7, 162.3, 155.3, 148.3, 144.3, 138.9, 136.54, 136.50, 134.4, 132.1, 132.0, 130.2, 129.6, 129.5, 129.2, 127.2, 126.6, 125.6, 125.2, 119.7, 117.7, 116.1, 115.9, 62.2, 58.8, 57.1, 54.0, 52.3, 39.3, 38.9, 27.8; ESI-MS m/z calculated for C32H33BrFN3O4S+ [M]+ 654.6; found 654.6 HRMS (ESI) for C32H34BrFN3O4S+ [M+H]+ 654.1437; found 654.1447.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)-2-naphthamide (21a) In a solution of 8a (30 mg, 0.08 mmol) in 2 mL of CH2Cl2, 2-naphthoyl chloride (22 mg, 0.12 mmol) and Et3N (12.9 µL, 0.09 mmol) were added. The reaction mixture was stirred for 1 h at RT followed by 2 mL of MeOH was added. After the mixture was stirred for 10 min, the crude product was purified by flash chromatography on silica gel (CH2Cl2/7 N NH3 in MeOH = 80:1) to afford 21a (37 mg, 86% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.31 (s, 1H), 7.95–89 (m, 3H), 7.83 (dd, J1 = 8.6 Hz, J2 = 1.7 Hz, 1H), 7.60–7.53 (m, 2H), 7.48 (d, J = 2.4 Hz, 1H), 7.22 (d, J = 2.4 Hz, 1H), 3.85 (s, 3H), 3.83 (s, 3H), 3.53–3.48 (m, 4H), 2.72 (t, J = 6.4 Hz, 2H), 2.70–2.64 (m, 4H), 1.90–1.83 (m, 2H), 1.09 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 170.3, 166.7, 155.3, 148.3, 136.4, 134.2, 133.1, 130.1, 130.0, 129.4, 128.9, 128.8, 128.0, 125.2, 124.9, 119.7, 117.7, 62.1, 57.0, 53.4, 52.3, 39.8, 38.8, 27.9, 11.9; ESI-MS m/z calculated for C27H32BrN3O4+ [M]+ 542.5; found 542.5 HRMS (ESI) for C27H33BrN3O4+ [M+H]+ 542.1654; found 542.1649.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)quinoline-4-carboxamide (21b) 21b was synthesized using 8a (30 mg, 0.08 mmol) and 4-quinolinecarboxylic acid (21 mg, 0.12 mmol) in the same procedures as 9a and obtained 27 mg (65% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.90 (d, J = 4.4 Hz, 1H), 8.16 (d, J = 8.4 Hz, 1H), 8.08 (d, J = 8.4 Hz, 1H), 7.84–7.80 (m, 1H), 7.68–7.64 (m, 1H), 7.54 (d, J = 4.4 Hz, 1H), 7.50 (d, J = 2.4 Hz, 1H), 7.28 (d, J = 2.4 Hz, 1H), 3.88 (s, 3H), 3.86 (s, 3H), 3.56–3.49 (m, 4H), 2.75–2.66 (m, 6H), 1.93–1.85 (m, 2H), 1.10 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.8, 166.7, 155.3, 151.2, 149.3, 148.3, 144.6, 131.7, 130.1, 129.9, 129.1, 126.7, 126.1, 125.3, 120.3, 119.8, 117.7, 62.2, 57.1, 53.3, 52.2, 39.5, 38.8, 28.0, 12.0; ESI-MS m/z calculated for C26H31BrN4O4+ [M]+ 543.5; found 543.4 HRMS (ESI) for C26H32BrN4O4+ [M+H]+ 543.1607; found 543.1622.
5-Bromo-N-(2-(ethyl(3-(4-(pyridin-4-yl)benzamido)propyl)amino)ethyl)-2,3-dimethoxybenzamide (21c) 21c was synthesized using 8a (30 mg, 0.08 mmol) and 4-(4-pyridyl)benzoic acid (24 mg, 0.12 mmol) in the same procedures as 9a and obtained 33 mg (75% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.61 (dd, J1 = 4.6 Hz, J2 = 1.6 Hz, 2H), 7.92 (d, J = 8.4 Hz, 2H), 7.82 (d, J = 8.5 Hz, 2H), 7.75 (dd, J1 = 4.6 Hz, J2 = 1.6 Hz, 2H), 7.49 (d, J = 2.3 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 3.87 (s, 3H), 3.84 (s, 3H), 3.51 (t, J = 6.4 Hz, 2H), 3.46 (t, J = 6.8 Hz, 2H), 2.71 (t, J = 6.4 Hz, 2H), 2.70–2.63 (m, 4H), 1.88–1.81 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.5, 166.7, 155.3, 150.9, 149.7, 148.3, 141.8, 136.6, 130.0, 129.3, 128.4, 125.3, 123.4, 119.8, 117.7, 62.1, 57.1, 53.4, 52.3, 40.6, 39.8, 38.8, 27.8, 11.9; ESI-MS m/z calculated for C28H33BrN4O4+ [M]+ 569.5; found 569.5 HRMS (ESI) for C28H34BrN4O4+ [M+H]+ 569.1763; found 569.1775.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)-1H-indole-2-carboxamide (21d) 21d was synthesized using 8a (30 mg, 0.08 mmol) and indole-2-carboxylic acid (19 mg, 0.12 mmol) in the same procedures as 9a and obtained 17 mg (42% yield) as a yellow oil. (1H NMR, 400 MHz, MeOD): δ = 7.57 (d, J = 8.0 Hz, 1H), 7.50 (d, J = 2.4 Hz, 1H), 7.42 (dd, J1 = 8.3 Hz, J2 = 0.6 Hz, 1H), 7.23 (d, J = 2.4 Hz, 1H), 7.22–7.18 (m, 1H), 7.06–7.02 (m, 1H), 7.00 (d, J = 0.6 Hz, 1H), 3.86 (s, 3H), 3.83 (s, 3H), 3.51 (t, J = 6.4 Hz, 2H), 3.44 (t, J = 6.9 Hz, 2H), 2.71 (t, J = 6.4 Hz, 2H), 2.68–2.63 (m, 4H), 1.87–1.80 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.8, 164.3, 155.3, 148.3, 138.3, 132.4, 130.0, 129.1, 125.2, 125.1, 122.8, 121.2, 119.7, 117.7, 113.1, 104.2, 62.1, 57.0, 53.4, 52.3, 39.1, 38.8, 28.1, 11.9; ESI-MS m/z calculated for C25H31BrN4O4+ [M]+ 531.5; found 531.4 HRMS (ESI) for C25H32BrN4O4+ [M+H]+ 531.1607; found 531.1596.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)imidazo[1,2-a]pyridine-2-carboxamide (21e) 21e was synthesized using 8a (30 mg, 0.08 mmol) and imidazo[1,2-a]pyridine-2-carobxylic acid (20 mg, 0.12 mmol) in the same procedures as 9a and obtained 14 mg (34% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.40 (d, J = 6.8 Hz, 1H), 8.13 (s, 1H), 7.45 (d, J = 9.2 Hz, 1H), 7.34–7.30 (m, 1H), 7.10 (d, J = 2.4 Hz, 1H), 7.04 (d, J = 2.3 Hz, 1H), 6.93 (td, J1 = 6.7 Hz, J2 = 0.6 Hz, 1H), 3.78 (s, 3H), 3.77 (s, 3H), 3.58 (t, J = 5.8 Hz, 2H), 3.50 (t, J = 6.3 Hz, 2H), 2.73 (t, J = 5.6 Hz, 2H), 2.70–2.66 (m, 4H), 1.86–1.79 (m, 2H), 1.10 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 167.6, 164.8, 155.0, 147.7, 146.2, 140.4, 131.3, 128.7, 128.1, 124.3, 119.0, 118.5, 117.1, 116.0, 114.9, 62.0, 56.9, 53.9, 53.8, 48.6, 40.4, 38.8, 26.8, 11.8; ESI-MS m/z calculated for C24H30BrN5O4+ [M]+ 532.4; found 532.4 HRMS (ESI) for C24H31BrN5O4+ [M+H]+ 532.1559; found 532.1559.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)isonicotinamide (21f) 21f was synthesized using 8a (30 mg, 0.08 mmol) and isonicotinic acid (15 mg, 0.12 mmol) in the same procedures as 9a and obtained 15 mg (40% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.66 (dd, J1 = 4.5 Hz, J2 = 1.7 Hz, 2H), 7.73 (dd, J1 = 4.5 Hz, J2 = 1.7 Hz, 2H), 7.49 (d, J = 2.4 Hz, 1H), 7.28 (d, J = 2.4 Hz, 1H), 3.87 (d, J = 2.4 Hz, 6H), 3.50 (t, J = 6.5 Hz, 2H), 3.45 (t, J = 6.9 Hz, 2H), 2.70 (t, J = 6.4 Hz, 2H), 2.67–2.61 (m, 4H), 1.86–1.79 (m, 2H), 1.08 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 167.7, 166.7, 155.3, 151.1, 148.3, 144.1, 130.1, 125.2, 123.0, 119.8, 117.7, 62.1, 57.1, 53.4, 52.2, 39.7, 38.8, 28.0, 12.0; ESI-MS m/z calculated for C22H29BrN4O4+ [M]+ 493.4; found 493.4 HRMS (ESI) for C22H30BrN4O4+ [M+H]+ 493.1450; found 493.1451.
5-Bromo-N-(2-(ethyl(3-(4-(thiophen-3-yl)benzamido)propyl)amino)ethyl)-2,3-dimethoxybenzamide (21g) 21g was synthesized using 8a (30 mg, 0.08 mmol) and 4-(thiophen-3-yl)benzoic acid (25 mg, 0.12 mmol) in the same procedures as 9a and obtained 6 mg (15% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.81 (d, J = 8.5 Hz, 2H), 7.74 (d, J = 2.1 Hz, 1H), 7.72 (d, J = 8.5 Hz, 2H), 7.51 (d, J = 2.2 Hz, 2H), 7.50 (d, J = 2.4 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 3.86 (s, 3H), 3.84 (s, 3H), 3.51 (t, J = 6.5 Hz, 2H), 3.45 (t, J = 6.8 Hz, 2H), 2.72 (t, J = 6.4 Hz, 2H), 2.69–2.63 (m, 4H), 1.87–1.80 (m, 2H), 1.09 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 169.9, 166.8, 155.3, 148.3, 142.5, 140.3, 134.1, 130.0, 129.0, 127.9, 127.3, 127.2, 125.2, 122.9, 119.8, 117.7, 62.1, 57.1, 54.0, 52.4, 39.7, 38.8, 27.9, 11.9; ESI-MS m/z calculated for C27H33BrN3O4S+ [M+H]+ 575.5; found 575.2 HRMS (ESI) for C27H33BrN3O4S+ [M+H]+ 574.1375; found 574.1381.
5-Bromo-N-(2-((3-(3-(dimethylamino)benzamido)propyl)(ethyl)amino)ethyl)-2,3-dimethoxybenzamide (21h) 21h was synthesized using 8a (30 mg, 0.08 mmol) and 3-dimethylaminobenzoic acid (20 mg, 0.12 mmol) in the same procedures as 9a and obtained 3 mg (7% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 8.23 (t, J = 1.8 Hz, 1H), 7.99 (d, J = 7.9 Hz, 1H), 7.85 (dd, J1 = 7.9 Hz, J2 = 2.3 Hz, 1H), 7.70 (t, J = 8.0 Hz, 1H), 7.52 (d, J = 2.4 Hz, 1H), 7.32 (d, J = 2.4 Hz, 1H), 3.91 (s, 3H), 3.89 (s, 3H), 3.83 (t, J = 5.9 Hz, 2H), 3.55 (t, J = 6.4 Hz, 2H), 3.47–3.43 (m, 2H), 3.43–3.37 (m, 4H), 3.34 (s, 6H), 2.17–2.10 (m, 2H), 1.39 (t, J = 7.2 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 168.8, 168.3, 155.3, 148.5, 144.7, 137.7, 132.1, 129.8, 129.5, 125.3, 124.8, 121.0, 120.2, 117.6, 62.2, 57.2, 55.0, 53.4, 52.1, 47.1, 38.0, 36.5, 25.6, 9.2; ESI-MS m/z calculated for C25H35BrN4O4+ [M]+ 535.5; found 535.4 HRMS (ESI) for C25H36BrN4O4+ [M+H]+ 535.1920; found 535.1934.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)thiophen- e-3-carboxamide (21i) 21i was synthesized using 8a (30 mg, 0.08 mmol) and 3-thiophenecaroboxylic acid (15 mg, 0.12 mmol) in the same procedures as 9a and obtained 24 mg (63% yield) as a colorless oil. (1H NMR, 400 MHz, MeOD): δ = 7.99 (dd, J1 = 2.7 Hz, J2 = 1.6 Hz, 1H), 7.50 (d, J = 2.4 Hz, 1H), 7.47–7.43 (m, 2H), 7.28 (d, J = 2.4 Hz, 1H), 3.87 (s, 6H), 3.50 (t, J = 6.5 Hz, 2H), 3.39 (t, J = 7.0 Hz, 2H), 2.71 (t, J = 6.4 Hz, 2H), 2.68–2.61 (m, 4H), 1.84–1.77 (m, 2H), 1.07 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.7, 165.7, 155.3, 148.3, 138.7, 130.1, 129.7, 127.6, 127.5, 125.2, 119.7, 117.7, 62.1, 57.1, 53.3, 52.2, 48.7, 39.2, 38.7, 27.9, 11.9; ESI-MS m/z calculated for C21H29BrN3O4S+ [M+H]+ 499.4; found 499.2 HRMS (ESI) for C21H29BrN3O4S+ [M+H]+ 498.1062; found 498.1060.
N-(3-((2-(5-Bromo-2,3-dimethoxybenzamido)ethyl)(ethyl)amino)propyl)-1-methyl-1H-indole-2-carboxamide (21j) 21j was synthesized using 8a (30 mg, 0.08 mmol) and 1-methylindole-2-carboxylic acid (21 mg, 0.12 mmol) in the same procedures as 9a and obtained 8 mg (20% yield) as a yellow oil. (1H NMR, 400 MHz, MeOD): δ = 7.58 (d, J = 8.0 Hz, 1H), 7.50 (d, J = 2.3 Hz, 1H), 7.43 (d, J = 8.4 Hz, 1H), 7.29 (t, J = 7.3 Hz, 1H), 7.25 (d, J = 2.4 Hz, 1H), 7.10 (t, J = 7.6 Hz, 1H), 6.95 (s, 1H), 4.00 (s, 3H), 3.87 (s, 3H), 3.85 (s, 3H), 3.54 (t, J = 6.4 Hz, 2H), 3.44 (t, J = 6.8 Hz, 2H), 2.76 (t, J = 6.3 Hz, 2H), 2.74–2.67 (m, 4H), 1.89–1.82 (m, 2H), 1.12 (t, J = 7.1 Hz, 3H) (13C NMR, 100 MHz, MeOD): δ = 166.8, 165.2, 155.3, 148.3, 140.6, 133.5, 130.0, 127.8, 125.2, 125.1, 122.9, 121.5, 119.7, 117.7, 111.2, 105.8, 62.1, 57.0, 54.9, 53.4, 52.3, 39.1, 38.7, 31.9, 27.9, 11.8; ESI-MS m/z calculated for C26H34BrN4O4+ [M+H]+ 546.5; found 546.3 HRMS (ESI) for C26H34BrN4O4+ [M+H]+ 545.1763; found 545.1745.

4.3. Statistical Analysis

4.3.1. Radioligand Binding Assays

Ki values for D2 and D3 receptors were measured using [125I]IABN in human D2 and D3 receptors expressed in HEK cells, respectively. A filtration binding assay was used to characterize membrane-associated receptor binding properties [49]. The details for the procedures were described in the literature [26].

4.3.2. β-Arrestin Recruitment Assay

CHO-K1 cells which were overexpressed human D3 receptors were cultured in assaycompleteTM cell culture kit 107. Cells were seeded at a density of 25,000 cells per well of 96-well plate, and incubated at 5% CO2, 37 °C. Two days later, test compounds were dissolved in DMSO, and diluted with 11-point series in phosphate-buffered saline (PBS). Prepared compounds were added to the cells, and it was incubated for 30 min at 5% CO2, 37 °C. Then, cells were treated with 30 nM (EC80) of dopamine, and the plate was incubated another 90 min. PathHunterTM detection reagent was added to each well, and then plate was incubated for 80 min at RT in the dark. The chemiluminescent signal was measured by PerkinElmer Enspire plate reader (PerkinElmer, Boston, MA). Data were analyzed by Prism followed by non-linear regression.

4.3.3. Molecular Docking and Molecular Dynamics Simulations (MDS)

The 4 compounds with different N-alkyl groups (9a, 20a, 20b, and 20c) and the best candidate 21c were selected and performed for molecular docking and MDS studies on the D3 receptor. The protonated status at physiological pH of each compound was predicted by using Open Babel v3.1.0 [65]. Then, the molecular docking studies and MDS were performed by following the previous protocols [29]. In brief, molecular docking studies were performed via the AutoDock 4.2 [66] plugin on PyMOL (pymol.org). The X-ray structure of the D3 receptor (PDB: 3PBL, resolution: 2.89 Å) was obtained from the RCSB Protein Data Bank (www.rcsb.org (accessed on 19 May 2022)). Heteroatoms were removed from the crystal structure and polar hydrogens were added. Non-polar hydrogens were removed from selected compounds. A grid box with a dimension of 30 × 30 × 28.2 Å3 was applied for covering OBS and SBS bindings. The Lamarckian Genetic Algorithm with a maximum of 2,500,000 energy evaluations was used to calculated 100 protein–ligand binding poses for each compound. The D3 receptor−ligand complex that reproduced the crystallographic ligand binding pose with good docking score was subjected for the evaluation.
The CHARMM-GUI web-sever [67] was used for MDS preparation. The topology and parameter files of protonated compounds were generated by the Ligand Reader and Modeler module [68,69]. The Bilayer Membrane Builder module [70,71] was used for building the MDS system with FF19SB force field. The protein–ligand complexes generated from docking studies were aligned to the D3 receptor structure obtained from the Orientations of Protein in Membranes (OPM) database [72], and the POPC membrane were placed by using the OPM D3 receptor model. The protein, ligand, and membrane complexes were solvated in a TIP3P water box, and then Monte-Carlo sampling was used to add 0.15 M NaCl for charge neutralization. The MDS studies were performed via Amber18 [73] on the high-performance computing (HPC) cluster at Center for Biomedical Image Computing and Analytics at the University of Pennsylvania. The input files of system minimization, 6 steps equilibration including 2 steps NVT ensemble and 4 steps NPT ensemble, and 5 copies of 200 ns production run for MDS were generated from the last step of Membrane Builder [70,71] on the CHARMM-GUI web-sever [67].
The 50 to 200 ns of production simulation with a total of 7500 frames (1500 frames of 5 production simulation copies) for each compound were used for further MDS analysis. The interactions between a ligand and protein in the production simulations were calculated by using the software BINANA v2.1 [74].

5. Conclusions

A new scaffold was designed based on metoclopramide and identified having high affinity and subtype selectivity for the D3 receptor versus the D2 receptor. Initially, 9a having 4-(thiophen-2-yl)benzamide was recognized as a lead compound showing high binding affinity and subtype selectivity for the D3 receptor (Ki D2 = 169 nM and D3 = 1 nM). Although different aryl carboxamides exhibited excellent binding affinities preferring D3 receptors, 21c was the most potent (IC50 = 1.3 nM) for competing with dopamine in the β-arrestin recruitment assay. Furthermore, the comprehensive screening of 21c revealed the minimal off-target binding for other CNS targets. Molecular docking or MDS demonstrated that interactions between 21c and the D3 receptor were comparable with fallypride that was known for potent D2/D3 antagonists. These results suggested that 21c may have a greater potential for competing with synaptic dopamine for binding to the D3 receptor. Overall, this novel scaffold can be developed as high-affinity D3 receptor antagonists that bind with low affinity at D2 receptors and other CNS receptors.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24010432/s1.

Author Contributions

Conceptualization, H.Y.K. and R.H.M.; Methodology, H.Y.K., J.Y.L., C.-J.H. and M.T.; Validation, R.R.L. and R.H.M.; Formal analysis, H.Y.K., J.Y.L., C.-J.H. and R.H.M.; Investigation, H.Y.K., J.Y.L., C.-J.H., M.T. and R.H.M.; Resources, R.R.L. and R.H.M.; Data curation, R.R.L. and R.H.M.; Writing—original draft, H.Y.K., J.Y.L., C.-J.H. and R.H.M.; Writing—review and editing, H.Y.K., J.Y.L., R.R.L. and R.H.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from National Institute on Drug Abuse, grant number DA029840.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Off-target binding profiles for GPCRs were generously provided by the National Institute of Mental Health’s Psychoactive Drug Screening Program, Contact# HHSN-271-2018-00023-C (NIMH PDSP). The NIMH PDSP is Directed by Bryan L. Roth at the University of North Carolina at Chapel Hill and Project Officer Jamie Driscoll at NIMH, Bethesda MD, USA. The molecular dynamic simulation studies were conducted on the high-performance computing cluster (https://www.med.upenn.edu/cbica/cubic.html) at the University of Pennsylvania Center for Biomedical Image Computing and Analytics and supported by the National Institutes of Health, Grant Number: 1S10OD023495-01.

Conflicts of Interest

The authors declare no competing financial interest.

Abbreviations

aq: aqueous; Boc, tert-butoxycarbonyl; br, broad; DIAD, diisopropyl azodicarboxylate; DMF, N, N-dimethylformamide; GPCR, G-protein coupled receptor; HBTU, hexafluorophosphate benzotriazole tetramethyl uronium; IC50, half-maximum inhibitory concentration; Ki, inhibition constant; MeCN, acetonitrile; RT, room temperature; SAR, structure–activity relationship; TFA, trifluoroacetic acid; THF, tetrahydrofuran; PPh3, triphenylphosphine.

References

  1. Luedtke, R.R.; Rangel-Barajas, C.; Malik, M.; Reichert, D.E.; Mach, R.H. Bitropic D3 Dopamine Receptor Selective Compounds as Potential Antipsychotics. Curr. Pharm. Des. 2015, 21, 3700–3724. [Google Scholar] [CrossRef]
  2. Strange, P.G. Antipsychotic drug action: Antagonism, inverse agonism or partial agonism. Trends Pharmacol. Sci. 2008, 29, 314–321. [Google Scholar] [CrossRef] [PubMed]
  3. Volkow, N.D.; Fowler, J.S.; Wang, G.J.; Swanson, J.M. Dopamine in drug abuse and addiction: Results from imaging studies and treatment implications. Mol. Psychiatry 2004, 9, 557–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gilbert, J.G.; Newman, A.H.; Gardner, E.L.; Ashby, C.R., Jr.; Heidbreder, C.A.; Pak, A.C.; Peng, X.Q.; Xi, Z.X. Acute administration of SB-277011A, NGB 2904, or BP 897 inhibits cocaine cue-induced reinstatement of drug-seeking behavior in rats: Role of dopamine D3 receptors. Synapse 2005, 57, 17–28. [Google Scholar] [CrossRef] [PubMed]
  5. Sokoloff, P.; Le Foll, B. The dopamine D3 receptor, a quarter century later. Eur. J. Neurosci. 2017, 45, 2–19. [Google Scholar] [CrossRef]
  6. Sokoloff, P.; Giros, B.; Martres, M.P.; Bouthenet, M.L.; Schwartz, J.C. Molecular cloning and characterization of a novel dopamine receptor (D3) as a target for neuroleptics. Nature 1990, 347, 146–151. [Google Scholar] [CrossRef] [PubMed]
  7. Newman, A.H.; Blaylock, B.L.; Nader, M.A.; Bergman, J.; Sibley, D.R.; Skolnick, P. Medication discovery for addiction: Translating the dopamine D3 receptor hypothesis. Biochem. Pharmacol. 2012, 84, 882–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Newman, A.H.; Grundt, P.; Nader, M.A. Dopamine D3 receptor partial agonists and antagonists as potential drug abuse therapeutic agents. J. Med. Chem. 2005, 48, 3663–3679. [Google Scholar] [CrossRef]
  9. You, Z.B.; Gao, J.T.; Bi, G.H.; He, Y.; Boateng, C.; Cao, J.; Gardner, E.L.; Newman, A.H.; Xi, Z.X. The novel dopamine D3 receptor antagonists/partial agonists CAB2-015 and BAK4-54 inhibit oxycodone-taking and oxycodone-seeking behavior in rats. Neuropharmacology 2017, 126, 190–199. [Google Scholar] [CrossRef]
  10. Newman, A.H.; Xi, Z.X.; Heidbreder, C. Current Perspectives on Selective Dopamine D(3) Receptor Antagonists/Partial Agonists as Pharmacotherapeutics for Opioid and Psychostimulant Use Disorders; Springer: Berlin/Heidelberg, Germany, 2022. [Google Scholar]
  11. You, Z.B.; Bi, G.H.; Galaj, E.; Kumar, V.; Cao, J.; Gadiano, A.; Rais, R.; Slusher, B.S.; Gardner, E.L.; Xi, Z.X.; et al. Dopamine D(3)R antagonist VK4-116 attenuates oxycodone self-administration and reinstatement without compromising its antinociceptive effects. Neuropsychopharmacology 2019, 44, 1415–1424. [Google Scholar] [CrossRef]
  12. Boeckler, F.; Gmeiner, P. Dopamine D3 receptor ligands: Recent advances in the control of subtype selectivity and intrinsic activity. Biochim. Biophys. Acta 2007, 1768, 871–887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wang, Q.; Mach, R.H.; Luedtke, R.R.; Reichert, D.E. Subtype selectivity of dopamine receptor ligands: Insights from structure and ligand-based methods. J. Chem. Inf. Model. 2010, 50, 1970–1985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Newman, A.H.; Beuming, T.; Banala, A.K.; Donthamsetti, P.; Pongetti, K.; LaBounty, A.; Levy, B.; Cao, J.; Michino, M.; Luedtke, R.R.; et al. Molecular determinants of selectivity and efficacy at the dopamine D3 receptor. J. Med. Chem. 2012, 55, 6689–6699. [Google Scholar] [CrossRef] [PubMed]
  15. Chien, E.Y.; Liu, W.; Zhao, Q.; Katritch, V.; Han, G.W.; Hanson, M.A.; Shi, L.; Newman, A.H.; Javitch, J.A.; Cherezov, V.; et al. Structure of the human dopamine D3 receptor in complex with a D2/D3 selective antagonist. Science 2010, 330, 1091–1095. [Google Scholar] [CrossRef] [Green Version]
  16. Yuan, J.; Chen, X.; Brodbeck, R.; Primus, R.; Braun, J.; Wasley, J.W.; Thurkauf, A. NGB 2904 and NGB 2849: Two highly selective dopamine D3 receptor antagonists. Bioorg. Med. Chem. Lett. 1998, 8, 2715–2718. [Google Scholar] [CrossRef]
  17. Reavill, C.; Taylor, S.G.; Wood, M.D.; Ashmeade, T.; Austin, N.E.; Avenell, K.Y.; Boyfield, I.; Branch, C.L.; Cilia, J.; Coldwell, M.C.; et al. Pharmacological actions of a novel, high-affinity, and selective human dopamine D(3) receptor antagonist, SB-277011-A. J. Pharmacol. Exp. Ther. 2000, 294, 1154–1165. [Google Scholar]
  18. Mach, R.H.; Huang, Y.; Freeman, R.A.; Wu, L.; Vangveravong, S.; Luedtke, R.R. Conformationally-flexible benzamide analogues as dopamine D3 and sigma 2 receptor ligands. Bioorg. Med. Chem. Lett. 2004, 14, 195–202. [Google Scholar] [CrossRef]
  19. Pilla, M.; Perachon, S.; Sautel, F.; Garrido, F.; Mann, A.; Wermuth, C.G.; Schwartz, J.C.; Everitt, B.J.; Sokoloff, P. Selective inhibition of cocaine-seeking behaviour by a partial dopamine D3 receptor agonist. Nature 1999, 400, 371–375. [Google Scholar] [CrossRef]
  20. Stemp, G.; Ashmeade, T.; Branch, C.L.; Hadley, M.S.; Hunter, A.J.; Johnson, C.N.; Nash, D.J.; Thewlis, K.M.; Vong, A.K.; Austin, N.E.; et al. Design and synthesis of trans-N-[4-[2-(6-cyano-1,2,3, 4-tetrahydroisoquinolin-2-yl)ethyl]cyclohexyl]-4-quinolinecarboxamide (SB-277011): A potent and selective dopamine D(3) receptor antagonist with high oral bioavailability and CNS penetration in the rat. J. Med. Chem. 2000, 43, 1878–1885. [Google Scholar] [CrossRef]
  21. Mach, R.H.; Tu, Z.; Xu, J.; Li, S.; Jones, L.A.; Taylor, M.; Luedtke, R.R.; Derdeyn, C.P.; Perlmutter, J.S.; Mintun, M.A. Endogenous dopamine (DA) competes with the binding of a radiolabeled D(3) receptor partial agonist in vivo: A positron emission tomography study. Synapse 2011, 65, 724–732. [Google Scholar] [CrossRef] [Green Version]
  22. Maramai, S.; Gemma, S.; Brogi, S.; Campiani, G.; Butini, S.; Stark, H.; Brindisi, M. Dopamine D3 Receptor Antagonists as Potential Therapeutics for the Treatment of Neurological Diseases. Front. Neurosci. 2016, 10, 451. [Google Scholar] [CrossRef] [PubMed]
  23. Shaik, A.B.; Kumar, V.; Bonifazi, A.; Guerrero, A.M.; Cemaj, S.L.; Gadiano, A.; Lam, J.; Xi, Z.X.; Rais, R.; Slusher, B.S.; et al. Investigation of Novel Primary and Secondary Pharmacophores and 3-Substitution in the Linking Chain of a Series of Highly Selective and Bitopic Dopamine D3 Receptor Antagonists and Partial Agonists. J. Med. Chem. 2019, 62, 9061–9077. [Google Scholar] [CrossRef] [PubMed]
  24. Lee, B.; Taylor, M.; Griffin, S.A.; McInnis, T.; Sumien, N.; Mach, R.H.; Luedtke, R.R. Evaluation of Substituted N-Phenylpiperazine Analogs as D3 vs. D2 Dopamine Receptor Subtype Selective Ligands. Molecules 2021, 26, 3182. [Google Scholar] [CrossRef] [PubMed]
  25. Bonifazi, A.; Newman, A.H.; Keck, T.M.; Gervasoni, S.; Vistoli, G.; Del Bello, F.; Giorgioni, G.; Pavletic, P.; Quaglia, W.; Piergentili, A. Scaffold Hybridization Strategy Leads to the Discovery of Dopamine D(3) Receptor-Selective or Multitarget Bitopic Ligands Potentially Useful for Central Nervous System Disorders. ACS Chem. Neurosci. 2021, 12, 3638–3649. [Google Scholar] [CrossRef]
  26. Reilly, S.W.; Riad, A.A.; Hsieh, C.J.; Sahlholm, K.; Jacome, D.A.; Griffin, S.; Taylor, M.; Weng, C.C.; Xu, K.; Kirschner, N.; et al. Leveraging a Low-Affinity Diazaspiro Orthosteric Fragment to Reduce Dopamine D3 Receptor (D3R) Ligand Promiscuity across Highly Conserved Aminergic G-Protein-Coupled Receptors (GPCRs). J. Med. Chem. 2019, 62, 5132–5147. [Google Scholar] [CrossRef]
  27. Wassouf, Z.; Schulze-Hentrich, J.M. Alpha-synuclein at the nexus of genes and environment: The impact of environmental enrichment and stress on brain health and disease. J. Neurochem. 2019, 150, 591–604. [Google Scholar] [CrossRef]
  28. Zingales, V.; Torrisi, S.A.; Leggio, G.M.; Bucolo, C.; Drago, F.; Salomone, S. Pharmacological and Genetic Evidence of Dopamine Receptor 3-Mediated Vasoconstriction in Isolated Mouse Aorta. Biomolecules 2021, 11, 418. [Google Scholar] [CrossRef]
  29. Hsieh, C.J.; Riad, A.; Lee, J.Y.; Sahlholm, K.; Xu, K.; Luedtke, R.R.; Mach, R.H. Interaction of Ligands for PET with the Dopamine D3 Receptor: In Silico and In Vitro Methods. Biomolecules 2021, 11, 529. [Google Scholar] [CrossRef]
  30. Micheli, F.; Arista, L.; Bonanomi, G.; Blaney, F.E.; Braggio, S.; Capelli, A.M.; Checchia, A.; Damiani, F.; Di-Fabio, R.; Fontana, S.; et al. 1,2,4-Triazolyl azabicyclo [3.1.0]hexanes: A new series of potent and selective dopamine D(3) receptor antagonists. J. Med. Chem. 2010, 53, 374–391. [Google Scholar] [CrossRef]
  31. Mugnaini, M.; Iavarone, L.; Cavallini, P.; Griffante, C.; Oliosi, B.; Savoia, C.; Beaver, J.; Rabiner, E.A.; Micheli, F.; Heidbreder, C.; et al. Occupancy of brain dopamine D3 receptors and drug craving: A translational approach. Neuropsychopharmacology 2013, 38, 302–312. [Google Scholar] [CrossRef] [Green Version]
  32. Murphy, A.; Nestor, L.J.; McGonigle, J.; Paterson, L.; Boyapati, V.; Ersche, K.D.; Flechais, R.; Kuchibatla, S.; Metastasio, A.; Orban, C.; et al. Acute D3 Antagonist GSK598809 Selectively Enhances Neural Response During Monetary Reward Anticipation in Drug and Alcohol Dependence. Neuropsychopharmacology 2017, 42, 1925–1926. [Google Scholar] [CrossRef] [PubMed]
  33. Micheli, F.; Bacchi, A.; Braggio, S.; Castelletti, L.; Cavallini, P.; Cavanni, P.; Cremonesi, S.; Cin, M.D.; Feriani, A.; Gehanne, S.; et al. 1,2,4-Triazolyl 5-Azaspiro[2.4]heptanes: Lead Identification and Early Lead Optimization of a New Series of Potent and Selective Dopamine D3 Receptor Antagonists. J. Med. Chem. 2016, 59, 8549–8576. [Google Scholar] [CrossRef] [PubMed]
  34. Reilly, S.W.; Griffin, S.; Taylor, M.; Sahlholm, K.; Weng, C.C.; Xu, K.; Jacome, D.A.; Luedtke, R.R.; Mach, R.H. Highly Selective Dopamine D3 Receptor Antagonists with Arylated Diazaspiro Alkane Cores. J. Med. Chem. 2017, 60, 9905–9910. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, J.; Levant, B.; Jiang, C.; Keck, T.M.; Newman, A.H.; Wang, S. Tranylcypromine substituted cis-hydroxycyclobutylnaphthamides as potent and selective dopamine D(3) receptor antagonists. J. Med. Chem. 2014, 57, 4962–4968. [Google Scholar] [CrossRef] [Green Version]
  36. Tan, L.; Zhou, Q.; Yan, W.; Sun, J.; Kozikowski, A.P.; Zhao, S.; Huang, X.P.; Cheng, J. Design and Synthesis of Bitopic 2-Phenylcyclopropylmethylamine (PCPMA) Derivatives as Selective Dopamine D3 Receptor Ligands. J. Med. Chem. 2020, 63, 4579–4602. [Google Scholar] [CrossRef]
  37. Bennacef, I.; Salinas, C.A.; Bonasera, T.A.; Gunn, R.N.; Audrain, H.; Jakobsen, S.; Nabulsi, N.; Weinzimmer, D.; Carson, R.E.; Huang, Y.; et al. Dopamine D3 receptor antagonists: The quest for a potentially selective PET ligand. Part 3: Radiosynthesis and in vivo studies. Bioorg. Med. Chem. Lett. 2009, 19, 5056–5059. [Google Scholar] [CrossRef]
  38. Keck, T.M.; John, W.S.; Czoty, P.W.; Nader, M.A.; Newman, A.H. Identifying Medication Targets for Psychostimulant Addiction: Unraveling the Dopamine D3 Receptor Hypothesis. J. Med. Chem. 2015, 58, 5361–5380. [Google Scholar] [CrossRef] [Green Version]
  39. Appel, N.M.; Li, S.H.; Holmes, T.H.; Acri, J.B. Dopamine D3 Receptor Antagonist (GSK598809) Potentiates the Hypertensive Effects of Cocaine in Conscious, Freely-Moving Dogs. J. Pharmacol. Exp. Ther. 2015, 354, 484–492. [Google Scholar] [CrossRef] [Green Version]
  40. Battiti, F.O.; Cemaj, S.L.; Guerrero, A.M.; Shaik, A.B.; Lam, J.; Rais, R.; Slusher, B.S.; Deschamps, J.R.; Imler, G.H.; Newman, A.H.; et al. The Significance of Chirality in Drug Design and Synthesis of Bitopic Ligands as D3 Receptor (D3R) Selective Agonists. J. Med. Chem. 2019, 62, 6287–6314. [Google Scholar] [CrossRef]
  41. Shaik, A.B.; Boateng, C.A.; Battiti, F.O.; Bonifazi, A.; Cao, J.; Chen, L.; Chitsazi, R.; Ravi, S.; Lee, K.H.; Shi, L.; et al. Structure Activity Relationships for a Series of Eticlopride-Based Dopamine D2/D3 Receptor Bitopic Ligands. J. Med. Chem. 2021, 64, 15313–15333. [Google Scholar] [CrossRef]
  42. Mukherjee, J.; Yang, Z.Y.; Das, M.K.; Brown, T. Fluorinated benzamide neuroleptics--III. Development of (S)-N-[(1-allyl-2-pyrrolidinyl)methyl]-5-(3-[18F]fluoropropyl)-2, 3-dimethoxybenzamide as an improved dopamine D-2 receptor tracer. Nucl. Med. Biol. 1995, 22, 283–296. [Google Scholar] [CrossRef] [PubMed]
  43. Mukherjee, J.; Constantinescu, C.C.; Hoang, A.T.; Jerjian, T.; Majji, D.; Pan, M.L. Dopamine D3 receptor binding of (18)F-fallypride: Evaluation using in vitro and in vivo PET imaging studies. Synapse 2015, 69, 577–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Halldin, C.; Farde, L.; Hogberg, T.; Mohell, N.; Hall, H.; Suhara, T.; Karlsson, P.; Nakashima, Y.; Swahn, C.G. Carbon-11-FLB 457: A radioligand for extrastriatal D2 dopamine receptors. J. Nucl. Med. 1995, 36, 1275–1281. [Google Scholar] [PubMed]
  45. Yang, D.; Kefi, S.; Audinot, V.; Millan, M.J.; Langlois, M. Benzamides derived from 1,2-diaminocyclopropane as novel ligands for human D2 and D3 dopamine receptors. Bioorg. Med. Chem. 2000, 8, 321–327. [Google Scholar] [CrossRef]
  46. Chen, P.J.; Taylor, M.; Griffin, S.A.; Amani, A.; Hayatshahi, H.; Korzekwa, K.; Ye, M.; Mach, R.H.; Liu, J.; Luedtke, R.R.; et al. Design, synthesis, and evaluation of N-(4-(4-phenyl piperazin-1-yl)butyl)-4-(thiophen-3-yl)benzamides as selective dopamine D3 receptor ligands. Bioorg. Med. Chem. Lett. 2019, 29, 2690–2694. [Google Scholar] [CrossRef]
  47. Tu, Z.; Li, S.; Cui, J.; Xu, J.; Taylor, M.; Ho, D.; Luedtke, R.R.; Mach, R.H. Synthesis and pharmacological evaluation of fluorine-containing D(3) dopamine receptor ligands. J. Med. Chem. 2011, 54, 1555–1564. [Google Scholar] [CrossRef] [Green Version]
  48. Rangel-Barajas, C.; Malik, M.; Taylor, M.; Neve, K.A.; Mach, R.H.; Luedtke, R.R. Characterization of [(3) H]LS-3-134, a novel arylamide phenylpiperazine D3 dopamine receptor selective radioligand. J. Neurochem. 2014, 131, 418–431. [Google Scholar] [CrossRef] [Green Version]
  49. Luedtke, R.R.; Freeman, R.A.; Boundy, V.A.; Martin, M.W.; Huang, Y.; Mach, R.H. Characterization of (125)I-IABN, a novel azabicyclononane benzamide selective for D2-like dopamine receptors. Synapse 2000, 38, 438–449. [Google Scholar] [CrossRef]
  50. Wang, T.; Li, Z.; Cvijic, M.E.; Krause, C.; Zhang, L.; Sum, C.S. Measurement of beta-Arrestin Recruitment for GPCR Targets. In Assay Guidance Manual; Markossian, S., Grossman, A., Brimacombe, K., Arkin, M., Auld, D., Austin, C., Baell, J., Chung, T.D.Y., Coussens, N.P., Dahlin, J.L., et al., Eds.; Bethesda: Rockville, MD, USA, 2004. [Google Scholar]
  51. Lefkowitz, R.J.; Shenoy, S.K. Transduction of receptor signals by beta-arrestins. Science 2005, 308, 512–517. [Google Scholar] [CrossRef]
  52. Zhao, X.; Jones, A.; Olson, K.R.; Peng, K.; Wehrman, T.; Park, A.; Mallari, R.; Nebalasca, D.; Young, S.W.; Xiao, S.H. A homogeneous enzyme fragment complementation-based beta-arrestin translocation assay for high-throughput screening of G-protein-coupled receptors. J. Biomol. Screen 2008, 13, 737–747. [Google Scholar] [CrossRef] [Green Version]
  53. Hayatshahi, H.S.; Xu, K.; Griffin, S.A.; Taylor, M.; Mach, R.H.; Liu, J.; Luedtke, R.R. Analogues of Arylamide Phenylpiperazine Ligands To Investigate the Factors Influencing D3 Dopamine Receptor Bitropic Binding and Receptor Subtype Selectivity. ACS Chem. Neurosci. 2018, 9, 2972–2983. [Google Scholar] [CrossRef] [PubMed]
  54. Besnard, J.; Ruda, G.F.; Setola, V.; Abecassis, K.; Rodriguiz, R.M.; Huang, X.P.; Norval, S.; Sassano, M.F.; Shin, A.I.; Webster, L.A.; et al. Automated design of ligands to polypharmacological profiles. Nature 2012, 492, 215–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chu, W.; Tu, Z.; McElveen, E.; Xu, J.; Taylor, M.; Luedtke, R.R.; Mach, R.H. Synthesis and in vitro binding of N-phenyl piperazine analogs as potential dopamine D3 receptor ligands. Bioorg. Med. Chem. 2005, 13, 77–87. [Google Scholar] [CrossRef] [PubMed]
  56. Vangveravong, S.; Zhang, Z.; Taylor, M.; Bearden, M.; Xu, J.; Cui, J.; Wang, W.; Luedtke, R.R.; Mach, R.H. Synthesis and characterization of selective dopamine D(2) receptor ligands using aripiprazole as the lead compound. Bioorg. Med. Chem. 2011, 19, 3502–3511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Xu, J.; Vangveravong, S.; Li, S.; Fan, J.; Jones, L.A.; Cui, J.; Wang, R.; Tu, Z.; Chu, W.; Perlmutter, J.S.; et al. Positron emission tomography imaging of dopamine D2 receptors using a highly selective radiolabeled D2 receptor partial agonist. Neuroimage 2013, 71, 168–174. [Google Scholar] [CrossRef] [Green Version]
  58. Peng, X.; Wang, Q.; Mishra, Y.; Xu, J.; Reichert, D.E.; Malik, M.; Taylor, M.; Luedtke, R.R.; Mach, R.H. Synthesis, pharmacological evaluation and molecular modeling studies of triazole containing dopamine D3 receptor ligands. Bioorg. Med. Chem. Lett. 2015, 25, 519–523. [Google Scholar] [CrossRef] [Green Version]
  59. Vilkman, H.; Kajander, J.; Aalto, S.; Vahlberg, T.; Nagren, K.; Allonen, T.; Syvalahti, E.; Hietala, J. The effects of lorazepam on extrastriatal dopamine D(2/3)-receptors-A double-blind randomized placebo-controlled PET study. Psychiatry Res. 2009, 174, 130–137. [Google Scholar] [CrossRef]
  60. Lober, S.; Hubner, H.; Tschammer, N.; Gmeiner, P. Recent advances in the search for D3- and D4-selective drugs: Probes, models and candidates. Trends Pharmacol. Sci. 2011, 32, 148–157. [Google Scholar] [CrossRef]
  61. Gao, M.; Wang, M.; Mock, B.H.; Glick-Wilson, B.E.; Yoder, K.K.; Hutchins, G.D.; Zheng, Q.H. An improved synthesis of dopamine D2/D3 receptor radioligands [11C]fallypride and [18F]fallypride. Appl. Radiat. Isot. 2010, 68, 1079–1086. [Google Scholar] [CrossRef]
  62. Pettit, G.R.; Piatak, D.M. Hydrogen Bromide-Acetic Acid Demethylation of 2,3-Dimethoxy-6-bromobenzoic Acid. An Example of Concomitant Bromine Migration1,2. J. Org. Chem. 1960, 25, 721–725. [Google Scholar] [CrossRef]
  63. Komiya, C.; Aihara, K.; Morishita, K.; Ding, H.; Inokuma, T.; Shigenaga, A.; Otaka, A. Development of an Intein-Inspired Amide Cleavage Chemical Device. J. Org. Chem. 2016, 81, 699–707. [Google Scholar] [CrossRef] [PubMed]
  64. Varseev, G.N.; Maier, M.E. A novel palladium-catalyzed arylation-dehydroaromatization reaction: Synthesis of 7-aryltetralones. Org. Lett. 2005, 7, 3881–3884. [Google Scholar] [CrossRef]
  65. O'Boyle, N.M.; Banck, M.; James, C.A.; Morley, C.; Vandermeersch, T.; Hutchison, G.R. Open Babel: An open chemical toolbox. J. Cheminform. 2011, 3, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Lee, J.; Cheng, X.; Swails, J.M.; Yeom, M.S.; Eastman, P.K.; Lemkul, J.A.; Wei, S.; Buckner, J.; Jeong, J.C.; Qi, Y.; et al. CHARMM-GUI Input Generator for NAMD, GROMACS, AMBER, OpenMM, and CHARMM/OpenMM Simulations Using the CHARMM36 Additive Force Field. J. Chem. Theory Comput. 2016, 12, 405–413. [Google Scholar] [CrossRef] [PubMed]
  68. Jo, S.; Kim, T.; Iyer, V.G.; Im, W. CHARMM-GUI: A web-based graphical user interface for CHARMM. J. Comput. Chem. 2008, 29, 1859–1865. [Google Scholar] [CrossRef] [PubMed]
  69. Kim, S.; Lee, J.; Jo, S.; Brooks, C.L., 3rd; Lee, H.S.; Im, W. CHARMM-GUI ligand reader and modeler for CHARMM force field generation of small molecules. J. Comput. Chem. 2017, 38, 1879–1886. [Google Scholar] [CrossRef]
  70. Klauda, J.B.; Venable, R.M.; Freites, J.A.; O'Connor, J.W.; Tobias, D.J.; Mondragon-Ramirez, C.; Vorobyov, I.; MacKerell, A.D., Jr.; Pastor, R.W. Update of the CHARMM all-atom additive force field for lipids: Validation on six lipid types. J. Phys. Chem. B 2010, 114, 7830–7843. [Google Scholar] [CrossRef] [Green Version]
  71. Venable, R.M.; Sodt, A.J.; Rogaski, B.; Rui, H.; Hatcher, E.; MacKerell, A.D., Jr.; Pastor, R.W.; Klauda, J.B. CHARMM all-atom additive force field for sphingomyelin: Elucidation of hydrogen bonding and of positive curvature. Biophys. J. 2014, 107, 134–145. [Google Scholar] [CrossRef] [Green Version]
  72. Lomize, M.A.; Pogozheva, I.D.; Joo, H.; Mosberg, H.I.; Lomize, A.L. OPM database and PPM web server: Resources for positioning of proteins in membranes. Nucleic Acids Res. 2012, 40, D370–D376. [Google Scholar] [CrossRef]
  73. Case, D.B.-S.I.; Brozell, S.; Cerutti, D.; Cheatham III, T.; Cruzeiro, V.; Darden, T.; Duke, R.; Ghoreishi, D.; Gilson, M.; Gohlke, H.; et al. AMBER 18; University of California: San Francisco, CA, USA, 2018. [Google Scholar]
  74. Durrant, J.D.; McCammon, J.A. BINANA: A novel algorithm for ligand-binding characterization. J. Mol. Graph. Model. 2011, 29, 888–893. [Google Scholar] [CrossRef] [PubMed]
Scheme 1. Synthesis of 3-fluoropropyl or bromo analogs 6ad. Reagents and conditions: (a) tert-butyl (2-aminoethyl) ethylcarbamate, HBTU, DIPEA, DMF, RT, 24 h; (b) TFA, CH2Cl2, 0 °C to RT, 1 h; (c) 2-(2-bromoethyl)-1,3-dioxolane or 2-(3-bromopropyl)-1,3-dioxolane, Na2CO3, MeCN, 65 °C, 72 h; (d) aq 4 N HCl, THF, RT, 3 h; (e) dimethylamine, sodium triacetoxy borohydride, dichloroethane, RT, 16 h.
Scheme 1. Synthesis of 3-fluoropropyl or bromo analogs 6ad. Reagents and conditions: (a) tert-butyl (2-aminoethyl) ethylcarbamate, HBTU, DIPEA, DMF, RT, 24 h; (b) TFA, CH2Cl2, 0 °C to RT, 1 h; (c) 2-(2-bromoethyl)-1,3-dioxolane or 2-(3-bromopropyl)-1,3-dioxolane, Na2CO3, MeCN, 65 °C, 72 h; (d) aq 4 N HCl, THF, RT, 3 h; (e) dimethylamine, sodium triacetoxy borohydride, dichloroethane, RT, 16 h.
Ijms 24 00432 sch001
Scheme 2. Synthesis of 9a,b or 11a,b for the SBS interactions. Reagents and conditions: (a) N-(3-bromopropyl)phthalimide or N-(4-bromobutyl)phthalimide, K2CO3, DMF, 65 °C, 16 h; (b) hydrazine hydrate, EtOH, 75 °C, 3 h; (c) 4-(thiophen-2-yl)benzoic acid, SOCl2, 3 h, then, 8a or 8b, CH2Cl2, RT, 16 h; (d) NaB(OAc)3, RT, 16 h; (e) 4-methyl-5-phenyl-4H-1,2,4-triazole-3-thiol, DIAD, PPh3, THF, RT, 24 h.
Scheme 2. Synthesis of 9a,b or 11a,b for the SBS interactions. Reagents and conditions: (a) N-(3-bromopropyl)phthalimide or N-(4-bromobutyl)phthalimide, K2CO3, DMF, 65 °C, 16 h; (b) hydrazine hydrate, EtOH, 75 °C, 3 h; (c) 4-(thiophen-2-yl)benzoic acid, SOCl2, 3 h, then, 8a or 8b, CH2Cl2, RT, 16 h; (d) NaB(OAc)3, RT, 16 h; (e) 4-methyl-5-phenyl-4H-1,2,4-triazole-3-thiol, DIAD, PPh3, THF, RT, 24 h.
Ijms 24 00432 sch002
Scheme 3. Synthesis of 12 and 14 to determine the capability of the OBS binding. Reagents and conditions: (a) CH3I, K2CO3, acetone, reflux, 16 h; (b) SOCl2, CH2Cl2, RT, 3 h, then, 3-bromopropylamine hydrobromide, RT, 16 h (c) N-methylethanamine, K2CO3, DMF, 65 °C, 30 min.
Scheme 3. Synthesis of 12 and 14 to determine the capability of the OBS binding. Reagents and conditions: (a) CH3I, K2CO3, acetone, reflux, 16 h; (b) SOCl2, CH2Cl2, RT, 3 h, then, 3-bromopropylamine hydrobromide, RT, 16 h (c) N-methylethanamine, K2CO3, DMF, 65 °C, 30 min.
Ijms 24 00432 sch003
Scheme 4. Synthesis of 20ac for different substituents on tert-amine. Reagents and conditions: (a) (1) ethyl trifluoroacetate, CH2Cl2, 0 °C to RT, 1 h; (2) [for 15a] allyl bromide, Et3N, MeOH, RT, 16 h, then, (Boc)2O, 4 h; [for 15b] 4-fluorobenzaldehyde, NaB(OAc)3, then, (Boc)2O, 4 h; (3) K2CO3, MeOH/H2O (9:1), reflux, 2 h (b) 15a or 15b, HBTU, DIPEA, DMF, RT, 24 h; (c) TFA, CH2Cl2, 0 °C to RT, 1 h; (d) N-(3-bromopropyl)phthalimide, K2CO3, DMF, 65 °C, 16 h; (e) hydrazine hydrate, EtOH, 75 °C, 2 h; (f) [for 20a or 20c] 4-(thiophen-2-yl)benzoic acid, SOCl2, 3 h, then, 19a or 19b, CH2Cl2, RT, 16 h; (g) [for 20b] 13, K2CO3, DMF, 65 °C, 16 h.
Scheme 4. Synthesis of 20ac for different substituents on tert-amine. Reagents and conditions: (a) (1) ethyl trifluoroacetate, CH2Cl2, 0 °C to RT, 1 h; (2) [for 15a] allyl bromide, Et3N, MeOH, RT, 16 h, then, (Boc)2O, 4 h; [for 15b] 4-fluorobenzaldehyde, NaB(OAc)3, then, (Boc)2O, 4 h; (3) K2CO3, MeOH/H2O (9:1), reflux, 2 h (b) 15a or 15b, HBTU, DIPEA, DMF, RT, 24 h; (c) TFA, CH2Cl2, 0 °C to RT, 1 h; (d) N-(3-bromopropyl)phthalimide, K2CO3, DMF, 65 °C, 16 h; (e) hydrazine hydrate, EtOH, 75 °C, 2 h; (f) [for 20a or 20c] 4-(thiophen-2-yl)benzoic acid, SOCl2, 3 h, then, 19a or 19b, CH2Cl2, RT, 16 h; (g) [for 20b] 13, K2CO3, DMF, 65 °C, 16 h.
Ijms 24 00432 sch004
Scheme 5. Synthesis of different aryl carboxamides 21aj. Reagents and conditions: (a) [for 21a] 2-naphthoyl chloride, Et3N, CH2Cl2, RT, 1 h; [for 21b to 21j] aryl carboxylic acids, SOCl2, 3 h, then, 8a, CH2Cl2, RT, 16 h.
Scheme 5. Synthesis of different aryl carboxamides 21aj. Reagents and conditions: (a) [for 21a] 2-naphthoyl chloride, Et3N, CH2Cl2, RT, 1 h; [for 21b to 21j] aryl carboxylic acids, SOCl2, 3 h, then, 8a, CH2Cl2, RT, 16 h.
Ijms 24 00432 sch005
Figure 1. Representative poses of MDS for (a) 9a, (b) 21c, (c) 20a, (d) 20b, and (e) 20c. The predicted interactions of each compound with residues in the OBS and the SBS of the D3 receptor were distinguished by the color. Red: hydrogen; cyan: π-interactions; green: halogen bond.
Figure 1. Representative poses of MDS for (a) 9a, (b) 21c, (c) 20a, (d) 20b, and (e) 20c. The predicted interactions of each compound with residues in the OBS and the SBS of the D3 receptor were distinguished by the color. Red: hydrogen; cyan: π-interactions; green: halogen bond.
Ijms 24 00432 g001
Figure 2. Summary of frequency of all contacts between selected ligands and residues in the D3 receptor binding sites (the OBS and SBS).
Figure 2. Summary of frequency of all contacts between selected ligands and residues in the D3 receptor binding sites (the OBS and SBS).
Ijms 24 00432 g002
Table 1. Binding affinities and potency of R1 and R2 modified analogs with different length of the alkyl linker a.
Table 1. Binding affinities and potency of R1 and R2 modified analogs with different length of the alkyl linker a.
Ijms 24 00432 i001
CmpdR1R2nKi ± SEM (nM) bD2/D3IC50
(nM) c
Imax
(% Control) d
D3D2
6aIjms 24 00432 i002Ijms 24 00432 i00321763 ± 9424564 ± 24452.6>100092.7 ± 5.4
6bIjms 24 00432 i004Ijms 24 00432 i00531236 ± 4382810 ± 5712.3430 ± 46798.4 ± 2.9
6cIjms 24 00432 i006Ijms 24 00432 i00721627 ± 1655416 ± 10433.3>100098.0 ± 5.6
6dIjms 24 00432 i008Ijms 24 00432 i0093141 ± 26449 ± 903.2152 ± 18594.0 ± 6.4
9aIjms 24 00432 i010Ijms 24 00432 i01111.0 ± 0.01169 ± 416914.0 ± 7.467.3 ± 18.9
9bIjms 24 00432 i012Ijms 24 00432 i01328.0 ± 0.8103 ± 1412.9104 ± 13670.8 ± 7.3
11aIjms 24 00432 i014Ijms 24 00432 i0152125 ± 2342 ± 232.7NT eNT
11bIjms 24 00432 i016Ijms 24 00432 i017322.2 ± 1.889.7 ± 3.64.6NTNT
a All compounds were converted to HCl salts prior to tests. b mean ± SEM; mean Ki ± SEM values were measured using [125I]IABN in D2 or D3 receptors highly expressed HEK cells. The radioligand binding assay was performed by three individual experiments. c The potency for D3 receptors was expressed as mean ± SD by three individual experiments. d Imax was obtained from a percentage of the maximum inhibition of a dopamine at EC80 concentration in the same assay. e NT; not tested.
Table 2. Binding affinities of different sized substituents on the tert-amine a.
Table 2. Binding affinities of different sized substituents on the tert-amine a.
Ijms 24 00432 i018
CmpdR3Ki ± SEM (nM) bD2/D3IC50
(nM) c
Imax
(% Control) d
D3D2
9aIjms 24 00432 i0191.0 ± 0.01169 ± 416914.0 ± 7.467.3 ± 18.9
20aIjms 24 00432 i0202.7 ± 0.4259 ± 2311026.5 ± 12.988.1 ± 20.4
20bIjms 24 00432 i0215.8 ± 1.0243 ± 1546.751.6 ± 40.8100.6 ± 7.9
20cIjms 24 00432 i022299 ± 101574 ± 1701.9NT eNT
a All compounds were converted to HCl salts prior to tests. b mean ± SEM; mean Ki ± SEM values were measured using [125I]IABN in D2 or D3 receptors highly expressed HEK cells. The radioligand binding assay was performed by three individual experiments. c The potency for D3 receptors was expressed as mean ± SD by three individual experiments. d Imax was obtained from a percentage of the maximum inhibition of a dopamine at EC80 concentration in the same assay. e NT; not tested.
Table 3. R3 optimization based on carboxamide moieties a.
Table 3. R3 optimization based on carboxamide moieties a.
Ijms 24 00432 i023
CmpdArKi ± SEM (nM) bD2/D3IC50
(nM) c
Imax
(% Control) d
cLogP e
D3D2
21aIjms 24 00432 i0241.2 ± 0.2169 ± 1214116.4 ± 7.766.1 ± 17.64.12
21bIjms 24 00432 i02513.2 ± 0.5525 ± 7239.819.6 ± 23.775.2 ± 4.83.21
21cIjms 24 00432 i0261.1 ± 0.1107 ± 597.31.3 ± 1.078.2 ± 18.03.47
21dIjms 24 00432 i0270.8 ± 0.2148 ± 9180.59.3 ± 12.057.7 ± 26.92.72
21eIjms 24 00432 i0282.8 ± 0.5142 ± 1650.74.3 ± 2.585.4 ± 6.02.41
21fIjms 24 00432 i0296.1 ± 0.6327 ± 3253.72.7 ± 0.685.7 ± 13.11.79
21gIjms 24 00432 i0302.5 ± 0.3312 ± 1812536.8 ± 35.750.1 ± 20.94.78
21hIjms 24 00432 i03111.2 ± 1.4248 ± 1922.14.1 ± 2.173.8 ± 14.93.41
21iIjms 24 00432 i0327.0 ± 0.5304 ± 743.42.7 ± 0.299.6 ± 20.73.05
21jIjms 24 00432 i0333.1 ± 0.5192 ± 2261.94.8 ± 0.881.5 ± 10.12.96
a All compounds were converted to HCl salts prior to tests. b mean ± SEM; mean Ki ± SEM values were measured using [125I]IABN in D2 or D3 receptors highly expressed HEK cells. The radioligand binding assay was performed by three individual experiments. c The potency for the D3 receptor was expressed as mean ± SD by three individual experiments. d Imax was obtained from a percentage of the maximum inhibition of a dopamine at EC80 concentration in the same assay. e NT; not tested.
Table 4. Molecular docking and MDS results of selected analogs.
Table 4. Molecular docking and MDS results of selected analogs.
CmpdDockingMDS
Distance to ASP110 (Å)Binding Energy (kcal/mol)Ligand RMSD (Å)
fallypride a2.7−7.712.08 ± 0.33
9a2.6−9.743.18 ± 0.54
20a2.9−10.112.18 ± 0.74
20b2.8−10.002.29 ± 0.60
20c2.9−10.223.00 ± 0.82
21c2.7−10.102.45 ± 0.49
a Values were obtained from the previous study [29].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, H.Y.; Lee, J.Y.; Hsieh, C.-J.; Taylor, M.; Luedtke, R.R.; Mach, R.H. Design and Synthesis of Conformationally Flexible Scaffold as Bitopic Ligands for Potent D3-Selective Antagonists. Int. J. Mol. Sci. 2023, 24, 432. https://doi.org/10.3390/ijms24010432

AMA Style

Kim HY, Lee JY, Hsieh C-J, Taylor M, Luedtke RR, Mach RH. Design and Synthesis of Conformationally Flexible Scaffold as Bitopic Ligands for Potent D3-Selective Antagonists. International Journal of Molecular Sciences. 2023; 24(1):432. https://doi.org/10.3390/ijms24010432

Chicago/Turabian Style

Kim, Ho Young, Ji Youn Lee, Chia-Ju Hsieh, Michelle Taylor, Robert R. Luedtke, and Robert H. Mach. 2023. "Design and Synthesis of Conformationally Flexible Scaffold as Bitopic Ligands for Potent D3-Selective Antagonists" International Journal of Molecular Sciences 24, no. 1: 432. https://doi.org/10.3390/ijms24010432

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop