Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Aug 15.
Published in final edited form as: Bioorg Med Chem Lett. 2020 Jun 4;30(16):127305. doi: 10.1016/j.bmcl.2020.127305

Synthesis and dopamine receptor pharmacological evaluations on ring C ortho halogenated 1-phenyl-benzazepines

Rajan Giri a,b, Hari K Namballa a, Ananta Sarker a, Ian Alberts c, Wayne W Harding a,b,d,*
PMCID: PMC7340849  NIHMSID: NIHMS1602201  PMID: 32631525

Abstract

A series of 1-phenyl-benzazepines containing bromine or chlorine substituents at the ortho position of the appended phenyl ring (2’-monosubstituted or 2’,6’- disubstituted patterns) were synthesized and evaluated for affinity towards dopamine D1R, D2R and D5R. As is typical of the 1-phenyl-benzazepine scaffold, the compounds displayed selectivity towards D1R and D5R; analogs generally lacked affinity for D2R. Interestingly, 2’,6’-dichloro substituted analogs showed modest D5R versus D1R selectivity whereas this selectivity was reversed in compounds with a 2’-halo substitution pattern. Compound 10a was identified as a D1R antagonist (Ki = 14 nM; IC50 = 9.4 nM).

Keywords: Dopamine, D1, D5, D2, Benzazepine

Graphical Abstract

graphic file with name nihms-1602201-f0001.jpg


The neurotransmitter dopamine is implicated in a number of physiological functions in both the periphery and central nervous system (CNS) such as locomotion, blood pressure regulation, cognition and emotion.17 Perturbations in dopaminergic neurotransmission underlie some CNS disorders such as Parkinson’s disease, schizophrenia and drug abuse.811 Therefore normalization of dopaminergic neurotransmission with pharmacological agents has been explored as a means to treat these conditions.

Dopamine exerts its pharmacological actions via agonist activity at 5 dopamine receptors (D1R – D5R). Dopamine receptors are classified as “D1-like” (constituted by D1 and D5 receptor sub-types – D1R and D5R) and “D2-like” (comprising D2R, D3R and D4R) based on the structure and function of the receptors and pharmacological studies.1215 The discovery of ligands that are highly selective for either the D1R or D5R sub-types has proved challenging due to the close transmembrane structural similarity between D1R and D5R (>80% homology in transmembrane regions). Thus, commercially available D1R ligands (i.e. “D1R-like” ligands) usually display similar affinity at D5R. It is only quite recently that D1R subtype selective ligands have been disclosed.16, 17 The availability of highly selective ligands for either D1R or D5R is of current interest as such compounds would be useful tools to unravel the individual roles of D1R and D5R in various physiological processes and serve as lead molecules for related CNS disorders.

The 1-phenylbenzazepine framework is a classical template for D1R-like ligands and numerous compounds along a continuum of functional activity (i.e. full agonists, partial agonists, antagonists) have been identified with this scaffold.1823 A number of these compounds are used as research tools in pharmacological studies. For example, SCH 23390 (1) is a widely used D1R-like antagonist tool; it displays very high affinity for D1R and D5R (0.2 and 0.3 nM respectively).24 SKF 38393 (2) is a widely used D1R-like agonist with strong affinity for D1R and D5R (1.0 and 0.5 nM respectively).25 Fenoldopam (3), a peripherally restricted D1R-like partial agonist is currently the only compound from this class that is in use clinically (it is used as a fast-acting anti-hypertensive drug).26

There have been several structure-activity relationship (SAR) studies on 1-phenylbenzazepines as D1R-like receptor ligands and these studies have established that the aryl substituent groups as well as the nitrogen substituent can significantly impact D1R-like affinity, D1R-like selectivity versus “D2-like” receptors as well as functional activity.2732 Although several SAR studies have been performed on the scaffold, it has not been determined how halogen substituents in the ortho position of ring C in the 1-phenyl-benzazepine framework impacts affinity and selectivity for dopamine receptors; there is no data available concerning the D1R versus D5R affinity of the compounds. Given this gap in the SAR of 1-phenyl-benzazepines, we set out to examine the role of ring C ortho halo substituents on D1R/D5R affinity in this scaffold. We hypothesized that such substituents might cause differences in receptor interactions with the substituents themselves and/or lead to modified conformations of the molecule as a whole that could directly influence D1R and D5R affinity and selectivity. Thus, we set out to synthesize a set of 1-phenylbenzazepine derivatives with variations in the ring A moiety (either a catechol or protected catechol motif), nitrogen alkyl group substituent and ring C ortho halogenated motif.

The analogs were synthesized as shown in Scheme 1. The epoxides 9 were available commercially or could be readily synthesized from the corresponding styrenes 8. We initially attempted reaction of amine 4 and epoxides 9 to form amino alcohols 5 without the use of any additives/catalysts, as is typically done for the synthesis of 1-phenyl-benzazepine syntheses,3335 but found the reactions to be low yielding. Presumably this less than favorable outcome was due to the steric hindrance in the epoxides used. Lewis acids or lithium salts have often been used to promote ring opening of epoxides with amine nucleophiles.3638 As LiNTf2 was reported to provide high yields in such reactions,39 we examined the use of this reagent and were happy to find that the reactions of 4 and 9 to form 5 proceeded in reasonable yield. Acid-catalyzed cyclization of 5 afforded the benzazepine framework of compounds 6a-d. Compounds 6a-d served as key intermediates from which the synthesis diverged to prepare other analogs with variations in the phenolic moiety and/or N-substituent.

Thus, treatment of compounds 6b with BBr3 gave the catecholic compound 7. The secondary amine in compounds 6b-d was methylated via reductive amination to give analogs 10a-c respectively. Treatment of compounds 6b-d with allyl bromide allowed for the preparation of N-allylated analogs 11a-c. Compounds 10c and 11c in turn were demethylated by reaction with BBr3 affording catechols 12 and 13 respectively.

The binding affinity of compounds 6a-d, 7, 10a-c, 11a-c, 12 and 13 were assessed at dopamine D1, D2 and D5 receptors. Data for these assessments are presented in Table 1 as Ki values in nM. Compounds 6a-d had D1R affinities ranging from 72 to 147 nM. Affinities at D5R for this sub-group of compounds were slightly lower overall (ranging from 82 to 483 nM), so that on a whole, compounds 6a-d were slightly more selective for D1R over D5R. Compound 6c is interesting as it is the only compound of the 6a-6d subset that was D5R selective over D1R (3-fold). Compounds 6a, 6b and 6d displayed modest selectivity for D1R over D5R (up to 3-fold).

Table 1.

Binding affinity of analogs at D1R, D2R and D5R

graphic file with name nihms-1602201-t0002.jpg
Ki(nM)a
Cmpd # R R1 X X1 D1b D2C D5d
6a Me H H H 126.4 ± 9.1 nae 309.9 ± 23
6b Me H H Cl 147.4 ± 4.1 na 482.8 ± 29.4
6c Me H Cl Cl 76.1 ± 3.6 na 25.4 ± 2.6
6d Me H H Br 72 ± 5.6 na 82 ± 6.2
7 H H H Br 26 ± 3.1 na 67 ±7.1
10a Me Me H C1 14 ± 2.3 na 46.4 ± 3.8
10b Me Me Cl Cl 144.6 ± 9.0 na 49.1 ± 3.7
10c Me Me H Br 16 ± 1.4 na 47 ± 3.2
11a Me Allyl H Cl 48.3 ± 8.2 na 264.4 ± 13.6
11b Me Allyl Cl Cl 1044 ± 59.2 1507.8 ± 89.2 479.1 ± 39.2
11c Me Allyl H Br 41 ± 2.8 na 501 ±64
12 H Me H Br 59.4 ± 4.9 na 223.2 ± 18.6
13 H Allyl H Br 132.9 ± 8.9 na 442.2 ± 56
(+)− 4.04 ± 0.2
Butaclamol
Haloperidol 5.58 ± 0.3
SKF 83566 3.95 ± 0.2
a

Experiments carried out in triplicate;

b

[3H]SCH23390 used as radioligand;

c

[3H]N-methylspiperone used as radioligand;

d

[3H]SCH23390 used as radioligand;

e

na – not active (<50% inhibition in a primary assay when tested at 10μM).

Compound 7 with a bromo substituent, showed strong D1R and D5R affinity. In comparing phenol 7 to its C7 methoxy analog 6d, it is apparent that the effect of cleavage of the C7 methoxy group to give the catechol 7, results in a roughly 3-fold increase in D1R affinity; improvement in affinity at D5R was more modest.

Compounds 10a-c are the N-methylated analogs of 6b-d respectively; compounds 10a-c as a group displayed stronger D1R affinity than their N-des-methyl counterparts. Compounds 10a and 10c had the strongest D1R affinity of any compound evaluated for this study (Ki of 14 and 16 nM for 10a and 10c respectively). The compounds in this sub-group with mono-halogen substituents (10a and 10c) showed modest D1R selectivity over D5R (3-fold and 4-fold respectively), but this selectivity was reversed in the dichloro substituted compound 10b.

Similar trends as for the N-methylated analogs 10a-c were seen for the N-allylated analogs 11a-c. In that regard, both the mono-halo substituted analogs 11a and 11c were D1R selective whereas the dichloro substituted analog was D5R selective. In general, the N-allyl analogs displayed lower affinity for D1R and D5R than their N-methyl congeners.

The catecholic analogs 12 and 13 are the O-demethylated analogs of 10c and 11c respectively; both 12 and 13 showed lower D1R and D5R affinity than their methylated precursors and both also had modest D1R selectivity versus D5R (in the 3- to 4-fold range).

In analysis of the effect of monohalogenated versus dihalogenated substitutions in the pendant aryl ring, interesting observations emerge. In the case of the 6b/6c pair, the dihalogenated compound 6c showed higher D1R and D5R affinities than the monohalogenated congener 6b. However, a similar change in the 10a/10b pair and the 11a/11b pair resulted in diminished D1R and D5R affinities for the corresponding dihalogenated analogues. This result indicates that the presence of an N-alkyl substituent is more favorable for binding of the monohalogenated versus their dihalogenated congeners at D1R/D5R, whereas absence of such a substituent leads to a stronger preference towards binding of the dihalogenated versus monohalogenated variants.

Comparison of data for the 6b/6d, 10a/10c and 11a/11c compound pairs enabled an analysis of the effect of monochloro versus monobromo substitution in the analogue series. In the case of the 6b/6d pair, the bromo analogue 6d had higher D1R and D5R affinity than the chloro analogue 6b. However, in the case of the 10a/10c and 11a/11c pairs, changing from a chloro group to a bromo group did not result in a similar increase in affinity of the brominated analogues for the D1R as was seen for 6b/6d; affinities for the chloro and bromo variants were similar (e.g. 14 nM and 16 nM for 10a and 10c respectively at D1R). Meanwhile, at the D5R, affinities of the bromo and chloro analogues were similar (for 10a/10c) or were worse for the bromo analogue (for 11a/11c). Therefore, it appears that in this series, the presence of an N-alkyl substituent group does not lead to a strong preference for binding of the monobromo versus monochloro variants at D1R; however, the absence of an N-alkyl substituent leads to stronger binding of the monobromo versus monochloro congeners at D1R.

We selected the compound with the highest D1-like receptor affinity for further evaluation of functional activity. Thus, compound 10a was evaluated for agonist and antagonist activity in D1R assays that measured cAMP modulation by Eurofins Lead Hunter Discovery Services. As expected, (based on structural similarity to 3) 10a displayed strong antagonist activity in these assays (IC50 = 9.4 nM for 10a; IC50 of positive control SCH 39166 = 1.5 nM). No agonist activity was detected for 10a.

In order to provide insights into the important receptor-ligand interactions between the ortho halogen substituted 1-phenyl-benzapines and the D1R and D5R, computational docking studies were conducted for the series of analogs in Table 1. In this context, we explored the docked ligand poses and identified key interactions that have a significant impact on binding to the dopamine receptors for this ligand series. These efforts focused mainly on the compounds 7, 10a and 10c, which displayed the best experimental binding affinities to D1R.

Homology models of D1R and D5R were generated and utilized in the docking studies. The D1R homology model was constructed from the high-resolution crystal structure of the human β2-adrenergic G protein-coupled receptor (GPCR) with pdb code 2RH1 followed by induced fit docking with several halogenated 1-phenyl-benzazepine analogs.40 In a similar manner, the D5R homology model was created from the high-resolution crystal structure of the β1-adrenergic GPCR with pdb code 6H7J followed by induced fit docking with the benzazepine analogs.41 Models of appropriate amino acid backbone and side-chain orientations in the ligand binding site. The homology model building procedure involved application of the Schrödinger Prime Structure Prediction, Induced Fit Docking and Glide software tools in conjunction with manual intervention to support the formation of known key receptor-ligand interactions. The docking runs of the 1-phenyl-benzazepine analogs into the D1R and D5R binding sites utilized the Schrödinger Glide methodology in Standard Precision (SP) mode. Using this approach, the Glidescore scoring function provided an estimate of the ligand binding affinities for the highest ranked poses of the ligand series in the D1R and D5R targets. The binding poses for the compounds 7, 10a and 10c (docked as the R enantiomers), which gave the best D1R experimental affinities, are depicted in Figures 2A and 2B.

Figure 2.

Figure 2.

Docked poses of compounds 7 (blue carbon atoms), 10a (green carbon atoms) and 10c (pink carbon atoms) in A - the D1R target and B - the D5R target. The receptor targets are depicted by secondary structure elements and grey carbon atoms for select residues. Key quaternary N – Asp salt bridges are depicted by the pink dashed lines, H-bonding interactions by the yellow dashed lines, aromatic H-bonding by the turquoise dashed lines and π-π stacking by the blue dashed lines. Docking studies were performed with the R enantiomers.

Compounds 7, 10a and 10c give very similar docked poses in the D1R binding pocket as shown in Figure 2A with binding energies in the range −7.8 kcal/mol to −8.2 kcal/mol. The docked poses display the quaternary N - Asp103 salt bridge, H-bonding interactions of the ligand hydroxyl group to the Asn292 side chain, and for compound 7 with an additional hydroxyl group in the catechol moiety there is also a H-bond to the Ser198 sidechain, as well as an aromatic H-bond involving the pendant phenyl group and Ser188.

In the D5R binding site, the main receptor-ligand interactions for the docked poses of compounds 7, 10a and 10c comprise the quaternary N - Asp120 salt bridge, hydrogen bonding interactions of the ligand hydroxyl group to the Asn316 or Ser229 sidechain, and again for compound 7 there is another hydrogen bond with its second catechol hydroxyl group to Ser233, as well as π-π hydrophobic interactions involving the ligand aromatic rings with Phe312 and Trp116. Compounds 10a and 10c form docked poses with binding energies of −8.2 kcal/mol and −8.1 kcal/mol, respectively, which are very similar to those in the D1R binding pocket.

Overall, the computationally predicted binding energies for the docked series of halogen substituted 1-phenyl-benzapine derivatives in Table 1 are similar in both the D1R and D5R structures or a little better in D5R as a consequence of the slightly stronger hydrogen bonding and π-π hydrophobic interactions. In this context, the docking scores do not align with the selectivity trends derived from the experimental binding affinities. This is at least partially attributable to the modest nature of the observed D1R/D5R experimental selectivities of the ring C halogenated analogs. Furthermore, D1R and D5R are very similar structurally in the ligand binding pocket, which provides justification for the close computational binding energies for most of the compounds in these two target sites. The docking outcomes for the compounds in Table 1 involved the R enantiomers whereas the affinity data were obtained with racemic mixtures and this could also have an impact on the match between the experimental and computational results. Docking simulations were investigated with the S enantiomers, however, they generated similar trends compared to the R enantiomers with, in general, slightly worse predicted binding energies in both the D1R and D5R targets.

In conclusion, this study extends the available SAR information on 1-phenyl-benzazepines as D1R-like ligands with regards to the effect of ring C ortho halogen substituents. As is evident from examination of the data, the compounds in this study maintain selectivity for D1-like receptors over D2R, with modest selectivity for either D1R or D5R. As compared to known 1-phenyl-benzapeine D1R-like tools such as 1 and 2, it is apparent that the ortho halogen group does not significantly improve D1R or D5R affinity. However, one of the findings from this work is that compounds with di-ortho-halo substituents (i.e. C2’/C6’ substitution) favor binding to D5R, whereas compounds with a mono-ortho-halo (C2’) substituent favor D1R binding over D5R. In addition, the SAR data suggests that the most favorable outcome for good D1R affinity is to have either mono-ortho-halogenation in tandem with N-alkyl substitution or di-orthohalogenation without N-alkyl substitution.

Evaluation of the functional activity of 10a reaffirms the idea that an 8-hydroxy-7-methoxy moiety favors antagonist rather than agonist activity. This result is in line with the generally accepted view that a catechol motif is required for agonist activity in the 1-phenyl-benzazepine scaffold.

Our molecular docking studies revealed interactions that were relevant for affinity of the molecules at D1R and D5R, but were unable to resolve interactions necessary for the observed modest D1R sub-type selectivity of 7, 10a and 10c. Examination of larger sets of compounds with ortho halogenated patterns in future, including enantiopure analogs, may provide a larger body of data to aid in the challenging optimization of these ligands towards D1R or D5R potency and sub-type selectivity.

Supplementary Material

1

Figure 1.

Figure 1.

Structures of typical 1-phenylbenzazepine D1R-like ligands – Fenoldopam, SCH 23390 and SKF 38393.

Reagents and Conditions:

Reagents and Conditions:

(a) 1. mCPBA, DCM, rt, 12h; 2. NaOH, rt, (88 – 96%); (b) 9, LiNTf2, THF, reflux, 24 h, (56 −76)%; (c) 1. TFA, H2SO , rt, 5h; 2. NaOAc, (43 – 54%); (d) BBr3 , DCM, 0 °C, 4 h, (56 – 80%); (e) HCHO, Na(OAc)3BH, ACN, rt, 12 h, (23 – 48%); (f) Allyl Bromide, TEA, ACN, rt, 16 h, (56 – 64%).

Acknowledgments

This publication was made possible by Grant Number 1SC1DA049961-01 from the National Institutes of Health (NIH). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NIH or its divisions. Ki determinations, and receptor binding profiles were generously provided by the National Institute of Mental Health’s Psychoactive Drug Screening Program, Contract # HHSN-271-2008-00025-C (NIMH PDSP). The NIMH PDSP is directed by Bryan L. Roth MD, PhD at the University of North Carolina at Chapel Hill and Project Officer Jamie Driscol at NIMH, Bethesda MD, USA. For experimental details please refer to the PDSP website http://pdsp.med.unc.edu/ and click on “Binding Assay” or “Functional Assay” on the menu bar.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declaration of interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  • 1.Bell C Endogenous renal dopamine and control of blood pressure. Clin Exp Hypertens A. 1987;9(5–6): 955–975. [DOI] [PubMed] [Google Scholar]
  • 2.Caruana MP, Heber M, Brigden G, Raftery EB. Effects of fenoldopam, a specific dopamine receptor agonist, on blood pressure and left ventricular function in systemic hypertension. Br J Clin Pharmacol. 1987;24(6): 721–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Eilam D, Clements KV, Szechtman H. Differential effects of D1 and D2 dopamine agonists on stereotyped locomotion in rats. Behav Brain Res. 1991;45(2): 117–124. [DOI] [PubMed] [Google Scholar]
  • 4.Ashby FG, Valentin VV, von Meer SS. Differential effects of dopamine-directed treatments on cognition. Neuropsychiatr Dis Treat. 2015;11: 1859–1875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Robbins TW. Dopamine and cognition. Curr Opin Neurol. 2003;16 Suppl 2: S1–2. [DOI] [PubMed] [Google Scholar]
  • 6.Nieoullon A, Coquerel A. Dopamine: a key regulator to adapt action, emotion, motivation and cognition. Curr Opin Neurol. 2003;16 Suppl 2: S3–9. [PubMed] [Google Scholar]
  • 7.Schwartz J The dopaminergic system in the periphery. J Pharmacol. 1984;15(4): 401–414. [PubMed] [Google Scholar]
  • 8.Sohur US, Gray DL, Duvvuri S, Zhang Y, Thayer K, Feng G. Phase 1 Parkinson’s Disease Studies Show the Dopamine D1/D5 Agonist PF-06649751 is Safe and Well Tolerated. Neurol Ther. 2018;7(2): 307–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Szasz JA, Viorelia C, Mihaly I, et al. [Dopamine agonists in Parkinson’s disease therapy - 15 years of experience of the Neurological Clinics from Tirgu Mures. A cross-sectional study]. Ideggyogy Sz. 2019;72(5–6): 187–193. [DOI] [PubMed] [Google Scholar]
  • 10.Perez de la Mora M, Hernandez-Mondragon C, Crespo-Ramirez M, Rejon-Orantes J, Borroto-Escuela DO, Fuxe K. Conventional and Novel Pharmacological Approaches to Treat Dopamine-Related Disorders: Focus on Parkinson’s Disease and Schizophrenia. Neuroscience. 2019. [DOI] [PubMed] [Google Scholar]
  • 11.Solinas M, Belujon P, Fernagut PO, Jaber M, Thiriet N. Dopamine and addiction: what have we learned from 40 years of research. J Neural Transm (Vienna). 2019;126(4): 481–516. [DOI] [PubMed] [Google Scholar]
  • 12.Beaulieu JM, Espinoza S, Gainetdinov RR. Dopamine receptors - IUPHAR Review 13. Br J Pharmacol. 2015;172(1): 1–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Beaulieu JM, Gainetdinov RR. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol Rev. 2011;63(1): 182–217. [DOI] [PubMed] [Google Scholar]
  • 14.Missale C, Nash SR, Robinson SW, Jaber M, Caron MG. Dopamine receptors: from structure to function. Physiol Rev. 1998;78(1): 189–225. [DOI] [PubMed] [Google Scholar]
  • 15.Vallone D, Picetti R, Borrelli E. Structure and function of dopamine receptors. Neurosci Biobehav Rev. 2000;24(1): 125–132. [DOI] [PubMed] [Google Scholar]
  • 16.Felsing DE, Jain MK, Allen JA. Advances in Dopamine D1 Receptor Ligands for Neurotherapeutics. Curr Top Med Chem. 2019. [DOI] [PubMed] [Google Scholar]
  • 17.Hall A, Provins L, Valade A. Novel Strategies To Activate the Dopamine D1 Receptor: Recent Advances in Orthosteric Agonism and Positive Allosteric Modulation. J Med Chem. 2019;62(1): 128–140. [DOI] [PubMed] [Google Scholar]
  • 18.Zhang J, Xiong B, Zhen X, Zhang A. Dopamine D1 receptor ligands: where are we now and where are we going. Med Res Rev. 2009;29(2): 272–294. [DOI] [PubMed] [Google Scholar]
  • 19.Neumeyer JL, Kula NS, Bergman J, Baldessarini RJ. Receptor affinities of dopamine D1 receptor-selective novel phenylbenzazepines. Eur J Pharmacol. 2003;474(2–3): 137–140. [DOI] [PubMed] [Google Scholar]
  • 20.Wu WL, Burnett DA, Spring R, et al. Dopamine D1/D5 receptor antagonists with improved pharmacokinetics: design, synthesis, and biological evaluation of phenol bioisosteric analogues of benzazepine D1/D5 antagonists. J Med Chem. 2005;48(3): 680–693. [DOI] [PubMed] [Google Scholar]
  • 21.Breese GR, Criswell HE, McQuade RD, Iorio LC, Mueller RA. Pharmacological evaluation of SCH-12679: evidence for an in vivo antagonism of D1-dopamine receptors. J Pharmacol Exp Ther. 1990;252(2): 558–567. [PubMed] [Google Scholar]
  • 22.Kaiser C, Dandridge PA, Garvey E, et al. Absolute stereochemistry and dopaminergic activity of enantiomers of 2,3,4,5-tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine. J Med Chem. 1982;25(6): 697–703. [DOI] [PubMed] [Google Scholar]
  • 23.Andersen PH, Jansen JA. Dopamine receptor agonists: selectivity and dopamine D1 receptor efficacy. Eur J Pharmacol. 1990;188(6): 335–347. [DOI] [PubMed] [Google Scholar]
  • 24.Bourne JA. SCH 23390: the first selective dopamine D1-like receptor antagonist. CNS Drug Rev. 2001;7(4): 399–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Seeman P, Van Tol HH. Dopamine receptor pharmacology. Trends Pharmacol Sci. 1994;15(7): 264–270. [DOI] [PubMed] [Google Scholar]
  • 26.Brogden RN, Markham A. Fenoldopam: a review of its pharmacodynamic and pharmacokinetic properties and intravenous clinical potential in the management of hypertensive urgencies and emergencies. Drugs. 1997;54(4): 634–650. [DOI] [PubMed] [Google Scholar]
  • 27.Zhang J, Huang J, Song Z, et al. Structural manipulation on the catecholic fragment of dopamine D1 receptor agonist 1-phenyl-N-methyl-benzazepines. Eur J Med Chem. 2014;85: 16–26. [DOI] [PubMed] [Google Scholar]
  • 28.Zhang J, Chen X, Yu L, Zhen X, Zhang A. Synthesis of 6-substituted 1-phenylbenzazepines and their dopamine D1 receptor activities. Bioorg Med Chem. 2008;16(21): 9425–9431. [DOI] [PubMed] [Google Scholar]
  • 29.Pfeiffer FR, Wilson JW, Weinstock J, et al. Dopaminergic activity of substituted 6-chloro-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepines. J Med Chem. 1982;25(4): 352–358. [DOI] [PubMed] [Google Scholar]
  • 30.Neumeyer JL, Baindur N, Niznik HB, Guan HC, Seeman P. (+/−)-3-Allyl-6-bromo-7,8-dihydroxy-1-phenyl-2,3,4,5-tetrahydro-1H-3- benzazepin, a new high-affinity D1 dopamine receptor ligand: synthesis and structure-activity relationship. J Med Chem. 1991;34(12): 3366–3371. [DOI] [PubMed] [Google Scholar]
  • 31.Ross ST, Franz RG, Gallagher G Jr., et al. Dopamine agonists related to 3-allyl-6-chloro-2,3,4,5-tetrahydro-1-(4-hydroxyphenyl)-1H-3-benzaz epi ne-7, 8-diol. 6-Position modifications. J Med Chem. 1987;30(1): 35–40. [DOI] [PubMed] [Google Scholar]
  • 32.Weinstock J, Ladd DL, Wilson JW, et al. Synthesis and renal vasodilator activity of some dopamine agonist 1-aryl-2,3,4,5-tetrahydro-1H-3-benzazepine-7,8-diols: halogen and methyl analogues of fenoldopam. J Med Chem. 1986;29(11): 2315–2325. [DOI] [PubMed] [Google Scholar]
  • 33.Tan ES, Miyakawa M, Bunzow JR, Grandy DK, Scanlan TS. Exploring the Structure-Activity Relationship of the Ethylamine Portion of 3-Iodothyronamine for Rat and Mouse Trace Amine-Associated Receptor 1. J Med Chem. 2007;50(12): 2787–2798. [DOI] [PubMed] [Google Scholar]
  • 34.Baindur N, Tran M, Niznik HB, Guan HC, Seeman P, Neumeyer JL. (±)-3-Allyl-7-halo-8-hydroxy-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepines as selective high affinity D1 dopamine receptor antagonists: synthesis and structure-activity relationship. J Med Chem. 1992;35(1): 67–72. [DOI] [PubMed] [Google Scholar]
  • 35.Ross ST, Franz RG, Gallagher G JR, et al. Dopamine agonists related to 3-allyl-6-chloro-2,3,4,5-tetrahydro-1-(4-hydroxyphenyl)-1H-3-benzazepine-7,8-diol, 6-position modifications. J Med Chem. 1987;30(1): 35–40. [DOI] [PubMed] [Google Scholar]
  • 36.Auge J, Leroy F. Lithium trifluoromethanesulfonate-catalyzed aminolysis of oxiranes. Tetrahedron Lett. 1996;37(43): 7715–7716. [Google Scholar]
  • 37.Sekar G, Singh VK. An Efficient Method for Cleavage of Epoxides with Aromatic Amines. J Org Chem. 1999;64(1): 287–289. [DOI] [PubMed] [Google Scholar]
  • 38.Chini M, Crotti P, Favero L, Macchia F, Pineschi M. Lanthanide(III) trifluoromethanesulfonates as extraordinarily effective new catalysts for the aminolysis of 1,2-epoxides. Tetrahedron Lett. 1994;35(3): 433–436. [Google Scholar]
  • 39.Cossy J, Bellosta V, Hamoir C, Desmurs J-R. Regioselective ring opening of epoxides by nucleophiles mediated by lithium bistrifluoromethanesulfonimide. Tetrahedron Lett. 2002;43(39): 7083–7086. [Google Scholar]
  • 40.Cherezov V, Rosenbaum DM, Hanson MA, et al. High-resolution crystal structure of an engineered human beta2-adrenergic G protein-coupled receptor. Science. 2007;318(5854): 1258–1265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Warne T, Edwards PC, Dore AS, Leslie AGW, Tate CG. Molecular basis for high-affinity agonist binding in GPCRs. Science. 2019;364(6442): 775–778. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES