Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Res Sq ; 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38196573

ABSTRACT

Although aminergic GPCRs are the target for ~25% of approved drugs, developing subtype selective drugs is a major challenge due to the high sequence conservation at their orthosteric binding site. Bitopic ligands are covalently joined orthosteric and allosteric pharmacophores with the potential to boost receptor selectivity, driven by the binding of the secondary pharmacophore to non-conserved regions of the receptor. Although bitopic ligands have great potential to improve current medications by reducing off-target side effects, the lack of structural information on their binding mode impedes rational design. Here we determine the cryo-EM structure of the hD3R coupled to a GO heterotrimer and bound to the D3R selective bitopic agonist FOB02-04A. Structural, functional and computational analyses provide new insights into its binding mode and point to a new TM2-ECL1-TM1 region, which requires the N-terminal ordering of TM1, as a major determinant of subtype selectivity in aminergic GPCRs. This region is underexploited in drug development, expands the established secondary binding pocket in aminergic GPCRs and could potentially be used to design novel and subtype selective drugs.

2.
J Med Chem ; 64(20): 15313-15333, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34636551

ABSTRACT

The crystal structure of the dopamine D3 receptor (D3R) in complex with eticlopride inspired the design of bitopic ligands that explored (1) N-alkylation of the eticlopride's pyrrolidine ring, (2) shifting of the position of the pyrrolidine nitrogen, (3) expansion of the pyrrolidine ring system, and (4) incorporation of O-alkylations at the 4-position. Structure activity relationships (SAR) revealed that moving the N- or expanding the pyrrolidine ring was detrimental to D2R/D3R binding affinities. Small pyrrolidine N-alkyl groups were poorly tolerated, but the addition of a linker and secondary pharmacophore (SP) improved affinities. Moreover, O-alkylated analogues showed higher binding affinities compared to analogously N-alkylated compounds, e.g., O-alkylated 33 (D3R, 0.436 nM and D2R, 1.77 nM) vs the N-alkylated 11 (D3R, 6.97 nM and D2R, 25.3 nM). All lead molecules were functional D2R/D3R antagonists. Molecular models confirmed that 4-position modifications would be well-tolerated for future D2R/D3R bioconjugate tools that require long linkers and or sterically bulky groups.


Subject(s)
Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/metabolism , Salicylamides/pharmacology , Dose-Response Relationship, Drug , Humans , Ligands , Models, Molecular , Molecular Structure , Salicylamides/chemical synthesis , Salicylamides/chemistry , Structure-Activity Relationship
3.
J Med Chem ; 64(21): 16088-16105, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34699207

ABSTRACT

Linkers are emerging as a key component in regulating the pharmacology of bitopic ligands directed toward G-protein coupled receptors (GPCRs). In this study, the role of regio- and stereochemistry in cyclic aliphatic linkers tethering well-characterized primary and secondary pharmacophores targeting dopamine D2 and D3 receptor subtypes (D2R and D3R, respectively) is described. We introduce several potent and selective D2R (rel-trans-16b; D2R Ki = 4.58 nM) and D3R (rel-cis-14a; D3R Ki = 5.72 nM) agonists while modulating subtype selectivity in a stereospecific fashion, transferring D2R selectivity toward D3R via inversion of the stereochemistry around these cyclic aliphatic linkers [e.g., (-)-(1S,2R)-43 and (+)-(1R,2S)-42]. Pharmacological observations were supported with extensive molecular docking studies. Thus, not only is it an innovative approach to modulate the pharmacology of dopaminergic ligands described, but a new class of optically active cyclic linkers are also introduced, which can be used to expand the bitopic drug design approach toward other GPCRs.


Subject(s)
Dopamine Agonists/pharmacology , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D3/drug effects , Dopamine Agonists/chemistry , HEK293 Cells , Humans , Ligands , Molecular Docking Simulation , Radioligand Assay , Stereoisomerism
4.
J Med Chem ; 64(11): 7778-7808, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34011153

ABSTRACT

The need for safer pain-management therapies with decreased abuse liability inspired a novel drug design that retains µ-opioid receptor (MOR)-mediated analgesia, while minimizing addictive liability. We recently demonstrated that targeting the dopamine D3 receptor (D3R) with highly selective antagonists/partial agonists can reduce opioid self-administration and reinstatement to drug seeking in rodent models without diminishing antinociceptive effects. The identification of the D3R as a target for the treatment of opioid use disorders prompted the idea of generating a class of ligands presenting bitopic or bivalent structures, allowing the dual-target binding of the MOR and D3R. Structure-activity relationship studies using computationally aided drug design and in vitro binding assays led to the identification of potent dual-target leads (23, 28, and 40), based on different structural templates and scaffolds, with moderate (sub-micromolar) to high (low nanomolar/sub-nanomolar) binding affinities. Bioluminescence resonance energy transfer-based functional studies revealed MOR agonist-D3R antagonist/partial agonist efficacies that suggest potential for maintaining analgesia with reduced opioid-abuse liability.


Subject(s)
Dopamine Antagonists/chemistry , Ligands , Receptors, Dopamine D3/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/therapeutic use , Animals , Binding Sites , Biphenyl Compounds/chemistry , Biphenyl Compounds/metabolism , Biphenyl Compounds/therapeutic use , Disease Models, Animal , Dopamine Antagonists/metabolism , Dopamine Antagonists/therapeutic use , Drug Design , Fluorescence Resonance Energy Transfer , Mice , Molecular Docking Simulation , Opioid-Related Disorders/drug therapy , Pain/drug therapy , Pain Management , Receptors, Dopamine D3/agonists , Receptors, Dopamine D3/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Structure-Activity Relationship
5.
Biomolecules ; 11(4)2021 04 13.
Article in English | MEDLINE | ID: mdl-33924613

ABSTRACT

The dopamine D2/D3 receptor (D2R/D3R) agonists are used as therapeutics for Parkinson's disease (PD) and other motor disorders. Selective targeting of D3R over D2R is attractive because of D3R's restricted tissue distribution with potentially fewer side-effects and its putative neuroprotective effect. However, the high sequence homology between the D2R and D3R poses a challenge in the development of D3R selective agonists. To address the ligand selectivity, bitopic ligands were designed and synthesized previously based on a potent D3R-preferential agonist PF592,379 as the primary pharmacophore (PP). This PP was attached to various secondary pharmacophores (SPs) using chemically different linkers. Here, we characterize some of these novel bitopic ligands at both D3R and D2R using BRET-based functional assays. The bitopic ligands showed varying differences in potencies and efficacies. In addition, the chirality of the PP was key to conferring improved D3R potency, selectivity, and G protein signaling bias. In particular, compound AB04-88 exhibited significant D3R over D2R selectivity, and G protein bias at D3R. This bias was consistently observed at various time-points ranging from 8 to 46 min. Together, the structure-activity relationships derived from these functional studies reveal unique pharmacology at D3R and support further evaluation of functionally biased D3R agonists for their therapeutic potential.


Subject(s)
Dopamine Agonists/pharmacology , Receptors, Dopamine D3/metabolism , Aminopyridines/chemistry , Aminopyridines/pharmacology , Binding Sites , Dopamine Agonists/chemical synthesis , Energy Transfer , HEK293 Cells , Humans , Luminescence , Morpholines/chemistry , Morpholines/pharmacology , Protein Binding , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/chemistry , Stereoisomerism , Structure-Activity Relationship
6.
ACS Med Chem Lett ; 11(10): 1956-1964, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33062179

ABSTRACT

In this study, starting from our selective D3R agonist FOB02-04A (5), we investigated the chemical space around the linker portion of the molecule via insertion of a hydroxyl substituent and ring-expansion of the trans-cyclopropyl moiety into a trans-cyclohexyl scaffold. Moreover, to further elucidate the importance of the primary pharmacophore stereochemistry in the design of bitopic ligands, we investigated the chiral requirements of (+)-PD128907 ((+)-(4a R ,10b R )-2)) by synthesizing and resolving bitopic analogues in all the cis and trans combinations of its 9-methoxy-3,4,4a,10b-tetrahydro-2H,5H-chromeno[4,3-b][1,4] oxazine scaffold. Despite the lack of success in obtaining new analogues with improved biological profiles, in comparison to our current leads, a "negative" result due to a poor or simply not improved biological profile is fundamental toward better understanding chemical space and optimal stereochemistry for target recognition. Herein, we identified essential structural information to understand the differences between orthosteric and bitopic ligand-receptor binding interactions, discriminate D3R active and inactive states, and assist multitarget receptor recognition. Exploring stereochemical complexity and developing extended D3R SAR from this new library complements previously described SAR and inspires future structural and computational biology investigation. Moreover, the expansion of chemical space characterization for D3R agonism may be utilized in machine learning and artificial intelligence (AI)-based drug design, in the future.

7.
J Med Chem ; 63(5): 1779-1797, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31499001

ABSTRACT

The genesis of designing bivalent or bitopic molecules that engender unique pharmacological properties began with Portoghese's work directed toward opioid receptors, in the early 1980s. This strategy has evolved as an attractive way to engineer highly selective compounds for targeted G-protein coupled receptors (GPCRs) with optimized efficacies and/or signaling bias. The emergence of X-ray crystal structures of many GPCRs and the identification of both orthosteric and allosteric binding sites have provided further guidance to ligand drug design that includes a primary pharmacophore (PP), a secondary pharmacophore (SP), and a linker between them. It is critical to note the synergistic relationship among all three of these components as they contribute to the overall interaction of these molecules with their receptor proteins and that strategically designed combinations have and will continue to provide the GPCR molecular tools of the future.


Subject(s)
Chemistry, Pharmaceutical/methods , Drug Design , Drug Discovery/methods , Receptors, G-Protein-Coupled/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Chemistry, Pharmaceutical/trends , Drug Discovery/trends , Humans , Receptors, Dopamine/chemistry , Receptors, Dopamine/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, Opioid/chemistry , Receptors, Opioid/metabolism
8.
J Med Chem ; 62(13): 6287-6314, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31257877

ABSTRACT

Because of the large degree of homology among dopamine D2-like receptors, discovering ligands capable of discriminating between the D2, D3, and D4 receptor subtypes remains a significant challenge. Previous work has exemplified the use of bitopic ligands as a powerful strategy in achieving subtype selectivity for agonists and antagonists alike. Inspired by the potential for chemical modification of the D3 preferential agonists (+)-PD128,907 (1) and PF592,379 (2), we synthesized bitopic structures to further improve their D3R selectivity. We found that the (2S,5S) conformation of scaffold 2 resulted in a privileged architecture with increased affinity and selectivity for the D3R. In addition, a cyclopropyl moiety incorporated into the linker and full resolution of the chiral centers resulted in lead compound 53 and eutomer 53a that demonstrate significantly higher D3R binding selectivities than the reference compounds. Moreover, the favorable metabolic stability in rat liver microsomes supports future studies in in vivo models of dopamine system dysregulation.


Subject(s)
Dopamine Agonists/chemistry , Dopamine Agonists/metabolism , Indoles/chemistry , Indoles/metabolism , Receptors, Dopamine D3/metabolism , Animals , Dopamine Agonists/chemical synthesis , Drug Design , HEK293 Cells , Humans , Indoles/chemical synthesis , Ligands , Microsomes, Liver/metabolism , Rats , Stereoisomerism , Structure-Activity Relationship
9.
Eur J Med Chem ; 168: 461-473, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30844609

ABSTRACT

A series of derivatives obtained by moving the aromatic moiety on the 1,4-dioxane ring of compounds 1-3 from position 6 to position 2 or 3 was prepared and evaluated for the affinity for 5-HT1A receptor (5-HT1AR) and α1-adrenoceptor (α1-AR) subtypes. Moreover, the flexible 2-ethanolamine linker of the most interesting compounds was replaced by the more conformationally constrained piperazine ring. In vitro functional studies performed on derivatives showing the highest affinities for 5-HT1AR highlighted that the shifting of the diphenyl moiety of derivatives 2 and 13 from position 6 to position 3 of the 1,4-dioxane nucleus, affording 11 and 16, respectively, modulated the 5-HT1AR functional profile from agonism to antagonism. Docking simulations, performed on the human 5-HT1AR, further rationalized the biological results, delving into the features which modulate the shift between agonist and antagonist activity. Interestingly, compound 11, endowed with mixed 5-HT1AR/α1d-AR antagonist profile, showed antiproliferative and cytotoxic effects on both PC-3 and DU-145 prostate cancer cell lines higher than those of the α1d-AR antagonist 2 and the 5-HT1AR antagonist 16. The experiments performed in the presence of the endogenous agonists norepinephrine and serotonin confirmed the involvement of both receptor systems in the antitumor activity of 11.


Subject(s)
Antineoplastic Agents/pharmacology , Dioxanes/pharmacology , Prostatic Neoplasms/drug therapy , Receptor, Serotonin, 5-HT1A/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dioxanes/chemical synthesis , Dioxanes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Male , Models, Molecular , Molecular Structure , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...