Research paper
Novel androgen receptor antagonist identified by structure-based virtual screening, structural optimization, and biological evaluation

https://doi.org/10.1016/j.ejmech.2020.112156Get rights and content

Highlights

  • A novel hit (C18) with IC50 of 2.4 μM against AR transcriptional activity in LNCaP cell was identified through SBVS.

  • Optimization of C18 resulted in the discovery of a more potent AR antagonist (AT2) with 16-fold improved anti-AR potency.

  • AT2 could effectively inhibit the transcriptional function of AR and block the nuclear translocation of AR.

Abstract

Androgen receptor (AR) plays important roles in the development of prostate cancer (PCa), and therefore it has been regarded as the most important therapeutic target for both hormone-sensitive prostate cancer (HSPC) and advanced PCa. In this study, a novel hit (C18) with IC50 of 2.4 μM against AR transcriptional activity in LNCaP cell was identified through structure-based virtual screening based on molecular docking and free energy calculations. The structure-activity relationship analysis and structural optimization of C18 resulted in the discovery of a structural analogue (AT2), a more potent AR antagonist with 16-fold improved anti-AR potency. Further assays indicated that AT2 was capable of effectively inhibiting the transcriptional function of AR and blocking the nuclear translocation of AR like the second-generation AR antagonists. The antagonists discovered in this study may be served as the promising lead compounds for the development of AR-driven PCa therapeutics.

Introduction

Prostate cancer (PCa) is the most diagnosed cancer among men worldwide [1]. Androgen receptor (AR) is a member of the nuclear receptor superfamily of ligand-activated transcription factor. Abnormal activation of the AR signaling pathway plays a pivotal role in the development and progression of PCa [2,3]. Currently, a vital approach to prevent the excessive activation of androgens is the treatment with AR antagonists to block the binding of androgens to AR [2,3]. Androgen deprivation therapy (ADT), the most important treatment for advanced PCa, reduces the levels of androgen production by surgical or pharmacological castration. Unfortunately, most patients usually develop into castration-resistant prostate cancer (CRPC) after 2 years of ADT treatment. The possible mechanisms of CRPC include AR mutations, AR amplifications, and active AR splice variants [4]. In general, existing studies show both PCa and CRPC are closely related to AR [[5], [6], [7]].

Current clinically used first-generation or second-generation AR antagonists, such as R-bicalutamide and Enzalutamide (Enz), have achieved great success against androgen-dependent PCa and improved the survival rate of PCa patients [2,8]. However, after the initially effective response, efficacy of these AR antagonists is suffered from the rapid emergence of drug resistance [[9], [10], [11], [12]]. One of the main reasons for drug resistance is the acquired point mutations at the AR ligand-binding pocket (LBP). An explanation for this reason is that some point mutations in the AR LBP, such as W741L, W741C, and T877 A, would create a more spacious LBP, thus converting these antagonists to agonists and inducing drug resistance [[13], [14], [15]]. A widely used approach to deal with this situation in drug design is to modify the original antagonists into larger chemical structures [[16], [17], [18], [19]]. Nevertheless, in general, larger molecules often confront several problems associated with unfavorable permeability and physiological distribution [20]. Therefore, it is still urgent to discover new AR antagonists with novel scaffolds to improve clinical outcomes.

A common approach to design antagonists with novel scaffolds is to use virtual screening (VS), which has also been used to identify novel hits of AR. For instance, the AR antagonist 6-(3,4-dihydro-1H-isoquinolin-2-yl)-N-(6-methylpyridin-2-yl)nicotinamide (DIMN) was screened out by structure-based VS (SBVS) based on the crystal structure of the AR-metribolone complex [21]. Another AR antagonist 5,5a,6,10b-tetrahydroindeno[2,1-b]indole (VPC-12060) was discovered by SBVS and ligand-based virtual screening (LBVS) [22]. Wang et al. identified pyrazolopyramidine analogs as novel potent AR antagonists also by combining SBVS and LBVS [23]. Recently, we reported the identification of a series of novel AR ligands, including AR agonists and antagonists, through an integrated strategy by combining SBVS based on the crystal AR structures in complex with its agonists [24]. It has been proved that SBVS is potent to discover novel AR antagonists.

Molecular docking has been recognized as the most popular method for structure-based drug design [[25], [26], [27]]. It can predict the binding conformations of ligands to the target, and rank the ligands by scoring functions [[28], [29], [30]]. Compared with most scoring functions in molecular docking, the Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) method can achieve a better balance between identifying the binding poses and predicting the binding free energies, and therefore it has been widely used in SBVS and lead optimization [[31], [32], [33]]. In recent years, variable dielectric MM/GBSA (VD-MM/GBSA), a novel modified MM/GBSA algorithm, has attracted increasing attention [[34], [35], [36], [37]].

In this study, a multi-step SBVS strategy based on molecular docking and VD-MM/GBSA rescoring was employed to screen the Specs database, and 32 compounds were finally purchased for bioassay verification. Three of them exhibited strong bioactivities, and the competitive ligand binding assay showed that 1 compound (C18) was targeting the LBP of AR. Structural optimization based on molecular dynamics (MD) simulation and structure-activity relationship (SAR) analysis was then applied to the hit of C18, which resulted in the discovery of AT2, a more promising AR antagonist with 16 folds improved anti-AR potency relative to C18. Moreover, AT2 displayed much better anti-proliferative effects than Enz in three androgen independent cell lines, including PC3, C4-2, and DU145. The qPCR and immunofluorescence assays illustrated that AT2 can inhibit AR transcriptional activity and block nuclear translocation of AR. Our study provided valuable clues for the development of a novel class of AR therapeutic antagonists to combat PCa.

Section snippets

Evaluation of candidate compounds and discovery of hit C18

The schematic workflow of the SBVS protocol used in this study is presented in Fig. 1. By applying molecular docking, VD-MM/GBSA rescoring and structural clustering, a number of potential AR antagonists were identified. As a result, a total of 32 potential active candidates were purchased and submitted to bioassays. At first, the LNCaP-ARR2PB-eGFP-based transcriptional activity assay was performed to determine the antagonistic activities of the screened compounds at the concentration of 10 μM [

Conclusion

In this study, the SBVS strategy based on molecular docking and VD-MM/GBSA rescoring was employed to discover novel AR antagonists against PCa. A novel hit (C18) with promising anti-AR activities in a group of bioassays was identified. The subsequent structural optimization focusing on the change of the spatial conformations and hydrophobicity leads to a more potent AR antagonist (AT2) without toxicity at its effective concentration against AR. Besides, AT2 could efficiently antagonize AR

General synthetic procedure for compounds CT1∼CT6

A mixture of 2-naphthylamine (1, 0.02 mol) and appropriate benzaldehyde (2a-c, 0.022 mol) in dry dichloromethane was stirred under reflux overnight. After completion of the reaction, the mixture was concentrated to give appropriate Schiff base 3a-c, which were used directly without further purification. Then a mixture of 3a-c (0.02 mol), 3,4-dihydro-2H-pyran or 2-norbornylene (0.022 mol) and indium trichloride (0.002 mol) in dichloromethane/acetonitrile (1:1, 20 mL) was stirred at room

Declaration of competing interest

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.

Acknowledgments

This study was supported by National Key R&D Program of China (2016YFA0202900), Key R&D Program of Zhejiang Province (2020C03010), National Natural Science Foundation of China (21575128, 81773632), and Zhejiang Provincial Natural Science Foundation of China (LZ19H300001).

References (49)

  • W. Zhou et al.

    Discovery of novel androgen receptor ligands by structure-based virtual screening and bioassays

    Dev. Reprod. Biol.

    (2018)
  • S.I. Virtanen et al.

    Case-specific performance of MM-PBSA, MM-GBSA, and SIE in virtual screening

    J. Mol. Graph. Model.

    (2015)
  • H. Borgmann et al.

    Moving towards precision urologic oncology: targeting enzalutamide-resistant prostate cancer and mutated forms of the androgen receptor using the novel inhibitor darolutamide (ODM-201)

    Eur. Urol.

    (2018)
  • F. Bray et al.

    Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries

    Ca - Cancer J. Clin.

    (2018)
  • D. Li et al.

    A magic drug target: androgen receptor

    Med. Res. Rev.

    (2019)
  • N. Elshan et al.

    Molecules targeting the androgen receptor (AR) signaling axis beyond the AR-Ligand binding domain

    Med. Res. Rev.

    (2019)
  • C. Cai et al.

    Intratumoral androgen biosynthesis in prostate cancer pathogenesis and response to therapy

    Endocr. Relat. Canc.

    (2011)
  • M.H. Tan et al.

    Androgen receptor: structure, role in prostate cancer and drug discovery

    Acta Pharmacol. Sin.

    (2015)
  • D. Leveque

    Enzalutamide in metastatic prostate cancer

    N. Engl. J. Med.

    (2019)
  • P.A. Watson et al.

    Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer

    Nat. Rev. Canc.

    (2015)
  • J.D. Joseph et al.

    A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509

    Canc. Discov.

    (2013)
  • T. Hara et al.

    Novel mutations of androgen receptor: a possible mechanism of bicalutamide withdrawal syndrome

    Canc. Res.

    (2003)
  • M.S. Ozers et al.

    The androgen receptor T877A mutant recruits LXXLL and FXXLF peptides differently than wild-type androgen receptor in a time-resolved fluorescence resonance energy transfer assay

    Biochemistry

    (2007)
  • K. Steketee et al.

    Broadened ligand responsiveness of androgen receptor mutants obtained by random amino acid substitution of H874 and mutation hot spot T877 in prostate cancer

    Int. J. Canc.

    (2002)
  • Cited by (11)

    • Discovery of novel non-steroidal selective glucocorticoid receptor modulators by structure- and IGN-based virtual screening, structural optimization, and biological evaluation

      2022, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Then cell nuclei were visualized by DAPI staining (Beyotime, China; # C1005). Images were recorded and analyzed as previously reported [43]. RAW264.7 macrophages cells were pretreated with 10 μM indicated compounds and after 1 h later, cells were stimulated with 20 ng/mL LPS.

    • Inhibitory potential of a benzoxazole derivative, 4FI against SNRPG∼RING finger domain protein complex as a lead compound in the discovery of anti-cancer drugs: A molecular dynamics simulation approach

      2022, Informatics in Medicine Unlocked
      Citation Excerpt :

      Hence, the binding of the 4FI ligand is expected to reshape the sorting signal binding groove in the SNRPG ∼ RING finger domain complex in a way that optimal van der Waals contacts between the complex and the ligand are possible and non-polar solvent accessible surface area is diminished upon binding. Molecular recognition in drug-target interactions is known to be attributable to the contributions from both electrostatic and van der Waals interactions, solvation/desolvation and entropy effects [36,61,62]. The MM/GBSA calculations have the advantage of prediction accuracy at a much lower computational costs in comparison to the much more time-consuming methods widely used in the field of small-molecule drug design, which include post-processing of structure-based virtual screening [63,64].

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text