Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Synthon-based ligand discovery in virtual libraries of over 11 billion compounds

Abstract

Structure-based virtual ligand screening is emerging as a key paradigm for early drug discovery owing to the availability of high-resolution target structures1,2,3,4 and ultra-large libraries of virtual compounds5,6. However, to keep pace with the rapid growth of virtual libraries, such as readily available for synthesis (REAL) combinatorial libraries7, new approaches to compound screening are needed8,9. Here we introduce a modular synthon-based approach—V-SYNTHES—to perform hierarchical structure-based screening of a REAL Space library of more than 11 billion compounds. V-SYNTHES first identifies the best scaffold–synthon combinations as seeds suitable for further growth, and then iteratively elaborates these seeds to select complete molecules with the best docking scores. This hierarchical combinatorial approach enables the rapid detection of the best-scoring compounds in the gigascale chemical space while performing docking of only a small fraction (<0.1%) of the library compounds. Chemical synthesis and experimental testing of novel cannabinoid antagonists predicted by V-SYNTHES demonstrated a 33% hit rate, including 14 submicromolar ligands, substantially improving over a standard virtual screening of the Enamine REAL diversity subset, which required approximately 100 times more computational resources. Synthesis of selected analogues of the best hits further improved potencies and affinities (best inhibitory constant (Ki) = 0.9 nM) and CB2/CB1 selectivity (50–200-fold). V-SYNTHES was also tested on a kinase target, ROCK1, further supporting its use for lead discovery. The approach is easily scalable for the rapid growth of combinatorial libraries and potentially adaptable to any docking algorithm.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: V-SYNTHES approach to modular screening of Enamine REAL Space.
Fig. 2: Assessment of VLS computational performance for V-SYNTHES and standard VLS.
Fig. 3: The top five CB2 hits identified by V-SYNTHES.
Fig. 4: Selection and characterization of the best analogue series for CB2 hits from V-SYNTHES screening.

Similar content being viewed by others

Data availability

Chemical structures, synthetic methods, detailed results of biochemical characterization are presented in this paper and its Supplementary Information.

Code availability

V-SYNTHES scripts and example files have been deposited at GitHub (https://github.com/katritchlab/V-SYNTHES).

References

  1. Shoichet, B. K. & Kobilka, B. K. Structure-based drug screening for G-protein-coupled receptors. Trends Pharmacol. Sci. 33, 268–272 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Katritch, V., Cherezov, V. & Stevens, R. C. Structure-function of the G protein-coupled receptor superfamily. Annu. Rev. Pharmacol. Toxicol. 53, 531–556 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Renaud, J.-P. et al. Cryo-EM in drug discovery: achievements, limitations and prospects. Nat. Rev. Drug Discov. 17, 471–492 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Congreve, M., de Graaf, C., Swain, N. A. & Tate, C. G. Impact of GPCR structures on drug discovery. Cell 181, 81–91 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Stein, R. M. et al. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 579, 609–614 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Lyu, J. et al. Ultra-large library docking for discovering new chemotypes. Nature 566, 224–229 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Grygorenko, O. O. et al. Generating multibillion chemical space of readily accessible screening compounds. iScience 23, 101681 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gorgulla, C. et al. An open-source drug discovery platform enables ultra-large virtual screens. Nature 580, 663–668 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Graff, D. E., Shakhnovich, E. I. & Coley, C. W. Accelerating high-throughput virtual screening through molecular pool-based active learning. Chem. Sci. 12, 7866–7881 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Engels, M. F. & Venkatarangan, P. Smart screening: approaches to efficient HTS. Curr. Opin. Drug Discov. Dev. 4, 275–283 (2001).

    CAS  Google Scholar 

  11. Villoutreix, B. O., Eudes, R. & Miteva, M. A. Structure-based virtual ligand screening: recent success stories. Comb. Chem. High Throughput Screen. 12, 1000–1016 (2009).

    Article  CAS  PubMed  Google Scholar 

  12. Abagyan, R. & Totrov, M. High-throughput docking for lead generation. Curr. Opin. Chem. Biol. 5, 375–382 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Irwin, J. J. & Shoichet, B. K. Docking screens for novel ligands conferring new biology. J. Med. Chem. 59, 4103–4120 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Ertl, P. Cheminformatics analysis of organic substituents: identification of the most common substituents, calculation of substituent properties, and automatic identification of drug-like bioisosteric groups. J. Chem. Inf. Comput. Sci. 43, 374–380 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Bohacek, R. S., McMartin, C. & Guida, W. C. The art and practice of structure-based drug design: a molecular modeling perspective. Med. Res. Rev. 16, 3–50 (1996).

    Article  CAS  PubMed  Google Scholar 

  16. REAL Space (Enamine, 2020); https://enamine.net/library-synthesis/real-compounds/real-space-navigator

  17. Guzmán, M. Cannabinoids: potential anticancer agents. Nat. Rev. Cancer 3, 745–755 (2003).

    Article  PubMed  Google Scholar 

  18. Contino, M., Capparelli, E., Colabufo, N. A. & Bush, A. I. Editorial: the CB2 cannabinoid system: a new strategy in neurodegenerative disorder and neuroinflammation. Front. Neurosci. 11, 196 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Lunn, C. A. et al. Biology and therapeutic potential of cannabinoid CB2 receptor inverse agonists. Br. J. Pharmacol. 153, 226–239 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Corey, E. J. General methods for the construction of complex molecules. Pure Appl. Chem. 14, 19–38 (1967).

    Article  CAS  Google Scholar 

  21. Baell, J. B. & Holloway, G. A. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J. Med. Chem. 53, 2719–2740 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Li, X. et al. Crystal structure of the human cannabinoid receptor CB2. Cell 176, 459–467 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kroeze, W. K. et al. PRESTO-Tango as an open-source resource for interrogation of the druggable human GPCRome. Nat. Struct. Mol. Biol. 22, 362–369 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gaulton, A. et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 40, D1100–D1107 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Xing, C. et al. Cryo-EM structure of the human cannabinoid receptor CB2-Gi signaling complex. Cell 180, 645–654 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Wei, L., Surma, M., Shi, S., Lambert-Cheatham, N. & Shi, J. Novel insights into the roles of rho kinase in cancer. Arch. Immunol. Ther. Exp. 64, 259–278 (2016).

    Article  CAS  Google Scholar 

  27. Chin, V. T. et al. Rho-associated kinase signalling and the cancer microenvironment: novel biological implications and therapeutic opportunities. Expert Rev. Mol. Med. 17, e17 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Baker, M. Fragment-based lead discovery grows up. Nat. Rev. Drug Discov. 12, 5–7 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Schulz, M. N. & Hubbard, R. E. Recent progress in fragment-based lead discovery. Curr. Opin. Pharmacol. 9, 615–621 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Davis, B. J. & Hubbard, R. E. in Structural Biology in Drug Discovery 79–98 (2020).

  31. Zheng, Z. et al. Structure-based discovery of new antagonist and biased agonist chemotypes for the kappa opioid receptor. J. Med. Chem. 60, 3070–3081 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. de Graaf, C. et al. Crystal structure-based virtual screening for fragment-like ligands of the human histamine H1 receptor. J. Med. Chem. 54, 8195–8206 (2011).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  33. Katritch, V. et al. Structure-based discovery of novel chemotypes for adenosine A2A receptor antagonists. J. Med. Chem. 53, 1799–1809 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chen, Y. & Shoichet, B. K. Molecular docking and ligand specificity in fragment-based inhibitor discovery. Nat. Chem. Biol. 5, 358–364 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Abagyan, R. A., Orry, A., Raush, E., Budagyan, L. & Totrov, M. ICM User’s Guide and Reference Manual v.3.9 (MolSoft, 2021).

  36. Bogolubsky, A. V. et al. A one-pot parallel reductive amination of aldehydes with heteroaromatic amines. ACS Comb. Sci. 16, 375–380 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Savych, O. et al. One-pot parallel synthesis of 5-(dialkylamino)tetrazoles. ACS Comb. Sci. 21, 635–642 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Katritch, V., Rueda, M. & Abagyan, R. Ligand-guided receptor optimization. Methods Mol. Biol. 857, 189–205 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Gatica, E. A. & Cavasotto, C. N. Ligand and decoy sets for docking to G protein-coupled receptors. J. Chem. Inf. Model. 52, 1–6 (2012).

    Article  CAS  PubMed  Google Scholar 

  40. Bottegoni, G., Kufareva, I., Totrov, M. & Abagyan, R. Four-dimensional docking: a fast and accurate account of discrete receptor flexibility in ligand docking. J. Med. Chem. 52, 397–406 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Real Compound Libraries (Enamine, 2020); https://enamine.net/library-synthesis/real-compounds/real-compound-libraries

  42. Nikas, S. P. et al. Probing the carboxyester side chain in controlled deactivation (−)-Δ8-tetrahydrocannabinols. J. Med. Chem. 58, 665–681 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Nikas, S. P. et al. Novel 1′,1′-chain substituted hexahydrocannabinols: 9β-hydroxy-3-(1-hexyl-cyclobut-1-yl)-hexahydrocannabinol (AM2389) a highly potent cannabinoid receptor 1 (CB1) agonist. J. Med. Chem. 53, 6996–7010 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jacobs, M. et al. The structure of dimeric ROCK I reveals the mechanism for ligand selectivity. J. Biol. Chem. 281, 260–268 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Anastassiadis, T., Deacon, S. W., Devarajan, K., Ma, H. & Peterson, J. R. Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat. Biotechnol. 29, 1039–1045 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the staff at the USC Center for Advanced Research Computing, and the Google Cloud Platform for Higher Education and Research for providing computational resources. The study was funded by National Institute on Drug Abuse grants R01DA041435 and R01DA045020 (to V.K. and A.M.), National Institute of Mental Health Grant R01MH112205 and Psychoactive Drug Screening Program (to B.L.R.) and the Michael Hooker Distinguished Professorship (to B.L.R.). B.H. was supported by NIGMS T32-GM118289.

Author information

Authors and Affiliations

Authors

Contributions

A.A.S. and A.V.S. developed V-SYNTHES algorithms, performed calculations and wrote the first draft of the manuscript. B.H. and N.A.P. performed calculations and compound selection for ROCK1. Y.L., M.K.J., J.P. and X.-P.H. performed functional and selectivity assays. C.I.-T., N.K.T., F.T., N.Z. and S.P.N. performed binding assays. N.P. performed full VLS on Google Cloud. O.S., D.S.R. and Y.S.M. developed the REAL Space library and performed compound synthesis. B.L.R. supervised the functional and selectivity assays. A.M. supervised binding assays for CB1 and CB2. V.K. conceived the study and supervised all of its computational aspects. All of the authors contributed to writing and editing the manuscript.

Corresponding authors

Correspondence to Bryan L. Roth, Alexandros Makriyannis or Vsevolod Katritch.

Ethics declarations

Competing interests

A.A.S. and V.K. filed a provisional patent on V-SYNTHES method (application no. 63159888, University of Southern California).

Additional information

Peer review information Nature thanks Charlotte Dean and Amy Newman for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Evaluation of SYNTHES performance on CB2 receptor with only docking score (without considering docking pose of MEL candidates in the binding pocket).

(a) The number of hits at each score threshold from V-SYNTHES and standard VLS (b) Enrichment in V-SYNTHES vs. Standard VLS at different score thresholds, with the red x-mark showing threshold that yields 100 V-SYNTHES hits in the two-component library.

Extended Data Fig. 2 Binding pocket of CB2 with selected dead-end atoms.

a) 3D illustration of a MEL compound binding pose (carbon atoms colored cyan) with a “non-productive” pose. (b-d) 2D schematics showing other possible non-productive cases, including dead-end subpockets. Dead-end water-colored red, pseudoatoms colored magenta.

Extended Data Fig. 3 Details of practical application V-SYNTHES algorithms to CB receptors screening.

a, b, Two-component (a) and three-component (b) reaction cases.

Extended Data Fig. 4 Concentration-response curves for V-SYNTHES hits in functional assays at CB1 and CB2 receptors (except those shown in main text Figure 3).

β-arrestin recruitment Tango assays were performed to assess antagonist activity of the compounds in (a,b) CB1 and (c,d) CB2 receptors. The compounds rimonabant or SR144528 served as positive controls. The assays were carried out in the presence of 100 nM (EC80) of the dual CB1/CB2 CP55,940 agonist. The data points are presented as mean ± SEM with n = 3 independent experiments, each one carried out in triplicate.

Extended Data Fig. 5 Competition binding curves for the best CB2 hit compounds from V-SYNTHES.

Radioligand binding assays were used to assess the binding affinities in rCB1 (a) and hCB2 (b). [3H]CP-55,940 was used as the radioligand. The data were presented as mean ± SEM with n = 3 independent experiments, each one carried out in triplicate.

Extended Data Fig. 6 Assessment of off-target selectivity for the best V-SYNTHES CB2 hits.

(a-c) Screening of compounds 673, 610 and 523 at 10 µM concentrations in GPCRome-Tango assays for >300 receptors. Dopamine D2 (DRD2) and 100 nM Quinpirole served as an assay control. The data are presented as mean ± SEM (n = 4) and the values of fold of basal > 3 are marked as significant hits. (d-o) Follow-up dose-response curves for targets with >3 fold increased activity. Known agonists or antagonist that showed activity served as positive controls. The data points are presented as mean ± SEM with n = 3 independent experiments, each assay carried out in triplicate.

Extended Data Fig. 7 Identification and characterization of CB1 and CB2 hits from standard VLS of 115M Enamine REAL compounds.

(a) Chemical structures of the hits from the standard VLS. (b-c). Concentration-response curves of the best hits in β-arrestin recruitment Tango assays for antagonist activity at CB1 (b) and CB2 (c) receptors. The compounds rimonabant or SR144528 served as positive controls. The assays were carried out in the presence of 100 nM (EC80) of the dual CB1/CB2 CP55,940 agonist. The data points are presented as mean ± SEM with n = 3 independent experiments, each one carried out in triplicate. (d) Functional potencies and binding affinities of the hit compounds from standard VLS. The 95% Confidence Intervals (CI) were calculated from n = 3 independent assays, with 16 dose-response points for functional Ki values and 8 dose-response points for affinity Ki values, except for values marked with *, roughly estimated from three-point assays.

Extended Data Fig. 8 Competition binding curves for the best CB2 hit compounds from standard VLS.

Radioligand binding assays were used to assess the binding affinities in hCB2. [3H]CP-55,940 was used as the radioligand. The data were presented as mean ± SEM with n = 3 independent experiments, each one carried out in triplicate.

Extended Data Fig. 9 Chemical structures for series of the SAR-by-catalog analogues of antagonists, discovered by V-SYNTHES.

Shown are 60 analogues of 523 (a), 610 (b), and 673 (c) with inhibitory activity >40% in the single point functional assays. All 104 analogues tested are shown in Supplementary Information Table S3.

Extended Data Fig. 10 Functional potency and binding affinity assessment of the SAR-by-catalog analogues of the antagonist 523, discovered by V-SYNTHES.

Table compounds with CB2 potency better than 500 nM are shown, antagonists with affinities better than 10 nM highlighted in bold, >50-fold selective by italic. Functional Ki values and 95% Confidence Intervals were calculated from n = 4 independent assays with 16 dose-response points. Affinity Ki values and 95% Confidence Intervals were calculated from n = 3 independent assays with 8 dose-response points.

Extended Data Fig. 11 Concentration-response curves for series of the SAR-by-catalog analogues of 523, 610 and 673 antagonists, discovered by V-SYNTHES.

The β-arrestin recruitment Tango assays were performed to assess the antagonist activity of the best hits at CB1 (a-i), and CB2 (j-o) receptors. Note that the six best analogues of 523 shown in Fig. 4 are excluded here. The compounds rimonabant and SR144528 served as positive controls. The assays were carried out in the presence of 100 nM (EC80) of the CP55,940 agonist. The data were presented as mean ± SEM with n = 3 independent experiments, each run carried out in triplicate.

Extended Data Fig. 12 Assessment of off-target selectivity for the best SAR-by-catalog compounds 733 and 747.

(a-b) Screening of compounds 733 and 747 in GPCRome-Tango assay for >300 receptors at 10 µM concentrations. Dopamine D2 (DRD2) and 100 nM Quinpirole served as an assay control. The data are presented as mean ± SEM (n = 4) and the values of fold of basal > 3 marked as significant hits. (c-d) Follow-up dose-response curves for targets with >3 fold increased activity. Known agonists that showed activity served as positive controls. The data were presented as mean ± SEM with n = 3 independent experiments, each run carried out in triplicate.

Extended Data Fig. 13 Application of V-SYNTHES to the discovery of ROCK1 inhibitors.

(a,b) Computational assessment of V-SYNTHES performance vs standard VLS. (a) The number of candidate hits at each score threshold from V-SYNTHES and standard VLS. (b) Enrichment in V-SYNTHES vs. standard VLS at different score thresholds, with the red x-mark showing threshold that yields 100 hits in the two-component library. (c) Chemical structures of all selected by V-SYNTHES and synthesized compounds for ROCK1 kinase.

Extended Data Fig. 14 Experimental characterization of candidate ROCK1 inhibitors predicted by V-SYNTHES.

Full dose-response curves for the ROCK1 hits in (a) functional potency and (b) binding affinity at human ROCK1. The data points are presented as mean ± SEM from n = 3 independent experiments, each run carried out in triplicate. (c) Values of binding affinities and functional potencies for all candidate compounds predicted by V-SYNTHES. Bold font highlight hits with IC50<10 µM. Estimated values for curves that did not allow accurate fitting are marked with *.

Extended Data Fig. 15 Examples of typical Enamine REAL reactions.

(a) two-component reaction (b) three-component reaction.

Extended Data Table 1 Potencies and affinities of V-SYNTHES hits in functional and binding assays at CB1 and CB2 receptors

Supplementary information

Supplementary Information

Supplementary Figs. 1–4 and Supplementary Tables 1–4.

Reporting Summary

Supplementary File 2

Detailed synthesis protocol for all compounds in the paper.

Supplementary File 3

NMR and LC–MS spectra for all compounds in the paper.

Supplementary File 4

HRMS spectra for all compounds in the paper.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sadybekov, A.A., Sadybekov, A.V., Liu, Y. et al. Synthon-based ligand discovery in virtual libraries of over 11 billion compounds. Nature 601, 452–459 (2022). https://doi.org/10.1038/s41586-021-04220-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04220-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing