Skip to main content
Log in

Theoretical studies on the selectivity mechanisms of PI3Kδ inhibition with marketed idelalisib and its derivatives by 3D-QSAR, molecular docking, and molecular dynamics simulation

  • Original Paper
  • Published:
Journal of Molecular Modeling Aims and scope Submit manuscript

Abstract

Phosphoinositide 3-kinases (PI3Ks) are crucial for cell proliferation, metabolism, motility, and cancer progression. Since the selective PI3Kδ inhibitor, idelalisib, was firstly approved by the FDA in 2014, large numbers of selective PI3Kδ inhibitors have been reported, but the detailed mechanisms of selective inhibition to PI3Kδ for idelalisib or its derivatives have not been well addressed. In this study, 3D-QSAR with COMFA, molecular docking, and molecular dynamic (MD) simulations was used to explore the binding modes between PI3Kδ and idelalisib derivatives. Firstly, a reliable COMFA model (q2 = 0.59, ONC = 8, r2 = 0.966) was built and the contour maps showed that the electrostatic field had more significant contribution to the bioactivities of inhibitors. Secondly, two molecular docking methods including rigid receptor docking (RRD) and induced fit docking (IFD) were employed to predict the docking poses of all the studied inhibitors and revealed the selective binding mechanisms. And then, the results of the MD simulation and the binding free energy decomposition verified that the binding of PI3Kδ/inhibitors was mainly contributed from hydrogen bonding and hydrophobic interactions and some key residues for selective binding were highlighted. Finally, based on the models developed, 14 novel inhibitors were optimized and some showed satisfactory predicted bioactivity. Taken together, the results provided by this study may facilitate the rational design of novel and selective PI3Kδ inhibitors.

.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Fruman DA, Rommel C (2014) PI3K and cancer: lessons, challenges and opportunities. Nat. Rev. Drug Discov. 13(2):140–156

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Cantley LC (2002) The phosphoinositide 3-kinase pathway. Science 296(5573):1655–1657

    CAS  PubMed  Google Scholar 

  3. Carracedo A, Pandolfi PP (2008) The PTEN-PI3K pathway: of feedbacks and cross-talks. Oncogene 27(41):5527–5541

    CAS  PubMed  Google Scholar 

  4. Liu P, Cheng H, Roberts TM, Zhao JJ (2009) Targeting the phosphoinositide 3-kinase pathway in cancer. Nat. Rev. Drug Discov. 8(8):627–644

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Courtney KD, Corcoran RB, Engelman JA (2010) The PI3K pathway as drug target in human cancer. J. Clin. Oncol. 28(6):1075–1083

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Jiang BH, Liu LZ (2009) PI3K/PTEN signaling in angiogenesis and tumorigenesis. Adv. Cancer Res. 102:19–65

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Hennessy BT, Smith D, Ram P, Lu Y (2005) Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat. Rev. Drug Discov. 4(12):988–1004

    CAS  PubMed  Google Scholar 

  8. Zhu J, Wang M, Cao B, Hou T, Mao X (2014) Targeting the phosphatidylinositol 3-kinase/AKT pathway for the treatment of multiple myeloma. Curr. Med. Chem. 21(27):3173–3187

    CAS  PubMed  Google Scholar 

  9. Setti A, Kumar MJ, Babu KR, Rasagna A (2015) Potency and pharmacokinetics of broad spectrum and isoform-specific p110γ and δ inhibitors in cancers. J. Recept. Signal Transduct. Res. 36(1):26–36

    PubMed  Google Scholar 

  10. Zhu J, Pan P, Li Y, Wang M, Li D, Cao B, Mao X, Hou T (2014) Theoretical studies on beta and delta isoform-specific binding mechanisms of phosphoinositide 3-kinase inhibitors. Mol. BioSyst. 10(3):454–466

    CAS  PubMed  Google Scholar 

  11. Yuan TL, Cantley LC (2008) PI3K pathway alterations in cancer: variations on a theme. Oncogene 27(41):5497–5510

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Jabbour E, Ottmann OG, Deininger M, Hochhaus A (2014) Targeting the phosphoinositide 3-kinase pathway in hematologic malignancies. Haematologica 99(1):7–18

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhu J, Hou T, Mao X (2015) Discovery of selective phosphatidylinositol 3-kinase inhibitors to treat hematological malignancies. Drug Discov. Today 20(8):988–994

    CAS  PubMed  Google Scholar 

  14. Li T, Wang G (2014) Computer-aided targeting of the PI3K/Akt/mTOR pathway: toxicity reduction and therapeutic opportunities. Int. J. Mol. Sci. 15(10):18856–18891

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Li M, Sala V (2018) Phosphoinositide 3-kinase gamma inhibition protects from anthracycline cardiotoxicity and reduces tumor growth. Circulation 138(7):696–711

    CAS  PubMed  Google Scholar 

  16. Vyas P, Vohora D (2016) Phosphoinositide-3-kinases as the novel therapeutic targets for the inflammatory diseases: current and future perspectives. Curr. Drug Targets 18(14):1622–1640

    Google Scholar 

  17. Vangapandu HV, Jain N, Gandhi V (2017) Duvelisib: a phosphoinositide-3 kinase δ/γ inhibitor for chronic lymphocytic leukemia. Expert Opin. Investig. Drugs 26(5):625–632

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Lannutti BJ, Meadows SA, Herman SE, Kashishian A (2011) CAL-101, a p110δ selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood 117(2):591–594

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Sabbah DA, Vennerstrom JL, Zhong HA (2012) Binding selectivity studies of phosphoinositide 3-kinases using free energy calculations. J. Chem. Inf. Model. 52(12):3213–3224

    CAS  PubMed  Google Scholar 

  20. Kitchen DB, Decornez H, Furr JR, Bajorath J (2004) Docking and scoring in virtual screening for drug discovery: methods and applications. Nat. Rev. Drug Discov. 3(11):935–949

    CAS  PubMed  Google Scholar 

  21. Sabbah DA, Vennerstrom JL, Zhong HZ (2010) Docking studies on isoform-specific inhibition of phosphoinositide-3-kinases. J. Chem. Inf. Model. 50(10):1887–1898

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Shao S, Yu R, Yu Y, Li Y (2014) Dual-inhibitors of STAT5 and STAT3: studies from molecular docking and molecular dynamics simulations. J. Mol. Model. 20(8):2399

    PubMed  Google Scholar 

  23. Zhang C, Du C, Feng Z, Zhu J, Li Y (2015) Hologram quantitative structure activity relationship, docking, and molecular dynamics studies of inhibitors for CXCR4. Chem. Biol. Drug Des. 85(2):119–136

    CAS  PubMed  Google Scholar 

  24. Iqbal S, Krishnan DA, Gunasekaran K (2018) Identification of potential PKC inhibitors through pharmacophore designing, 3D–QSAR and molecular dynamics simulations targeting Alzheimer’s disease. J Biomol Struct Dyn 36(15):4029–4044

  25. Katari SK, Natarajan P, Swargam S, Kanipakam H, Pasala C (2016) Inhibitor design against JNK1 through e-pharmacophore modeling docking and molecular dynamics simulations. J. Recept. Signal Transduct. Res. 36(6):558–571

    CAS  PubMed  Google Scholar 

  26. Rajamanikandan S, Jeyakanthan J, Srinivasan P (2017) Molecular docking, molecular dynamics simulations, computational screening to design quorum sensing inhibitors targeting LuxP of Vibrio harveyi and its biological evaluation. Appl. Biochem. Biotechnol. 181(1):192–218

    CAS  PubMed  Google Scholar 

  27. Zondagh J, Balakrishnan V, Achilonu I, Dirr HW, Sayed Y (2018) Molecular dynamics and ligand docking of a hinge region variant of South African HIV-1 subtype C protease. J Mol Graph Model 82:1–11

    CAS  PubMed  Google Scholar 

  28. Xu C, Ren Y (2015) Molecular modeling studies of [6,6,5] Tricyclic Fused Oxazolidinones as FXa inhibitors using 3D-QSAR, Topomer CoMFA, molecular docking and molecular dynamics simulations. Bioorg. Med. Chem. Lett. 25(20):4522–4528

    CAS  PubMed  Google Scholar 

  29. Tang HJ, Yang L, Li JH, Chen J (2016) Molecular modelling studies of 3,5-dipyridyl-1,2,4-triazole derivatives as xanthine oxidoreductase inhibitors using 3D-QSAR, Topomer CoMFA, molecular docking and molecular dynamic simulations. J Taiwan Inst Chem E 68:64–73

    CAS  Google Scholar 

  30. Aksoydan B, Kantarcioglu I, Erol I, Salmas RE, Durdagi S (2018) Structure-based design of hERG-neutral antihypertensive oxazalone and imidazolone derivatives. J Mol Graph Model 79:103–117

    CAS  PubMed  Google Scholar 

  31. Zhao S, Zhu J, Xu L, Jin J (2017) Theoretical studies on the selective mechanisms of GSK3beta and CDK2 by molecular dynamics simulations and free energy calculations. Chem. Biol. Drug Des. 89(6):846–855

    CAS  PubMed  Google Scholar 

  32. Shen M, Zhou S, Li Y, Li D, Hou T (2013) Theoretical study on the interaction of pyrrolopyrimidine derivatives as LIMK2 inhibitors: insight into structure-based inhibitor design. Mol. BioSyst. 9(10):2435–2446

    CAS  PubMed  Google Scholar 

  33. Xu L, Li Y, Li L, Zhou S, Hou T (2012) Understanding microscopic binding of macrophage migration inhibitory factor with phenolic hydrazones by molecular docking, molecular dynamics simulations and free energy calculations. Mol. BioSyst. 8(9):2260–2273

    CAS  PubMed  Google Scholar 

  34. Ekhteiari Salmas R, Unlu A, Bektas M, Yurtsever M, Mestanoglu M, Durdagi S (2017) Virtual screening of small molecules databases for discovery of novel PARP-1 inhibitors: combination of in silico and in vitro studies. J. Biomol. Struct. Dyn. 35(9):1899–1915

    CAS  PubMed  Google Scholar 

  35. Patel L, Chandrasekhar J, Evarts J (2016) 2,4,6-Triaminopyrimidine as a novel hinge binder in a series of PI3Kδ selective inhibitors. J. Med. Chem. 59(7):3532–3548

    CAS  PubMed  Google Scholar 

  36. Sherman W, Beard H, Farid R (2010) Use of an induced fit receptor structure in virtual screening. Chem. Biol. Drug Des. 67(1):83–84

    Google Scholar 

  37. Somoza JR, David K (2015) Structural, biochemical, and biophysical characterization of idelalisib binding to phosphoinositide 3-kinase δ. J Bio Chem 290(13):8439–8446

    CAS  Google Scholar 

  38. Case DA, Cheatham TE, Darden T, Gohlke H (2005) The Amber biomolecular simulation programs. J. Comput. Chem. 26(16):1668–1688

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Duan Y, Wu C, Chowdhury S, Lee MC, Xiong GM (2003) A point-charge force field for molecular mechanics simulations of proteins based on condensed-phase quantum mechanical calculations. J. Comput. Chem. 24(16):1999–2012

    CAS  PubMed  Google Scholar 

  40. Wang JM, Wolf RM, Caldwell JW, Kollman PA, Case DA (2004) Development and testing of a general amber force field. J. Comput. Chem. 25(9):1157–1174

    CAS  PubMed  Google Scholar 

  41. Stewart JPJ (2010) Optimization of parameters for semiempirical methods I. Method. J Comput Chem 10(2):221–264

    Google Scholar 

  42. Bayly CI, Cieplak P, Cornell WD, Kollman PA (1993) A well-behaved electrostatic potential based method using charge restraints for deriving atomic charges: the resp model. J. Phys. Chem. 97(40):10269–10280

    CAS  Google Scholar 

  43. Darden T, York D, Pedersen L (1993) Particle mesh Ewald: an N·log(N) method for Ewald sums in large systems. J. Phys. Chem. 98(12):10089–10092

    CAS  Google Scholar 

  44. Vincent K, Van Gunsteren W, Hünenberger P (2001) A fast SHAKE algorithm to solve distance constraint equations for small molecules in molecular dynamics simulations. J. Comput. Chem. 22(5):501–508

    Google Scholar 

  45. Kollman PA, Massova I, Reyes C, Kuhn B, Huo S, Chong L, Lee M (2000) Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models. Cheminform 32(10):889–897

    Google Scholar 

  46. Hou T, Li Y, Wang W (2011) Prediction of peptides binding to the PKA RIIα subunit using a hierarchical strategy. Bioinformatics 27(13):1814–1821

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Sun H, Li Y, Shen M, Tian S, Xu L, Pan P, Guan Y, Hou T (2014) Assessing the performance of MM/PBSA and MM/GBSA methods. 5. Improved docking performance using high solute dielectric constant MM/GBSA and MM/PBSA rescoring. Phys. Chem. Chem. Phys. 16(40):22035–22045

    CAS  PubMed  Google Scholar 

  48. Chen F, Liu H, Sun H, Pan P, Li Y, Li D, Hou T (2016) Assessing the performance of the MM/PBSA and MM/GBSA methods. 6. Capability to predict protein–protein binding free energies and re-rank binding poses generated by protein–protein docking. Phys. Chem. Chem. Phys. 18(18):22129–22139

    CAS  PubMed  Google Scholar 

  49. Pan P, Yu H, Liu Q, Kong X, Chen H, Chen J, Liu Q, Li D, Kang Y, Sun H (2017) Combating drug-resistant mutants of anaplastic lymphoma kinase with potent and selective type-I1/2 inhibitors by stabilizing unique DFG-shifted loop conformation. Acs Cent Sci 3(11):1208–1220

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Hou T, Li Y, Wang W (2011) Prediction of peptides binding to the PKA RIIalpha subunit using a hierarchical strategy. Bioinformatics 27(13):1814–1821

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Sun H, Li Y, Li D, Hou T (2013) Insight into crizotinib resistance mechanisms caused by three mutations in ALK tyrosine kinase using free energy calculation approaches. J. Chem. Inf. Model. 53(9):2376–2389

    CAS  PubMed  Google Scholar 

  52. Sun H, Li Y, Tian S, Xu L, Hou T (2014) Assessing the performance of MM/PBSA and MM/GBSA methods. 4. Accuracies of MM/PBSA and MM/GBSA methodologies evaluated by various simulation protocols using PDBbind data set. Phys. Chem. Chem. Phys. 16(31):16719–16729

    CAS  PubMed  Google Scholar 

  53. Sun H, Duan L, Chen F, Liu H, Wang Z, Pan P, Zhu F (2018) Assessing the performance of MM/PBSA and MM/GBSA methods. 7. Entropy effects on the performance of end-point binding free energy calculation approaches. Phys. Chem. Chem. Phys. 20(21):14450–14460

    CAS  PubMed  Google Scholar 

  54. Chen F, Liu H, Sun H, Pan P, Li Y, Li D, Hou T (2016) Assessing the performance of the MM/PBSA and MM/GBSA methods. 6. Capability to predict protein-protein binding free energies and re-rank binding poses generated by protein-protein docking. Phys. Chem. Chem. Phys. 18(32):22129–22139

    CAS  PubMed  Google Scholar 

  55. Chen F, Sun H, Wang J, Zhu F, Liu H (2018) Assessing the performance of MM/PBSA and MM/GBSA methods. 8. Predicting binding free energies and poses of protein-RNA complexes. RNA 24(9):1183–1194

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Onufriev A, Donald Bashford A, Case DA (2000) Modification of the generalized born model suitable for macromolecules. J. Phys. Chem. B 104(15):3712–3720

    CAS  Google Scholar 

  57. Weiser J, Shenkin P, Still W (1999) Approximate atomic surfaces from linear combinations of pairwise overlaps (LCPO). J. Comput. Chem. 20(2):217–230

    CAS  Google Scholar 

  58. Berndt A, Miller S, Williams O, Le D (2010) The p110δ crystal structure uncovers mechanisms for selectivity and potency of novel PI3K inhibitors. Nat. Chem. Biol. 6(2):117–124

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Williams R, Berndt A, Miller S, Hon W, Zhang X (2009) Form and flexibility in phosphoinositide 3-kinases. Biochem. Soc. Trans. 37(4):615–626

    CAS  PubMed  Google Scholar 

  60. Safina BS, Sweeney ZK, Li J, Chan BK (2013) Identification of GNE-293, a potent and selective PI3Kδ inhibitor: navigating in vitro genotoxicity while improving potency and selectivity. Bioorg. Med. Chem. Lett. 23(17):4953–4959

    CAS  PubMed  Google Scholar 

Download references

Funding

The study was financially supported by the National Natural Science Foundation of China (No. 21807049, 81803430) and the Fundamental Research Funds for the Central Universities (JUSRP11892).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Jingyu Zhu or Jian Jin.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s note

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

Electronic supplementary material

ESM 1

(DOCX 2327 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, J., Ke, K., Xu, L. et al. Theoretical studies on the selectivity mechanisms of PI3Kδ inhibition with marketed idelalisib and its derivatives by 3D-QSAR, molecular docking, and molecular dynamics simulation. J Mol Model 25, 242 (2019). https://doi.org/10.1007/s00894-019-4129-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00894-019-4129-x

Keywords

Navigation