Skip to main content
Log in

Identification of Novel PI3Kα Inhibitor Against Gastric Cancer: QSAR-, Molecular Docking–, and Molecular Dynamics Simulation–Based Analysis

  • Original Article
  • Published:
Applied Biochemistry and Biotechnology Aims and scope Submit manuscript

Abstract

Gastric cancer (GC) is a malignant tumor with global incidence and death ranking fifth and fourth, respectively. GC patients nevertheless have a poor prognosis despite the effectiveness of more advanced chemotherapy and surgical treatment options. The second most frequently mutated gene in GC is PI3Kalpha, a confirmed oncogene that results in abnormal PI3K/AKT/mTOR signaling, causing enhanced translation, proliferation, and survival, and is mutated in 7–25% of GC patients. The protein PI3Kalpha was targeted in the present study by utilizing machine learning (ML), molecular docking, and simulation. A total of 9214 molecules from the DrugBank database were chosen for the first screening. A training set for 6770 compounds tested against PI3Kalpha was assessed to create a quantitative structure-activity relationship-based machine learning model using five different classification algorithms: random forest, random tree, J48 pruned tree, decision stump, and REPTree. Furthermore, consideration was given to the random forest classifier for screening based on its performance index (Kappa statistics, ROC, and MCC). Overall, 1539 of the 9214 drug bank compounds were predicted to be active. Thereafter, three pharmacological filters, Lipinski’s rule, Ghose filter, and Veber rule, were applied to test the drug-like properties of the screened compounds. Twenty-six of 1593 compounds showed excellent drug-like properties and were further considered for molecular docking. Thereafter, two compounds were screened as hits because they possessed the molecular docked position with the lowest binding energy and an excellent bonding profile. The binding stability of the selected compounds was further assessed through molecular dynamics simulations for up to 100 ns. Furthermore, compound 1-(3-(2,4-dimethylthiazol-5-YL)-4-oxo-2,4-dihydroindeno[1,2-C]pyrazol-5-YL)-3-(4-methylpiperazin-1-YL) urea was selected as a potential hit in the final screening by analyzing a number of parameters, including the Rg, RMSD, RMSF, H bonding, and SASA profile. Therefore, we conclude that compound 1-(3-(2, 4-dimethylthiazol-5-YL)-4-oxo-2,4-dihydroindeno[1,2-C]pyrazol-5-YL)-3-(4-methylpiperazin-1-YL) urea has efficient inhibitory potential against PI3Kalpha protein and could be utilized for the development of effective drugs against GC.

Graphical Abstract

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

Similar content being viewed by others

Data Availability

Initial X-ray structures are available at the Protein Data Bank (https://www.rcsb.org/). Datasets used for different model developments are available at ChEMBL (https://www.ebi.ac.uk/chembl/) and the DrugBank database (https://go.drugbank.com/). All other data are available in the main text.

References

  1. Iwu, C. D., & Iwu-Jaja, C. J. (2023). Gastric cancer epidemiology: Current trend and future direction. Hygiene, 3, 256–268.

    Article  Google Scholar 

  2. Ma, X., et al. (2023). Upregulation of PIK3IP1 monitors the anti-cancer activity of PI3Kα inhibitors in gastric cancer cells. Biochemical Parmacology, 207, 115380.

    Article  CAS  Google Scholar 

  3. Arcaro, A., & Guerreiro, A. S. (2007). The phosphoinositide 3-kinase pathway in human cancer: Genetic alterations and therapeutic implications. Curr Genomics, 8, 271–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Baghery, S., Khorasani, A., et al. (2021). The PI3K/Akt/mTOR signaling pathway in gastric cancer; from oncogenic variations to the possibilities for pharmacologic interventions. European Journal of Pharmacology, 898, 173983.

    Article  Google Scholar 

  5. Vincent, E. E., et al. (2011). Akt phosphorylation on Thr308 but not on Ser473 correlates with akt protein kinase activity in human non-small cell lung cancer. British Journal of Cancer, 104, 1755–1761.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Inoki, K., Li, Y., Zhu, T., Wu, J., & Guan, K. L. (2002). TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nature Cell Biology, 4, 648–657.

    Article  CAS  PubMed  Google Scholar 

  7. Wang, C., et al. (2015). 4EBP1/eIF4E and p70S6K/RPS6 axes play critical and distinct roles in hepatocarcinogenesis driven by AKT and N-Ras protooncogenes. Hepatology, 61, 200–213.

    Article  PubMed  Google Scholar 

  8. He, Y., et al. (2021). Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct Target Ther, 6, 425.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Riquelme, I., et al. (2016). The Gene expression status of the PI3K/AKT/mTOR pathway in gastric Cancer tissues and cell lines. Pathology Oncology Research : Por, 22, 797–805.

    Article  CAS  PubMed  Google Scholar 

  10. Nand, M., Maiti, P., Joshi, T., Chandra, et al. (2020). Virtual screening of anti-HIV1 compounds against SARS-CoV-2: Machine learning modeling, chemoinformatics and molecular dynamics simulation based analysis. Scientific Reports, 10, 20397.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nand, M., Maiti, P., Chandra, S., & Pande, V. (2018). Screening of Alk targeted Anti-lung Cancer inhibitors: An in Silico Exploration from Natural Domain. Int J Recent Sci Res, 9(1), 22925–22928.

    Google Scholar 

  12. Nand, M., Maiti, P., Pant, R., Kumari, M., Chandra, S., & Pande, V. (2017). Virtual screening of natural compounds as inhibitors of EGFR 696–1022 T790M associated with non-small cell lung cancer. Bioinformation, 12(6), 311–317.

    Article  Google Scholar 

  13. Nand, M., Maiti, P., Pande, V., & Chandra, S. (2016). Predictive model assisted in silico screening of anti-lung cancer activity of compounds from lichen source. Int J Recent Sci Res, 7(4), 10370–10373.

    Google Scholar 

  14. Maiti, P., Nand, M., Joshi, T., Ramakrishnan, M. A. (2020). Identification of luteolin – 7-glucoside and epicatechin gallate from Vernonia cinerea, as novel EGFR L858R kinase inhibitors against lung cancer: Docking and simulation-based study. Journal of Biomolecular Structure and Dynamics, 1–10.

  15. Sharma, P., Joshi, T., Joshi, T., Mathpal, S. (2021). In silico screening of natural compounds to inhibit interaction of human ACE2 receptor and spike protein of SARS-CoV-2 for the prevention of COVID-19. Journal of Biomolecular Structure and Dynamics.

  16. Maiti, P., Sharma, P., Nand, M., Bhatt, I. D., et al. (2022). Integrated machine learning and chemoinformatics-based screening of mycotic compounds against kinesin spindle protein Eg5 for lung cancer therapy. Molecules, 27, 1639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  18. Wishart, D. S., et al. (2018). DrugBank 5.0: A major update to the DrugBank database for 2018. Nucleic Acids Research, 46, D1074–D1082.

    Article  CAS  PubMed  Google Scholar 

  19. Yap, C. (2011). PaDEL-descriptor: An open source software to calculate molecular descriptors and fingerprints - yap – 2011. Journal of Computational Chemistry - Wiley Online Library, 32, 1466–1476.

    CAS  Google Scholar 

  20. Frank, E., Hall, M., Trigg, L., Holmes, G., & Witten, I. H. (2004). Data mining in bioinformatics using Weka. Bioinformatics, 20, 2479–2481.

    Article  CAS  PubMed  Google Scholar 

  21. Steinbeck, C., et al. (2003). The Chemistry Development Kit (CDK): An open-source Java library for chemo- and bioinformatics. Journal of Chemical Information and Computer Sciences, 43, 493–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Geete, A., Damre, M., & Kokkula, A. Drug Likeness Tool (DruLiTo) Chemistry Development Kit (CDK), Department of Pharmacoinformatics, NIPER, Mohali, 1, 1–7.

  23. Hou, Y., et al. (2022). Discovery of novel phosphoinositide-3-kinase α inhibitors with high selectivity, excellent bioavailability, and long-acting efficacy for gastric cancer. Journal of Medicinal Chemistry 65, 9873–9892.

    Article  CAS  PubMed  Google Scholar 

  24. Abraham, M. J., et al. (2015). GROMACS: High performance molecular simulations through multi-level parallelism from laptops to supercomputers. SoftwareX, 1–2, 19–25.

    Article  ADS  Google Scholar 

  25. Vanommeslaeghe, K., et al. (2010). CHARMM General Force Field (CGenFF): A force field for drug-like molecules compatible with the CHARMM all-atom additive biological force fields. Journal of Computational Chemistry, 31, 671–690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Maheshwari, S., et al. (2017). Kinetic and structural analyses reveal residues in phosphoinositide 3-kinase α that are critical for catalysis and substrate recognition. The Journal of Biological Chemistry, 292, 13541.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Gabelli, S., et al. (2010). Structural effects of oncogenic PI3Kα mutations. Current Topics in Microbiology and Immunology, 347, 43–53.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Maiti, P. (2023). Potent multi-target natural inhibitors against SARS-CoV-2 from medicinal plants of the Himalaya: A discovery from hybrid machine learning, chemoinformatics, and simulation assisted screening. Journal of Biomolecular Structure & Dynamics 1–14.

  29. Matsuoka, T., & Yashiro, M. (2014). The role of PI3K/Akt/mTOR signaling in gastric carcinoma. Cancers (Basel), 6, 1441–1463.

    Article  CAS  PubMed  Google Scholar 

  30. Berman, H. M., et al. (2000). The Protein Data Bank. Nucleic Acids Research, 28, 235–242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ew, Y. (2004). Synthesis and evaluation of indenopyrazoles as cyclin-dependent kinase inhibitors. Part 4: Heterocycles at C3. Bioorganic & medicinal chemistry letters 14.

  32. Joshi, T., et al. (2022). Computational investigation of drug bank compounds against 3 C-like protease (3CLpro) of SARS-CoV-2 using deep learning and molecular dynamics simulation. Mol Divers, 26, 2243–2256.

    Article  CAS  PubMed  Google Scholar 

  33. Maiti, P., Nand, M., Joshi, H., & Chandra, S. (2016). Molecular docking analysis and screening of plant compounds against lung cancer target EGFR T790M mutant. International Journal of Computational Bioinformatics and in Silico Modeling, 5(2), 787–792.

    Google Scholar 

  34. Mathpal, S., Joshi, T., Sharma, P., Maiti, P., & Nand, M. (2024). ChandraS., In silico screening of chalcone derivatives as promising EGFR-TK inhibitors for the clinical treatment of cancer, 3 Biotech, 14:18.

Download references

Acknowledgements

We extend our appreciation to our research team for their contribution to this study.

Funding

This study was financially supported by The Natural Science Foundation of Shandong Province under Grant Number ZR2020MH355, which funded the project “To explore the mechanism of Kunshen granules in the treatment of advanced gastric cancer based on MIR-92B via the PI3K/Akt pathway.”

Author information

Authors and Affiliations

Authors

Contributions

F.Y and T.L. conducted the machine learning and molecular docking experiments and planned outline of this research, Z.C. generated the idea and wrote the manuscript, and X.X., T.C., and critically reviewed and guided in the idea generation.

Corresponding author

Correspondence to Zhiqun Cao.

Ethics declarations

Ethical Approval

Not applicable.

Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher’s Note

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

Fang Yuan and Ting Li are Co-first Authors and equally contributed to this work.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuan, F., Li, T., Xu, X. et al. Identification of Novel PI3Kα Inhibitor Against Gastric Cancer: QSAR-, Molecular Docking–, and Molecular Dynamics Simulation–Based Analysis. Appl Biochem Biotechnol (2024). https://doi.org/10.1007/s12010-024-04898-3

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12010-024-04898-3

Keywords

Navigation