Skip to main content
Log in

Chlordiazepoxide against signalling, receptor and regulatory proteins of breast cancer: a structure-based in-silico approach

  • Original Paper
  • Published:
Medical Oncology Aims and scope Submit manuscript

Abstract

Among the most prevalent forms of cancer are breast, lung, colon-rectum, and prostate cancers, and breast cancer is a major global health challenge, contributing to 2.26 million cases with approximately 685,000 deaths worldwide in 2020 alone, typically beginning in the milk ducts or lobules that produce and transport milk during lactation and it is becoming challenging to treat as the tissues are developing resistance, which makes urgent calls for new multitargeted drugs. The multitargeted drug design provides a better solution, simultaneously targeting multiple pathways, even when the drug resists one, it remains effective for others. In this study, we included four crucial proteins that perform signalling, receptor, and regulatory action, namely- NUDIX Hydrolases, Dihydrofolate Reductase, HER2/neu Kinase and EGFR and performed multitargeted molecular docking studies against human-approved drugs using HTVS, SP and extra precise algorithms and filtered the poses with MM\GBSA, suggested a benzodiazepine derivative chlordiazepoxide, used as an anxiolytic agent, can be a multitargeted inhibitor with docking and MM\GBSA score ranging from − 4.628 to − 7.877 and − 18.59 to − 135.86 kcal/mol, respectively, and the most interacted residues were 6ARG, 6GLU, 3TRP, and 3VAL. The QikProp-based ADMET and DFT computations showed the suitability and stability of the drug candidate followed by 100 ns MD simulation in water and MMGBSA on trajectories, resulting in stable performance and many intermolecular interactions to make the complexes stable, which favours that chlordiazepoxide can be a multitargeted breast cancer inhibitor. However, experimental validation is needed before its use.

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
Fig. 7
Fig. 8

Similar content being viewed by others

Data availability

The manuscript provides all the data in figures and tables.

References

  1. Manoharan S, Pugalendhi P. Breast cancer: an overview. J Cell Tissue Res. 2010;10(3):2423.

    Google Scholar 

  2. Torre LA, et al. Global cancer in women: burden and trends. Cancer Epidemiol Biomark Prev. 2017;26(4):444–57.

    Article  Google Scholar 

  3. Russo J, et al. Cancer risk related to mammary gland structure and development. Microsc Res Tech. 2001;52(2):204–23.

    Article  CAS  PubMed  Google Scholar 

  4. Sahu A, et al. In-silico and in-vitro study reveals Ziprasidone as a potential aromatase inhibitor against breast carcinoma. Sci Rep. 2023;13(1):16545.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sathish D, et al. Medical imaging techniques and computer aided diagnostic approaches for the detection of breast cancer with an emphasis on thermography-a review. Int J Med Eng Inf. 2016;8(3):275–99.

    Google Scholar 

  6. Olopade OI, et al. Advances in breast cancer: pathways to personalized medicine. Clin Cancer Res. 2008;14(24):7988–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rana M, et al. Design, synthesis, X-ray crystal structures, anticancer, DNA binding, and molecular modelling studies of pyrazole–pyrazoline hybrid derivatives. RSC Adv. 2023;13(38):26766–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cherny NI, Paluch-Shimon S, Berner-Wygoda Y. Palliative care: needs of advanced breast cancer patients. Breast Cancer. 2018;10:231–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Famuyiwa SO, et al. Comprehensive computational studies of naturally occurring kuguacins as antidiabetic agents by targeting visfatin. Chem Afr. 2023;6:1–13.

    Google Scholar 

  10. Feng Y, et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 2018;5(2):77–106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Page BD, et al. Targeted NUDT5 inhibitors block hormone signaling in breast cancer cells. Nat Commun. 2018;9(1):250.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Lamb KM, et al. Elucidating features that drive the design of selective antifolates using crystal structures of human dihydrofolate reductase. Biochemistry. 2013;52(41):7318–26.

    Article  CAS  PubMed  Google Scholar 

  13. Ishikawa T, et al. Design and synthesis of novel human epidermal growth factor receptor 2 (HER2)/epidermal growth factor receptor (EGFR) dual inhibitors bearing a pyrrolo [3, 2-d] pyrimidine scaffold. J Med Chem. 2011;54(23):8030–50.

    Article  CAS  PubMed  Google Scholar 

  14. Stamos J, Sliwkowski MX, Eigenbrot C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J Biol Chem. 2002;277(48):46265–72.

    Article  CAS  PubMed  Google Scholar 

  15. Rose PW, et al. The RCSB Protein Data Bank: redesigned web site and web services. Nucl Acids Res. 2010;39(suppl 1):392–401.

    Google Scholar 

  16. Rana M, et al. Synthesis, single crystal, TD-DFT, molecular dynamics simulation and DNA binding studies of carbothioamide analog. J Mol Struct. 2023;1287: 135701.

    Article  CAS  Google Scholar 

  17. Release S. Protein preparation wizard. Epik. New York: Schrödinger LLC; 2023.

    Google Scholar 

  18. Schrödinger L. Maestro. New York: Schrödinger; 2023.

    Google Scholar 

  19. Jacobson MP, et al. A hierarchical approach to all-atom protein loop prediction. Proteins. 2004;55(2):351–67.

    Article  CAS  PubMed  Google Scholar 

  20. Release S. Prime. New York: Schrödinger LLC; 2023.

    Google Scholar 

  21. Release S. Epik. New York: Schrödinger; 2023.

    Google Scholar 

  22. Li H, Robertson AD, Jensen JH. Very fast empirical prediction and rationalization of protein pKa values. Proteins. 2005;61(4):704–21.

    Article  CAS  PubMed  Google Scholar 

  23. Ahmad S, et al. Multitargeted molecular dynamic understanding of butoxypheser against SARS-CoV-2: An in silico study. Nat Prod Commun. 2022;17(7):1934578X221115499.

    CAS  Google Scholar 

  24. Ahmad S, et al. Therapeutic protein-based vaccines. In: Protein-based therapeutics. Springer; 2023. p. 355–84.

    Chapter  Google Scholar 

  25. Jorgensen WL, Maxwell DS, Tirado-Rives J. Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids. J Am Chem Soc. 1996;118(45):11225–36.

    Article  CAS  Google Scholar 

  26. Jorgensen WL, Tirado-Rives J. The OPLS [optimized potentials for liquid simulations] potential functions for proteins, energy minimizations for crystals of cyclic peptides and crambin. J Am Chem Soc. 1988;110(6):1657–66.

    Article  CAS  PubMed  Google Scholar 

  27. Karwasra R, et al. Macrophage-targeted punicalagin nanoengineering to alleviate methotrexate-induced neutropenia: a molecular docking, DFT, and MD simulation analysis. Molecules. 2022;27(18):6034.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Karwasra R, et al. The incipient role of computational intelligence in oncology: drug designing, discovery, and development. In: Computational intelligence in oncology. Springer; 2022. p. 369–84.

    Chapter  Google Scholar 

  29. Kaul T, et al. Probing the effect of a plus 1bp frameshift mutation in protein-DNA interface of domestication gene, NAMB1, in wheat. J Biomol Struct Dyn. 2019;38(12):3633–47.

    Article  PubMed  Google Scholar 

  30. Huang R, et al. The NCGC pharmaceutical collection: a comprehensive resource of clinically approved drugs enabling repurposing and chemical genomics. Sci Transl Med. 2011;3(80):8016.

    Article  Google Scholar 

  31. Release S. LigPrep. New York: Schrödinger LLC; 2023.

    Google Scholar 

  32. Shah AA, et al. Structure-based virtual screening, molecular docking, molecular dynamics simulation, and metabolic reactivity studies of quinazoline derivatives for their anti-EGFR activity against tumor angiogenesis. Curr Med Chem. 2024;31(5):595–619.

    Article  CAS  PubMed  Google Scholar 

  33. Singh AP, et al. Computational screening and MM/GBSA-based MD simulation studies reveal the high binding potential of FDA-approved drugs against Cutibacterium acnes sialidase. J Biomol Struct Dyn. 2023;2023:1–11.

    Google Scholar 

  34. Friesner RA, et al. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein−ligand complexes. J Med Chem. 2006;49(21):6177–96.

    Article  CAS  PubMed  Google Scholar 

  35. Release S. Glide. New York: Schrödinger; 2023.

    Google Scholar 

  36. Alghamdi YS, et al. Unveiling the multitargeted potential of N-(4-Aminobutanoyl)-S-(4-methoxybenzyl)-L-cysteinylglycine (NSL-CG) against SARS CoV-2: a virtual screening and molecular dynamics simulation study. J Biomol Struct Dyn. 2023;41(14):6633–42.

    Article  CAS  PubMed  Google Scholar 

  37. Alturki NA, et al. In-silico screening and molecular dynamics simulation of drug bank experimental compounds against SARS-CoV-2. Molecules. 2022;27(14):4391.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Alzamami A, et al. Hemi-Babim and fenoterol as potential inhibitors of MPro and papain-like protease against SARS-CoV-2: an in-silico study. Medicina. 2022;58(4):515.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bhati R, et al. Structural–functional analysis and molecular characterization of arsenate reductase from Enterobacter cloacae RSC3 for arsenic biotransformation. 3 Biotech. 2023;13(9):305.

    Article  PubMed  Google Scholar 

  40. Lipinski CA. Lead-and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol. 2004;1(4):337–41.

    Article  CAS  PubMed  Google Scholar 

  41. Release S. QikProp. New York: Schrödinger LLC; 2023.

    Google Scholar 

  42. Ahmad S, et al. Reporting dinaciclib and theodrenaline as a multitargeted inhibitor against SARS-CoV-2: an in-silico study. J Biomol Struct Dyn. 2023;41(9):4013–23.

    Article  CAS  PubMed  Google Scholar 

  43. Ahmad S, Raza K. Identification of 5-nitroindazole as a multitargeted inhibitor for CDK and transferase kinase in lung cancer: a multisampling algorithm-based structural study. Mol Divers. 2023;2023:1–14.

    Google Scholar 

  44. Ahmad S, et al. Multisampling-based docking reveals Imidazolidinyl urea as a multitargeted inhibitor for lung cancer: an optimisation followed multi-simulation and in-vitro study. J Biomol Struct Dyn. 2023;2023:1–18.

    Google Scholar 

  45. Tripathi MK, et al. Fundamentals of molecular modeling in drug design. In: Computer Aided Drug Design (CADD): from ligand-based methods to structure-based approaches. Elsevier; 2022. p. 125–55.

    Chapter  Google Scholar 

  46. Yadav MK, et al. Predictive modeling and therapeutic repurposing of natural compounds against the receptor-binding domain of SARS-CoV-2. J Biomol Struct Dyn. 2022;2022:1–13.

    Google Scholar 

  47. Bochevarov AD, et al. Jaguar: a high-performance quantum chemistry software program with strengths in life and materials sciences. Int J Quantum Chem. 2013;113(18):2110–42.

    Article  CAS  Google Scholar 

  48. Ganji MD, Hosseini-Khah S, Amini-Tabar Z. Theoretical insight into hydrogen adsorption onto graphene: a first-principles B3LYP-D3 study. Phys Chem Chem Phys. 2015;17(4):2504–11.

    Article  Google Scholar 

  49. Karplus M, Petsko GA. Molecular dynamics simulations in biology. Nature. 1990;347(6294):631–9.

    Article  CAS  PubMed  Google Scholar 

  50. Ahmad S, et al. Natural product-based drug designing for treatment of human parasitic diseases. In: Natural product based drug discovery against human parasites: opportunities and challenges. Springer; 2023. p. 37–59.

    Chapter  Google Scholar 

  51. Yadav MK, et al. Application of machine learning–based approaches in stem cell research. In: Computational biology for stem cell research. Elsevier; 2024. p. 65–76.

    Chapter  Google Scholar 

  52. Bowers KJ, et al. Scalable algorithms for molecular dynamics simulations on commodity clusters. In: Proceedings of the 2006 ACM/IEEE Conference on supercomputing. 2006.

  53. Mark P, Nilsson L. Structure and dynamics of the TIP3P, SPC, and SPC/E water models at 298 K. J Phys Chem A. 2001;105(43):9954–60.

    Article  CAS  Google Scholar 

  54. McDonald I. NpT-ensemble Monte Carlo calculations for binary liquid mixtures. Mol Phys. 1972;23(1):41–58.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors extend their appreciation to Taif University, Saudi Arabia, for supporting this work through project number (TU-DSPP-2024-13).

Funding

This research was funded by Taif University, Saudi Arabia with project No: TU-DSPP-2024-13.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualisation, funding, analysis, visualisation, writing the first draft, supervising the whole research project and refining the manuscript for publication; Ahad Amer Alsaiari. Conceptualisation, analysis, visualisation, writing the first draft; Amal F.Gharib. Data Collection, Analysis, visualisation, writing the first draft, extensive editing; Maha Mahfouz Bakhuraysah. Data preparation, processing, analysis, figure conversion and editing first draft; Amani A. Alrehaili. Reviewed and edited the manuscript and helped to revise; Shatha M.Algethami. Data Collection, Analysis, visualisation, writing the first draft, extensive editing; Hayfa Ali Alsaif. Data Collection, Analysis, visualisation, writing the first draft; Norah Al harthi. Conceptualisation, supervision, analysis, software, visualisation, writing and reviewing-editing the first draft; Mohammed Ageeli Hakami.

Corresponding author

Correspondence to Mohammed Ageeli Hakami.

Ethics declarations

Conflict of interest

The authors declare no competing or conflicts of interest.

Additional information

Publisher's Note

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

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

Alsaiari, A.A., Gharib, A.F., Bakhuraysah, M.M. et al. Chlordiazepoxide against signalling, receptor and regulatory proteins of breast cancer: a structure-based in-silico approach. Med Oncol 41, 117 (2024). https://doi.org/10.1007/s12032-024-02366-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s12032-024-02366-w

Keywords

Navigation