Skip to main content

Advertisement

Log in

Design, synthesis and molecular docking of new [1,2,4] triazolo[4,3-a]quinoxaline derivatives as anticancer agents targeting VEGFR-2 kinase

  • Original Article
  • Published:
Molecular Diversity Aims and scope Submit manuscript

Abstract

Vascular endothelial growth factor receptor-2 (VEGFR-2) is critically involved in cancer angiogenesis. Blocking of VEGFR-2 signaling pathway proved effective suppression of tumor growth. Accordingly, two series of new triazoloquinoxaline-based derivatives were designed and synthesized as VEGFR-2 inhibitors. All in vitro cytotoxic activities of the synthesized compounds were evaluated against two human cancer cell lines (MCF-7 and HepG2). To confirm the potential mechanism of cytotoxicity, enzymatic assays against VEGFR-2 were estimated for all the target compounds. The results of VEGFR-2 inhibitory activity and cytotoxicity were in high correlation. Compound 22a exhibited the highest cytotoxic effect with IC50 values of 6.2 and 4.9 μM against MCF-7 and HepG2, respectively, comparing to sorafenib (IC50 = 3.53 and 2.18 μM). Such derivative showed the best VEGFR-2 inhibitory activity with an IC50 value of 3.9 nM, which is very close to that of sorafenib (IC50 = 3.13 nM). Moreover, compounds 22b, 23b, and 23e exhibited strong cytotoxic activity with IC50 values ranging from 11.7 to 15.3 μM. Also, these compounds showed promising VEGFR-2 inhibition with IC50 values of 4.2, 5.7, and 4.7 nM, respectively. In silico docking, ADMET, and toxicity studies were carried out for the synthesized compounds. The results revealed that some compounds have a good binding mode against VEGFR-2 and a high level of drug-likeness.

Graphic 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
Scheme 1
Scheme 2
Scheme 3
Scheme 4
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10

Similar content being viewed by others

References

  1. Phillips, CM, Lima EA, Woodall RT, Brock A, Yankeelov TE (2020) A hybrid model of tumor growth and angiogenesis: In silico experiments. PLoS One 15(4):e0231137.

    Article  CAS  Google Scholar 

  2. Li WW, Talcott KE, Zhai AW, Kruger EA, Li VW (2005) The role of therapeutic angiogenesis in tissue repair and regeneration. Adv Skin Wound Care 18(9):491–500

    Article  Google Scholar 

  3. Carmeliet P, Jain RK (2000) Angiogenesis in cancer and other diseases. Nature 407(6801):249–257.

    Article  CAS  Google Scholar 

  4. Makrilia N, Lappa T, Xyla V, Nikolaidis I, Syrigos K (2009) The role of angiogenesis in solid tumours: an overview. Eur J Intern Med 20(7):663–671

    Article  CAS  Google Scholar 

  5. Patel HM, Bari P, Karpoormath R, Noolvi M, Thapliyal N, Surana S, Jain P (2015) Design and synthesis of VEGFR-2 tyrosine kinase inhibitors as potential anticancer agents by virtual based screening. RSC Adv 5(70):56724–56771

    Article  CAS  Google Scholar 

  6. Folkman J (2002) Role of angiogenesis in tumor growth and metastasis, Seminars in oncology. Elsevier, Amsterdam, pp 15–18.

  7. Shibuya M (2013) Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem 153(1):13–19

    Article  CAS  Google Scholar 

  8. Frezzetti D, Gallo M, Roma C, D’Alessio A, Maiello MR, Bevilacqua S, Normanno N, De Luca A (2016) Vascular endothelial growth factor a regulates the secretion of different angiogenic factors in lung cancer cells. J Cell Physiol 231(7):1514–1521

    Article  CAS  Google Scholar 

  9. Stuttfeld E, Ballmer-Hofer K (2009) Structure and function of VEGF receptors. IUBMB Life 61(9):915–922

    Article  CAS  Google Scholar 

  10. El-Metwally SA, Abou-El-Regal MM, Eissa IH, Mehany AB, Mahdy HA, Elkady H, Elwan A, Elkaeed EB (2021) Discovery of thieno [2, 3-d] pyrimidine-based derivatives as potent VEGFR-2 kinase inhibitors and anti-cancer agents. Bioorganic Chem, 104947.

  11. Shahin MI, Abou El Ella DA, Ismail NS, Abouzid KA (2014) Design, synthesis and biological evaluation of type-II VEGFR-2 inhibitors based on quinoxaline scaffold. Bioorg Chem 56:16–26

    Article  CAS  Google Scholar 

  12. Huang L, Huang Z, Bai Z, Xie R, Sun L, Lin K (2012) Development and strategies of VEGFR-2/KDR inhibitors. Future Med Chem 4(14):1839–1852

    Article  CAS  Google Scholar 

  13. Zeidan MA, Mostafa AS, Gomaa RM, Abou-Zeid LA, El-Mesery M, Magda A-A, Selim KB (2019) Design, synthesis and docking study of novel picolinamide derivatives as anticancer agents and VEGFR-2 inhibitors. Eur J Med Chem 168:315–329

    Article  CAS  Google Scholar 

  14. Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, Kelley S (2006) Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discovery 5(10):835–844

    Article  CAS  Google Scholar 

  15. Woo HY, Heo J (2012) Sorafenib in liver cancer. Expert Opin Pharmacother 13(7):1059–1067

    Article  CAS  Google Scholar 

  16. Wilhelm SM, Dumas J, Adnane L, Lynch M, Carter CA, Schütz G, Thierauch KH, Zopf D (2011) Regorafenib (BAY 73–4506): a new oral multikinase inhibitor of angiogenic, stromal and oncogenic receptor tyrosine kinases with potent preclinical antitumor activity. Int J Cancer 129(1):245–255

    Article  CAS  Google Scholar 

  17. Strumberg D, Schultheis B (2012) Regorafenib for cancer. Expert Opin Investig Drugs 21(6):879–889

    Article  CAS  Google Scholar 

  18. Kakarala KK, Jamil K (2021) Identification of novel allosteric binding sites and multi-targeted allosteric inhibitors of receptor and non-receptor tyrosine kinases using a computational approach. J Biomolecular Struct Dyn, pp 1–22.

  19. Rini BI, Pal SK, Escudier BJ, Atkins MB, Hutson TE, Porta C, Verzoni E, Needle MN, McDermott DF (2020) Tivozanib versus sorafenib in patients with advanced renal cell carcinoma (TIVO-3): a phase 3, multicentre, randomised, controlled, open-label study. Lancet Oncol 21(1):95–104

    Article  CAS  Google Scholar 

  20. Albiges L, Barthélémy P, Gross-Goupil M, Negrier S, Needle M, Escudier B (2021) TiNivo: safety and efficacy of tivozanib-nivolumab combination therapy in patients with metastatic renal cell carcinoma. Ann Oncol 32(1):97–102

    Article  CAS  Google Scholar 

  21. Le Tourneau C, Raymond E, Faivre S (2007) Sunitinib: a novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Therapeutics Clinical Risk Management 3(2):341.

  22. Fala L (2015) Lenvima (Lenvatinib), a multireceptor tyrosine kinase inhibitor, approved by the FDA for the treatment of patients with differentiated Thyroid Cancer. Am Health Drug Benefits 8(Spec Feature), 176.

  23. Shirley M (2018) Fruquintinib: first global approval. Drugs 78(16):1757–1761

    Article  Google Scholar 

  24. Abdallah AE, Eissa SI, Al Ward MMS, Mabrouk RR, Mehany AB, El-Zahabi MA (2021) Design, synthesis and molecular modeling of new quinazolin-4 (3H)-one based VEGFR-2 kinase inhibitors for potential anticancer evaluation. Bioorganic Chem 109:104695.

  25. Alanazi MM, Mahdy HA, Alsaif NA, Obiadullah AJ, Alkahtani HM, Al-Mehizia AA, Alsubaie SM, Dahab MA, Eissa IH (2021) New bis ([1, 2, 4] triazolo)[4, 3-a: 3', 4'-c] quinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers: design, synthesis, in silico studies, and anticancer evaluation. Bioorganic Chem, 104949.

  26. Alsaif NA, Dahab MA, Alanazi MM, Obaidullah AJ, Al-Mehizia AA, Alanazi MM, Aldawas S, Mahdy HA, Elkady H (2021) New quinoxaline derivatives as VEGFR-2 inhibitors with anticancer and apoptotic activity: design, molecular modeling, and synthesis. Bioorganic Chem 110:104807.

    Article  CAS  Google Scholar 

  27. El-Adl E, El-Helby AGA, Sakr H, Eissa IH, El-Hddad SS, Shoman FM (2020) Design, synthesis, molecular docking and anticancer evaluations of 5-benzylidenethiazolidine-2, 4-dione derivatives targeting VEGFR-2 enzyme. Bioorganic Chem 102:104059.

    Article  CAS  Google Scholar 

  28. Abou-Seri SM, Eldehna WM, Ali MM, Abou El Ella DA (2016) 1-Piperazinylphthalazines as potential VEGFR-2 inhibitors and anticancer agents: synthesis and in vitro biological evaluation. Eur J Med Chem 107:165–179

    Article  CAS  Google Scholar 

  29. Eldehna WM, Fares M, Ibrahim HS, Aly MH, Zada S, Ali MM, Abou-Seri SM, Abdel-Aziz HA, Abou El Ella DA (2015) Indoline ureas as potential anti-hepatocellular carcinoma agents targeting VEGFR-2: Synthesis, in vitro biological evaluation and molecular docking. Eur J Med Chem 100:89–97

    Article  CAS  Google Scholar 

  30. Sobhy MK, Mowafy S, Lasheen DS, Farag NA, Abouzid KA (2019) 3D-QSAR pharmacophore modelling, virtual screening and docking studies for lead discovery of a novel scaffold for VEGFR 2 inhibitors: design, synthesis and biological evaluation. Bioorganic Chem 89:102988.

  31. El-Adl K, El-Helby AGA, Sakr H, Elwan A (2020) Design, synthesis, molecular docking and anti-proliferative evaluations of [1, 2, 4] triazolo [4, 3-a] quinoxaline derivatives as DNA intercalators and Topoisomerase II inhibitors. Bioorganic Chem 105:104399.

  32. El-Adl K, El-Helby A-GA, Sakr H, Elwan A (2021) [1, 2, 4] Triazolo [4, 3-a] quinoxaline and [1, 2, 4] triazolo [4, 3-a] quinoxaline-1-thiol-derived DNA intercalators: design, synthesis, molecular docking, in silico ADMET profiles and anti-proliferative evaluations. New J Chem 45(2):881–897

    Article  CAS  Google Scholar 

  33. Abbass EM, Khalil AK, Mohamed MM, Eissa IH, El-Naggar AM (2020) Design, efficient synthesis, docking studies, and anticancer evaluation of new quinoxalines as potential intercalative Topo II inhibitors and apoptosis inducers. Bioorganic Chem 104:104255.

  34. Khandan M, Sadeghian-Rizi S, Khodarahmi G, Hassanzadeh F (2018) Synthesis and cytotoxic evaluation of some novel quinoxalinedione diarylamide sorafenib analogues. Research in pharmaceutical sciences 13(2):168

    Article  Google Scholar 

  35. Sadeghian-Rizi S, Khodarahmi GA, Sakhteman A, Jahanian-Najafabadi A, Rostami M, Mirzaei M, Hassanzadeh F (2017) Biological evaluation, docking and molecular dynamic simulation of some novel diaryl urea derivatives bearing quinoxalindione moiety. Res Pharmaceutical Sci 12(6):500

    Article  Google Scholar 

  36. Alanazi MM, Eissa IH, Alsaif NA, Obaidullah AJ, Alanazi WA, Alasmari AF, Albassam H, Elkady H, Elwan A (2021) Design, synthesis, docking, ADMET studies, and anticancer evaluation of new 3-methylquinoxaline derivatives as VEGFR-2 inhibitors and apoptosis inducers. J Enzyme Inhib Med Chem 36(1):1760–1782

    Article  CAS  Google Scholar 

  37. Nawaf A. Alsaif, Mohammed M. Alanazi, Ahmad J. Obaidullah, Abdulrahman A. Al-Mehizia, Manal M. Alanazi, Saleh Aldawas, Alaa Elwan, Hazem Elkady (2021) Discovery of new VEGFR-2 inhibitors based on bis([1, 2, 4]triazolo)[4,3-a:3',4'-c]quinoxaline derivatives as anticancer agents and apoptosis inducers. J Enzyme Inhibition Med Chem 36(1):1093–1114.

  38. Mahdy HA, Ibrahim MK, Metwaly AM, Belal A, Mehany AB, El-Gamal KM, El-Sharkawy A, Elhendawy MA, Radwan MM, Elsohly MA (2020) Design, synthesis, molecular modeling, in vivo studies and anticancer evaluation of quinazolin-4 (3H)-one derivatives as potential VEGFR-2 inhibitors and apoptosis inducers. Bioorganic Chem 94:103422.

    Article  CAS  Google Scholar 

  39. Eissa IH, Ibrahim MK, Metwaly AM, Belal A, Mehany AB, Abdelhady AA, Elhendawy MA, Radwan MM, ElSohly MA, Mahdy HA (2021) Design, molecular docking, in vitro, and in vivo studies of new quinazolin-4 (3H)-ones as VEGFR-2 inhibitors with potential activity against hepatocellular carcinoma. Bioorganic Chem 107:104532.

    Article  CAS  Google Scholar 

  40. Nasser AA, Eissa IH, Oun MR, El-Zahabi MA, Taghour MS, Belal A, Saleh AM, Mehany AB, Luesch H, Mostafa AE (2020) Discovery of new pyrimidine-5-carbonitrile derivatives as anticancer agents targeting EGFR WT and EGFR T790M. Org Biomol Chem 18(38):7608–7634

    Article  CAS  Google Scholar 

  41. El-Zahabi MA, Sakr H, El-Adl K, Zayed M, Abdelraheem AS, Eissa SI, Elkady H, Eissa IH (2020) Design, synthesis, and biological evaluation of new challenging thalidomide analogs as potential anticancer immunomodulatory agents. Bioorganic Chem 104:104218.

    Article  CAS  Google Scholar 

  42. Eissa IH, El-Helby AGA, Mahdy HA, Khalifa MM, Elnagar HA, Mehany AB, Metwaly AM, Elhendawy MA, Radwan MM, ElSohly MA (2020) Discovery of new quinazolin-4 (3H)-ones as VEGFR-2 inhibitors: design, synthesis, and anti-proliferative evaluation. Bioorganic Chem 105:104380.

    Article  Google Scholar 

  43. Ibrahim M-K, Abd-Elrahman AA, Ayyad RR, El-Adl K, Mansour AM, Eissa IH (2013) Design and synthesis of some novel 2-(3-methyl-2-oxoquinoxalin-1 (2H)-yl)-N-(4-(substituted) phenyl) acetamide derivatives for biological evaluation as anticonvulsant agents. Bull Faculty Pharmacy Cairo University 51(1):101–111

    Article  Google Scholar 

  44. Russell MG, Carling RW, Atack JR, Bromidge FA, Cook SM, Hunt P, Isted C, Lucas M, McKernan RM, Mitchinson A (2005) Discovery of functionally selective 7, 8, 9, 10-tetrahydro-7, 10-ethano-1, 2, 4-triazolo [3, 4-a] phthalazines as GABAA receptor agonists at the α3 subunit. J Med Chem 48(5):1367–1383

    Article  CAS  Google Scholar 

  45. Seglen PO (1976) Preparation of isolated rat liver cells. Methods Cell Biol 13:29–83

    Article  CAS  Google Scholar 

  46. El-Helby AGA, Ayyad RR, Zayed MF, Abulkhair HS, Elkady H, El-Adl K (2019) Design, synthesis, in silico ADMET profile and GABA-A docking of novel phthalazines as potent anticonvulsants. Arch Pharm 352(5):1800387

    Article  Google Scholar 

  47. El-Helby A-GA, Ayyad RR, El-Adl K, Elkady H (2019) Phthalazine-1, 4-dione derivatives as non-competitive AMPA receptor antagonists: design, synthesis, anticonvulsant evaluation, ADMET profile and molecular docking. Molecular Diversity 23(2):283–298

    Article  CAS  Google Scholar 

  48. Hasegawa M, Nishigaki N, Washio Y, Kano K, Harris PA, Sato H, Mori I, West RI, Shibahara M, Toyoda H (2007) Discovery of novel benzimidazoles as potent inhibitors of TIE-2 and VEGFR-2 tyrosine kinase receptors. J Med Chem 50(18):4453–4470

    Article  CAS  Google Scholar 

  49. Mena-Ulecia K, Tiznado W, Caballero J (2015) Study of the differential activity of thrombin inhibitors using docking, QSAR, molecular dynamics, and MM-GBSA, PLoS One 10(11):e0142774.

  50. Xia X, Maliski EG, Gallant P, Rogers D (2004) Classification of kinase inhibitors using a Bayesian model. J Med Chem 47(18):4463–4470

    Article  CAS  Google Scholar 

  51. BIOVIA, QSAR, ADMET and Predictive Toxicology. https://www.3dsbiovia.com/products/collaborative-science/biovia-discovery-studio/qsar-admet-and-predictive-toxicology.html. (Accessed May 2020).

  52. Ismail RS, Abou-Seri SM, Eldehna WM, Ismail NS, Elgazwi SM, Ghabbour HA, Ahmed MS, Halaweish FT, Abou El Ella DA (2018) Novel series of 6-(2-substitutedacetamido)-4-anilinoquinazolines as EGFR-ERK signal transduction inhibitors in MCF-7 breast cancer cells. Eur J Med Chem 155:782–796

    Article  CAS  Google Scholar 

  53. Said MA, Eldehna WM, Nocentini A, Fahim SH, Bonardi A, Elgazar AA, Kryštof V, Soliman DH, Abdel-Aziz HA, Gratteri P (2020) Sulfonamide-based ring-fused analogues for CAN508 as novel carbonic anhydrase inhibitors endowed with antitumor activity: design, synthesis, and in vitro biological evaluation. Euro J Med Chem189:112019.

  54. Li, N, Wang Y, Li W, Li H, Yang L, Wang J, Mahdy HA, Mehany A, Jaiash DA, Santali EY (2020) Screening of some sulfonamide and sulfonylurea derivatives as anti-Alzheimer’s agents targeting BACE1 and PPARγ. J Chem.

  55. El-Zahabi MA, Elbendary ER, Bamanie FH, Radwan MF, Ghareib SA, Eissa IH (2019) Design, synthesis, molecular modeling and anti-hyperglycemic evaluation of phthalimide-sulfonylurea hybrids as PPARγ and SUR agonists. Bioorganic Chem 91:103115.

Download references

Acknowledgements

The authors extend their appreciation to the Deanship of Scientific Research at King Saud University for funding this work through research group no (RG-1441-364).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nawaf A. Alsaif.

Ethics declarations

Conflict of interest

The authors declare that no conflict of interest exists. The authors thank Dr. Mohamed R. Elnagar, Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt, for his technical assistance.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 12877 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alsaif, N.A., Elwan, A., Alanazi, M.M. et al. Design, synthesis and molecular docking of new [1,2,4] triazolo[4,3-a]quinoxaline derivatives as anticancer agents targeting VEGFR-2 kinase. Mol Divers 26, 1915–1932 (2022). https://doi.org/10.1007/s11030-021-10303-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11030-021-10303-6

Keywords

Navigation