Drug Res (Stuttg) 2023; 73(06): 355-364
DOI: 10.1055/a-2062-3571
Original Article

Interaction of Some Amino-Nitrile Derivatives with Vascular Endothelial Growth Factor Receptor 1 (VEGFR1) Using a Theoretical Model

Lauro Figueroa-Valverde
1   Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
,
Francisco Díaz-Cedillo
2   Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional. Santo Tomas, México
,
Marcela Rosas-Nexticapa
3   Facultad de Nutrición, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
,
Magdalena Alvarez-Ramirez
3   Facultad de Nutrición, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
,
Maria Virginia Mateu-Armad
3   Facultad de Nutrición, Universidad Veracruzana, Médicos y Odontologos, Unidad del Bosque Xalapa Veracruz, México
,
Maria López-Ramos
1   Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
,
Tomas López-Gutierrez
1   Laboratory of Pharmaco-Chemistry, Faculty of Chemical Biological Sciences, University Autonomous of Campeche, Campeche, Camp., México
› Author Affiliations

Abstract

Background Some studies indicate that the angiogenesis process is related to vascular endothelial growth factor, which can interact with endothelial cell surface receptors (VEGF-R1, VEGF-R2, and VEGF-R3); this biochemical process and other factors result in the promotion and growth of new blood vessels under normal conditions. However, some studies indicate that this phenomenon could also occur in cancer cells. It is important to mention that some amino derivatives have been prepared as VEGF-R1 inhibitors; however, their interaction with VEGF-R1 is not clear, perhaps due to different experimental approaches or differences in their chemical structure.

Objective The aim of this study was to evaluate the theoretical interaction of several amino-nitrile derivatives (Compounds 1 to 38) with VEGF-R1.

Methods The theoretical interaction of amino-nitrile derivatives with VEGF-R1 was carried out using the 3hng protein as the theoretical model. In addition, cabozantinib, pazopanib, regorafenib, and sorafenib were used as controls in the DockingServer program.

Results The results showed different amino acid residues involved in the interaction of amino-nitrile derivatives with the 3hng protein surface compared with the controls. In addition, the inhibition constant (Ki) was lower for Compounds 10 and 34 than for cabozantinib. Other results show that Ki for Compounds 9, 10, 14, 27–29 and 34–36 was lower in comparison with pazopanib, regorafenib, and sorafenib.

Conclusions All theoretical data suggest that amino-nitrile derivatives could produce changes in the growth of some cancer cell lines through VEGFR-1 inhibition. Therefore, these amino-nitrile derivatives could be a therapeutic alternative to treat some types of cancer.



Publication History

Received: 22 February 2023

Accepted: 20 March 2023

Article published online:
12 May 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Vane J, Änggård E, Botting R. Regulatory functions of the vascular endothelium. New Eng J Med 1990; 323: 27-36
  • 2 Bouïs D, Hospers G, Meijer C. et al. Endothelium in vitro: a review of human vascular endothelial cell lines for blood vessel-related research. Angiogenesis 2001; 4: 91-102
  • 3 Ge L, Xun C, Li W. et al. Extracellular vesicles derived from hypoxia-preconditioned olfactory mucosa mesenchymal stem cells enhance angiogenesis via miR-612. J Nanobiotech 2021; 19: 1-23
  • 4 Lee H, Xu Y, He L. et al. Role of venous endothelial cells in developmental and pathologic angiogenesis. Circulation 2021; 144: 1308-1322
  • 5 Mezu-Ndubuisi O, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediat Res 2021; 89: 1619-1626
  • 6 Hu Y, Tao R, Chen L. et al. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J Nanobiotech 2021; 19: 1-17
  • 7 Depaz- Linares G, Opperman R. et al. Prostaglandin E2 receptor 4 (EP4) as a therapeutic target to impede breast cancer-associated angiogenesis and lymphangiogenesis. Cancers 2021; 13: 942
  • 8 Shibuya M. Vascular endothelial growth factor receptor-1 (VEGFR-1/Flt-1): a dual regulator for angiogenesis. Angiogenesis 2006; 4: 225-230
  • 9 Rahimi N, Dayanir V, Lashkari K. Receptor chimeras indicate that the vascular endothelial growth factor receptor-1 (VEGFR-1) modulates mitogenic activity of VEGFR-2 in endothelial cells. J Biol Chem 2000; 275: 16986-16992
  • 10 Lee Y, Karl D, Maduekwe U. et al. Differential effects of VEGFR-1 and VEGFR-2 inhibition on tumor metastases based on host organ environment. Cancer Res 2010; 70: 8357-8367
  • 11 Wang X, Bove A, Simone G. et al. Molecular bases of VEGFR-2-mediated physiological function and pathological role. Front Cell Dev Biol 2020; 8: 599281
  • 12 Shibuya M. Vascular endothelial growth factor receptor-1 (VEGFR-1/Flt-1): a dual regulator for angiogenesis. Angiogenesis 2006; 9: 225-230
  • 13 Carmeliet P. VEGF as a key mediator of angiogenesis in cancer. Oncology 2005; 69: 4-10
  • 14 Zhao Y, Guo S, Deng J. et al. VEGF/VEGFR-targeted therapy and immunotherapy in non-small cell lung cancer: targeting the tumor microenvironment. Int J Biol Sci 2922 18: 3845-3858
  • 15 Beigom H, Barari A, Abbasi D. et al. The Effect of Eight-week Concurrent Training On The Plasma And Gene Expression Levels VEGFR-1 and VEGFR-2 in Men with Prostate Cancer. J Neyshabur Univ Med Sci 2020; 8: 118-131
  • 16 Mabeta P, Steenkamp V. The VEGF/VEGFR Axis Revisited: Implications for Cancer Therapy. International J Mol Sci 2022; 23: 1-14
  • 17 Bando H, Weich H, Brokelmann M. et al. Association between intratumoral free and total VEGF, soluble VEGFR-1, VEGFR-2 and prognosis in breast cancer. British J Cancer 2005; 92: 553-561
  • 18 Garvin S, Nilsson U, Dabrosin C. Effects of oestradiol and tamoxifen on VEGF, soluble VEGFR-1, and VEGFR-2 in breast cancer and endothelial cells. British J Cancer 2005; 93: 1005-1010
  • 19 Ustuner Z, Saip P, Yasasever V. et al. Prognostic and predictive value of vascular endothelial growth factor and its soluble receptors, VEGFR-1 and VEGFR-2 levels in the sera of small cell lung cancer patients. Med Oncol 2008; 25: 394-399
  • 20 Wang H, Li K, Dou K. et al. Expression of vascular endothelial growth factor (VEGF) and its receptors in human hepatocellular carcinoma cell lines. Chinese J Cell Mol Immun 2001; 359-361
  • 21 Li T, Zhu Y, Qin C. et al. Expression and prognostic significance of vascular endothelial growth factor receptor 1 in hepatocellular carcinoma. J Clin Pathol 2012; 65: 808-814
  • 22 Duke E, Barone A, Chatterjee S. et al. FDA Approval Summary: Cabozantinib for Differentiated Thyroid Cancer. Clin Cancer Res 2022; 28: 4173-4177
  • 23 Sloan B, Scheinfeld N. Pazopanib, a VEGF receptor tyrosine kinase inhibitor for cancer therapy. Curr Opinion Invest drugs 2008; 9: 1324-1335
  • 24 Wilhelm S, Carter C, Lynch M. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nature Rev Drug Dis 2006; 5: 835-844
  • 25 Ibrahim N, Yu Y, Walsh W. et al. Molecular targeted therapies for cancer: sorafenib monotherapy and its combination with other therapies. Oncol Rep 2012; 27: 1303-1311
  • 26 Liang X, Yang Q, Wu P. et al. The synthesis review of the approved tyrosine kinase inhibitors for anticancer therapy in 2015-2020. Bioorg Chem 2021; 113: 105011
  • 27 Bekaii-Saab T, Ou F, Ahn D. et al. Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study. Lancet Oncol 2019; 20: 1070-1082
  • 28 Zhang Q, Chen M, Wang Z. et al. Efficacy and safety comparison of regorafenib and fruquintinib in metastatic colorectal cancer-an observational cohort study in the real world. Clin Col Cancer 2022; 21: e152-e161
  • 29 Van-Boxtel W, Uijen M, Krens S. et al. Excessive toxicity of cabozantinib in a phase II study in patients with recurrent and/or metastatic salivary gland cancer. Eur J Cancer 2022; 161: 128-137
  • 30 Li J, Guo W, Bai Y. et al. Safety profile and adverse events of special interest for Fruquintinib in Chinese patients with previously treated metastatic colorectal cancer: Analysis of the phase 3 FRESCO trial. Adv Ther 2020; 37: 4585-4598
  • 31 Milling R, Grimm D, Krüger M. et al. Pazopanib, cabozantinib, and vandetanib in the treatment of progressive medullary thyroid cancer with a special focus on the adverse effects on hypertension. Int J Mol Sci 2018; 19: 1-15
  • 32 Arrieta M, Lázaro A, Rodríguez P. et al. Efectividad y seguridad de regorafenib y trifluridina/tipiracilo en cáncer colorrectal metastático. Rev OFIL 2020; 30: 99-104
  • 33 Kroschinsky F, Stölzel F, Von-Bonin S. et al. New drugs, new toxicities: severe side effects of modern targeted and immunotherapy of cancer and their management. Critical Care 2017; 21: 1-11
  • 34 Manley P, Furet P, Bold G. et al. Anthranilic acid amides: a novel class of antiangiogenic VEGF receptor kinase inhibitors. J Mic Chem 2002; 45: 5687-5693
  • 35 Egert-Schmidt A, Dreher J, Dunkel U. et al. Identification of 2-anilino-9-methoxy-5, 7- dihydro-6 H-pyrimido [5, 4-d][1] benzazepin-6-ones as dual PLK1/VEGF-R2 kinase inhibitor chemotypes by structure-based lead generation. J Med Chem 2010; 53: 2433-2442
  • 36 Kelley RK, Rimassa L, Cheng A. et al. Cabozantinib plus atezolizumab versus sorafenib for advanced hepatocellular carcinoma (COSMIC-312): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2022; 23: 995-1008
  • 37 Shiri P, Ramezanpour S, Amani A. et al. A patent review on efficient strategies for the total synthesis of pazopanib, regorafenib and lenvatinib as novel anti-angiogenesis receptor tyrosine kinase inhibitors for cancer therapy. Mole Diver 2022; 26: 2981-3002
  • 38 Zhang Y, Wang Y, Lei Z. et al. Regorafenib antagonizes BCRP-mediated multidrug resistance in colon cancer. Cancer Lett 2019; 442: 104-112
  • 39 Stăncioiu L, Gherman A, Brezeștean I. et al. Vibrational spectral analysis of Sorafenib and its molecular docking study compared to other TKIs. J Mol Struct 2022; 1248: 131507
  • 40 Figueroa-Valverde L, Diaz-Cedillo F, Gobato R. Design and synthesis of two Strychnidin-oxiran-naphthalenol derivatives and their theoretical evaluation as noradrenaline and serotonin reuptake inhibitors. Vietnam J Chem 2022; 60: 245-256
  • 41 Figueroa-Valverde L, Rosas-Nexticapa M, Montserra M. Synthesis and Theoretical Interaction of 3-(2-oxabicyclo [7.4. 0] trideca-1 (13), 9, 11-trien-7-yn-12-yloxy)-steroid Derivative with 17β-hydroxysteroid Dehydrogenase Enzyme Surface. Biointerface Res Appl Chem 2023; 13: 1-10
  • 42 Bakchi B, Krishna A, Sreecharan E. et al. An overview on applications of SwissADME web tool in the design and development of anticancer, antitubercular and antimicrobial agents: A medicinal chemist’s perspective. J Mol Struc 2022; 132712
  • 43 Aqeel M, Khan A, Ashraf Z. et al. In silico approach for the development of phenolic derivatives as potential anti-angiogenic agents against lysyl oxidase-like 2 enzyme. Future J Pharma Sci 2022; 8: 1-11
  • 44 Da-Rocha M, Marinho E, Marinho M. et al. Virtual screening in pharmacokinetics, bioactivity, and toxicity of the amburana cearensis secondary metabolites. Biointerface Res Appl Chem 2022; 12: 8471-8491
  • 45 Renhowe P, Pecchi S, Shafer C. et al. Design, structure-activity relationships and in vivo characterization of 4-amino-3-benzimidazol-2-ylhydroquinolin-2-ones: a novel class of receptor tyrosine kinase inhibitors. J Med chem 2009; 52: 278-292
  • 46 Abd-Elhameid M, Labib M, Negmeldin A. et al. Design, synthesis, and screening of orthoamino thiophene carboxamide derivatives on hepatocellular carcinomaas VEGFR-2 Inhibitors. J Enz Inh Med Chem 2018; 33: 1472-1493
  • 47 Sun H, Zhuo L, Dong H. et al. Discovery of 8-amino-substituted 2-phenyl-2, 7- naphthyridinone derivatives as new c-kit/vegfr-2 kinase inhibitors. Molecules 2019; 24: 1-13
  • 48 Asano T, Nakamura H, Uehara Y. et al. Design, Synthesis, and Biological Evaluation of Aminoboronic Acids as Growth-Factor Receptor Inhibitors of EGFR and VEGFR -1 Tyrosine Kinases. ChemBioChem 2004; 5: 483-490
  • 49 Wang J, Ishchenko A, Zhang W. et al. A highly accurate metadynamics-based Dissociation Free Energy method to calculate protein-protein and protein-ligand binding potencies. Sci Rep 2022; 12: 1-13
  • 50 Gapsys V, Hahn D, Tresadern G. et al. Pre-exascale computing of protein-ligand binding free energies with open source software for drug design. J Chem Inf Mod 2022; 62: 1172-1177
  • 51 Rosas-Nexticapa M, Figueroa-Valverde L. Alvarez-Ramirez Evaluation of Interaction of Some Quinolone Derivatives on RSK-4 Using a Theoretical Model. Clin Inv Cancer J 2022; 11: 16-20