898
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Novel N-Arylmethyl-aniline/chalcone hybrids as potential VEGFR inhibitors: synthesis, biological evaluations, and molecular dynamic simulations

ORCID Icon, , , , , , , , , ORCID Icon, ORCID Icon & ORCID Icon show all
Article: 2278022 | Received 25 Aug 2023, Accepted 25 Oct 2023, Published online: 20 Nov 2023

References

  • Diori K, Hamarat S. Reviewing cancer’s biology: an eclectic approach. J Egypt Natl Canc Inst. 2021;33:1–17.
  • Gasparini G, Longo R, Toi M, Ferrara N. Angiogenic inhibitors: a new therapeutic strategy in oncology. Nat Clin Pract Oncol. 2005;2(11):562–577.
  • Saman H, Raza SS, Uddin S, Rasul K. Inducing angiogenesis, a key step in cancer vascularization, and treatment approaches. Cancers (Basel). 2020;12(5):1172.
  • Liang P, Ballou B, Lv X, Si W, Bruchez MP, Huang W, Dong X. Monotherapy and combination therapy using anti‐angiogenic nanoagents to fight cancer. Adv Mater. 2021;33(15):2005155.
  • Haibe Y, Kreidieh M, El Hajj H, Khalifeh I, Mukherji D, Temraz S, Shamseddine A. Resistance mechanisms to anti-angiogenic therapies in cancer. Front Oncol. 2020;10:221.
  • Qian Y, Han Q, Chen W, Song J, Zhao X, Ouyang Y, Yuan W, Fan C. Platelet-rich plasma derived growth factors contribute to stem cell differentiation in musculoskeletal regeneration. Front Chem. 2017;5:89.
  • Shibuya M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti-and pro-angiogenic therapies. Genes Cancer. 2011;2(12):1097–1105.
  • Takahashi H, Shibuya M. The vascular endothelial growth factor (VEGF)/VEGF receptor system and its role under physiological and pathological conditions. Clin Sci (Lond)). 2005;109(3):227–241.
  • Pauli SA, Tang H, Wang J, Bohlen P, Posser R, Hartman T, Sauer MV, Kitajewski J, Zimmermann R. The vascular endothelial growth factor (vegf)/vegf receptor 2 pathway is critical for blood vessel survival in corpora lutea of pregnancy in the rodent. Endocrinol. 2005;146(3):1301–1311.
  • Dang Y-Z, Zhang Y, Li J-P, Hu J, Li W-W, Li P, Wei L-C, Shi M. High vegfr1/2 expression levels are predictors of poor survival in patients with cervical cancer. Medicine (Baltimore)). 2017;96(1):e5772.
  • Dakowicz D, Zajkowska M, Mroczko B. Relationship between vegf family members, their receptors and cell death in the neoplastic transformation of colorectal cancer. Int J Mol Sci. 2022;23(6):3375.
  • Ding M, Liu L, Hu C, Liu Y, Qiao Y, Jiang X. Expression of vegfr2 and nrp-1 in non-small cell lung cancer and their clinical significance. Chin J Cancer Res. 2014;26(6):669.
  • Choi SB, Han HJ, Kim WB, Song TJ, Choi S. Vegf overexpression predicts poor survival in hepatocellular carcinoma. Open Med (Wars)). 2017;12(1):430–439.
  • Song SH, Jeong IG, You D, Hong JH, Hong B, Song C, Jung WY, Cho YM, Ahn H, Kim C, et al. Vegf/vegfr2 and pdgf-b/pdgfr-β expression in non-metastatic renal cell carcinoma: A retrospective study in 1,091 consecutive patients. Int J Clin Exp Pathol. 2014;7(11):7681.
  • Otrock ZK, Makarem JA, Shamseddine A. Vascular endothelial growth factor family of ligands and receptors. Blood Cells Mol Dis. 2007;38(3):258–268.
  • Abdullaziz MA, Abdel-Mohsen HT, El Kerdawy AM, Ragab FA, Ali MM, Abu-Bakr SM, Girgis AS, El Diwani H. Design, synthesis, molecular docking and cytotoxic evaluation of novel 2-furybenzimidazoles as vegfr-2 inhibitors. Eur J Med Chem. 2017;136:315–329.
  • Kassab AE, Gedawy EM, Hamed MI, Doghish AS, Hassan R. Design, synthesis, anticancer evaluation, and molecular modelling studies of novel tolmetin derivatives as potential VEGFR-2 inhibitors and apoptosis inducers. J Enzyme Inhib Med Chem. 2021;36(1):922–939.
  • Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, Chinot O. Bevacizumab (avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 2020;86:102017.
  • Lee C, Chen R, Sun G, Liu X, Lin X, He C, Xing L, Liu L, Jensen LD, Kumar A, et al. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 2023;8(1):305.
  • Ciombor KK, Berlin J. Aflibercept—a decoy vegf receptor. Curr Oncol Rep. 2014;16(2):368.
  • De Groot AC. Dermatological drugs, topical agents and cosmetics. In: Side effects of drugs annual. Amsterdam, Netherlands: Elsevier; 1992.
  • Stanel SC, Sjöberg J, Salmonson T, Foggi P, Caleno M, Melchiorri D, Gravanis I, Tzogani K, Pignatti F. European medicines agency approval summary: Zaltrap for the treatment of patients with oxaliplatin-resistant metastatic colorectal cancer. ESMO Open. 2017;2(2):e000190.
  • Javle M, Smyth EC, Chau I. Ramucirumab: successfully targeting angiogenesis in gastric cancer. Clin Cancer Res. 2014;20(23):5875–5881.
  • Cheng Y, Zhang T, Xu Q. Therapeutic advances in non‐small cell lung cancer: Focus on clinical development of targeted therapy and immunotherapy. MedComm (2020)). 2021;2(4):692–729.
  • Ghalehbandi S, Yuzugulen J, Pranjol MZI, Pourgholami M. The role of vegf in cancer-induced angiogenesis and research progress of drugs targeting vegf. Eur J Pharmacol. 2023; 949:175586.
  • Yang F, Jove V, Xin H, Hedvat M, Van Meter TE, Yu H. Sunitinib induces apoptosis and growth arrest of medulloblastoma tumor cells by inhibiting stat3 and akt signaling pathways. Mol Cancer Res. 2010;8(1):35–45.
  • Grosse J, Warnke E, Wehland M, Pietsch J, Pohl F, Wise P, Magnusson NE, Eilles C, Grimm D. Mechanisms of apoptosis in irradiated and sunitinib-treated follicular thyroid cancer cells. Apoptosis. 2014;19(3):480–490.
  • Varinska L, van Wijhe M, Belleri M, Mitola S, Perjesi P, Presta M, Koolwijk P, Ivanova L, Mojzis J. Anti-angiogenic activity of the flavonoid precursor 4-hydroxychalcone. Eur J Pharmacol. 2012;691(1-3):125–133.
  • Lee YS, Lim SS, Shin KH, Kim YS, Ohuchi K, Jung SH, Bulletin P. Anti-angiogenic and anti-tumor activities of 2′-hydroxy-4′-methoxychalcone. Biol Pharm Bull. 2006;29(5):1028–1031.
  • Ahmed MF, Santali EY, El-Haggar R. Novel piperazine–chalcone hybrids and related pyrazoline analogues targeting vegfr-2 kinase; design, synthesis, molecular docking studies, and anticancer evaluation. J Enzyme Inhib Med Chem. 2021;36(1):307–318.
  • Eissa IH, El-Haggar R, Dahab MA, Ahmed MF, Mahdy HA, Alsantali RI, Elwan A, Masurier N, Fatahala S. Design, synthesis, molecular modeling and biological evaluation of novel benzoxazole-benzamide conjugates via a 2-thioacetamido linker as potential anti-proliferative agents, vegfr-2 inhibitors and apoptotic inducers. J Enzyme Inhib Med Chem. 2022;37(1):1587–1599.
  • Ahmed MF, El‐Haggar R, Almalki AH, Abdullah O, El Hassab MA, Masurier N, Hammad S. Novel hydrazone‐isatin derivatives as potential egfr inhibitors: synthesis and in vitro pharmacological profiling. Arch. Pharm. 2023;356(9):2300244.
  • Baccon-Sollier PL, Malki Y, Maye M, Ali LM, Lichon L, Cuq P, Vincent L-A, Masurier N. Imidazopyridine-fused [1, 3] diazepinones: Modulations of positions 2 to 4 and their impacts on the anti-melanoma activity. J Enzyme Inhib Med Chem. 2020;35(1):935–949.
  • Bellet V, Lichon L, Arama DP, Gallud A, Lisowski V, Maillard LT, Garcia M, Martinez J, Masurier N. Imidazopyridine-fused [1, 3]-diazepinones part 2: structure-activity relationships and antiproliferative activity against melanoma cells. Eur J Med Chem. 2017;125:1225–1234.
  • Peyressatre M, Arama DP, Laure A, González-Vera JA, Pellerano M, Masurier N, Lisowski V, Morris M. Identification of quinazolinone analogs targeting cdk5 kinase activity and glioblastoma cell proliferation. Front Chem. 2020;8:691.
  • Gallud A, Vaillant O, Maillard LT, Arama DP, Dubois J, Maynadier M, Lisowski V, Garcia M, Martinez J, Masurier N. Imidazopyridine-fused [1, 3]-diazepinones: synthesis and antiproliferative activity. Eur J Med Chem. 2014;75:382–390.
  • Modi SJ, Kulkarni V. Vascular endothelial growth factor receptor (vegfr-2)/kdr inhibitors: Medicinal chemistry perspective. Med Drug Discov. 2019;2:100009.
  • Boyd MR, Paull K. Some practical considerations and applications of the national cancer institute in vitro anticancer drug discovery screen. Drug Dev Res. 1995;34(2):91–109.
  • Alley MC, Scudiero DA, Monks A, Hursey ML, Czerwinski MJ, Fine DL, Abbott BJ, Mayo JG, Shoemaker RH, Boyd M. Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay. Cancer Res. 1988;48(3):589–601.
  • Shoemaker R. The nci60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6(10):813–823.
  • USA: Developmental Therapeutic Program [2021; https://dtp.cancer.gov/.
  • Koch A, Tamez P, Pezzuto J, Soejarto D. Evaluation of plants used for antimalarial treatment by the maasai of kenya. J Ethnopharmacol. 2005;101(1-3):95–99.
  • Badisa R, Ayuk-Takem L, Ikediobi C, Walker E. Selective anticancer activity of pure licamichauxiioic-b acid in cultured cell lines. Pharm Biol. 2006;44(2):141–145.
  • Wardihan Rusdi M, Alam G, Lukman  , Manggau MA. Selective cytotoxicity evaluation in anticancer drug screening of boehmeria virgata (forst) guill leaves to several human cell lines: Hela, widr, t47d and vero. Dhaka Univ J Pharm Sci. 2014;12(2):87–90.
  • Acton EM, Narayanan VL, Risbood PA, Shoemaker RH, Vistica DT, Boyd M. Anticancer specificity of some ellipticinium salts against human brain tumors in vitro. J Med Chem. 1994;37(14):2185–2189.
  • Ferrari G, Pintucci G, Seghezzi G, Hyman K, Galloway AC, Mignatti P. Vegf, a prosurvival factor, acts in concert with tgf-β1 to induce endothelial cell apoptosis. Proc Natl Acad Sci U S A. 2006;103(46):17260–17265.
  • Shibuya M. Vascular endothelial growth factor receptor‐2: its unique signaling and specific ligand, vegf‐e. Cancer Sci. 2003;94(9):751–756.
  • Rizvi SU, Siddiqui HL, Nisar M, Khan N, Khan IJ. Discovery and molecular docking of quinolyl-thienyl chalcones as anti-angiogenic agents targeting vegfr-2 tyrosine kinase. Bioorg Med Chem Lett. 2012;22(2):942–944.
  • Synowiec E, Hoser G, Bialkowska-Warzecha J, Pawlowska E, Skorski T, Blasiak J. Doxorubicin differentially induces apoptosis, expression of mitochondrial apoptosis-related genes, and mitochondrial potential in bcr-abl1-expressing cells sensitive and resistant to imatinib. Biomed Res Int. 2015;2015:673512–673519.
  • Pilco-Ferreto N, Calaf G. Influence of doxorubicin on apoptosis and oxidative stress in breast cancer cell lines. Int J Oncol. 2016;49(2):753–762.
  • Zinonos I, Labrinidis A, Liapis V, Hay S, Panagopoulos V, Denichilo M, Ponomarev V, Ingman W, Atkins GJ, Findlay D. Doxorubicin overcomes resistance to drozitumab by antagonizing inhibitor of apoptosis proteins (iaps). Anticancer Res. 2014;34(12):7007–7020.
  • Tacar O, Sriamornsak P, Dass C. Doxorubicin: an update on anticancer molecular action, toxicity and novel drug delivery systems. J Pharm Pharmacol. 2013;65(2):157–170.
  • Kwan YP, Saito T, Ibrahim D, Al-Hassan FMS, Ein Oon C, Chen Y, Jothy SL, Kanwar JR, Sasidharan S. Evaluation of the cytotoxicity, cell-cycle arrest, and apoptotic induction by euphorbia hirta in mcf-7 breast cancer cells. Pharm Biol. 2016;54(7):1223–1236.
  • Kim HS, Lee YS, Kim DK. Doxorubicin exerts cytotoxic effects through cell cycle arrest and fas-mediated cell death. Pharmacology. 2009;84(5):300–309.
  • Kciuk M, Gielecińska A, Mujwar S, Kołat D, Kałuzińska-Kołat Ż, Celik I, Kontek R. Doxorubicin—an agent with multiple mechanisms of anticancer activity. Cells. 2023;12(4):659.
  • Fan LL, Sun GP, Wei W, Wang ZG, Ge L, Fu WZ, Wang H. Melatonin and doxorubicin synergistically induce cell apoptosis in human hepatoma cell lines. World J Gastroenterol. 2010;16(12):1473–1481.
  • Mehraj U, Mir IA, Hussain MU, Alkhanani M, Wani NA, Mir MA. Adapalene and doxorubicin synergistically promote apoptosis of tnbc cells by hyperactivation of the erk1/2 pathway through ros induction. Front Oncol. 2022;12:938052.
  • Amalina ND, Salsabila IA, Zulfin UM, Jenie RI, Meiyanto E. In vitro synergistic effect of hesperidin and doxorubicin downregulates epithelial-mesenchymal transition in highly metastatic breast cancer cells. J Egypt Natl Canc Inst. 2023;35(1):6.
  • Miladiyah I, Yuanita E, Nuryadi S, Jumina J, Haryana SM, Mustofa M. Synergistic effect of 1, 3, 6-trihydroxy-4, 5, 7-trichloroxanthone in combination with doxorubicin on b-cell lymphoma cells and its mechanism of action through molecular docking. Curr Ther Res Clin Exp. 2020;92:100576.
  • Park HK, Lee JE, Lim J, Jo DE, Park SA, Suh PG, Kang B. Combination treatment with doxorubicin and gamitrinib synergistically augments anticancer activity through enhanced activation of bim. BMC Cancer. 2014;14(1):431.
  • Evan GI, Vousden K. Proliferation, cell cycle and apoptosis in cancer. Nature. 2001;411(6835):342–348.
  • Brooks R. Cell cycle commitment and the origins of cell cycle variability. Front Cell Dev Biol. 2021;9:698066.
  • Cohen G. Caspases: the executioners of apoptosis. Biochem J. 1997;326 (Pt 1) (Pt 1):1–16.
  • Reed J, Jurgensmeier J, Matsuyama S. Bcl-2 family proteins and mitochondria. Biochim Biophys Acta. 1998;1366(1-2):127–137.
  • Vogel C, Marcotte E. Insights into the regulation of protein abundance from proteomic and transcriptomic analyses. Nat Rev Genet. 2012;13(4):227–232.
  • Pollman MJ, Naumovski L, Gibbons G. Endothelial cell apoptosis in capillary network remodeling. J Cell Physiol. 1999;178(3):359–370.
  • Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, Hirth PK, Waltenberger J, Voelkel N. Inhibition of vegf receptors causes lung cell apoptosis and emphysema. J Clin Invest. 2000;106(11):1311–1319.
  • Orellana EA, Kasinski AL. Sulforhodamine b (srb) assay in cell culture to investigate cell proliferation. Bio Protoc. 2016;6(21):1984–1984.
  • Ciftci H, Sever B, Bayrak N, Yıldız M, Yıldırım H, Tateishi H, Otsuka M, Fujita M, TuYuN AF. In vitro cytotoxicity evaluation of plastoquinone analogues against colorectal and breast cancers along with in silico insights. Pharmaceuticals. 2022;15(10):1266.
  • Peña-Morán OA, Villarreal ML, Álvarez-Berber L, Meneses-Acosta A, Rodríguez-López V. Cytotoxicity, post-treatment recovery, and selectivity analysis of naturally occurring podophyllotoxins from bursera fagaroides var. Fagaroides on breast cancer cell lines. Molecules. 2016;21(8):1013.
  • Braga CB, Kido LA, Lima EN, Lamas CA, Cagnon VH, Ornelas C, Pilli RA. Enhancing the anticancer activity and selectivity of goniothalamin using ph-sensitive acetalated dextran (ac-dex) nanoparticles: a promising platform for delivery of natural compounds. ACS Biomater Sci Eng. 2020;6(5):2929–2942.
  • Abdelaal MR, Soror SH, Elnagar MR, Haffez H. Revealing the potential application of ec-synthetic retinoid analogues in anticancer therapy. Molecules. 2021;26(2):506.
  • Abdelaal MR, Ibrahim E, Elnagar MR, Soror SH, Haffez H. Augmented therapeutic potential of ec-synthetic retinoids in caco-2 cancer cells using an in vitro approach. Int J Mol Sci. 2022;23(16):9442.
  • Haffez H, Osman S, Ebrahim HY, Hassan Z. Growth inhibition and apoptotic effect of pine extract and abietic acid on mcf-7 breast cancer cells via alteration of multiple gene expressions using in vitro approach. Molecules. 2022;27(1):293.
  • Khedr MA, Abu-Zied KM, Zaghary WA, Aly AS, Shouman DN, Haffez H. Novel thienopyrimidine analogues as potential metabotropic glutamate receptors inhibitors and anticancer activity: synthesis, in-vitro, in-silico, and SAR approaches. Bioorg Chem. 2021;109:104729.
  • Lo KKW, Lee TKM, Lau JSY, Poon WL, Cheng SH. Luminescent biological probes derived from ruthenium (II) estradiol polypyridine complexes. Inorg Chem. 2008;47(1):200–208.
  • Böhm I, Schild H. Apoptosis: The complex scenario for a silent cell death. Mol Imaging Biol. 2003;5(1):2–14.
  • Su M, Li Y, Chung HY, Ye W. 2β-(isobutyryloxy) florilenalin, a sesquiterpene lactone isolated from the medicinal plant centipeda minima, induces apoptosis in human nasopharyngeal carcinoma cne cells. Molecules. 2009;14(6):2135–2146.
  • Corripio-Miyar Y, MacLeod CL, Mair I, Mellanby RJ, Moore BD, McNeilly TN. Self-adjuvanting calcium-phosphate-coated microcrystal-based vaccines induce pyroptosis in human and livestock immune cells. Vaccines (Basel). 2023;11(7):1229.
  • Saad MH, El-Moselhy TF, S E-D N, Mehany AB, Belal A, Abourehab MA, Tawfik HO, El-Hamamsy MH. Discovery of new symmetrical and asymmetrical nitrile-containing 1, 4-dihydropyridine derivatives as dual kinases and p-glycoprotein inhibitors: synthesis, in vitro assays, and in silico studies. J Enzyme Inhib Med Chem. 2022;37(1):2489–2511.
  • Hassan A, Mubarak FA, Shehadi IA, Mosallam AM, Temairk H, Badr M, Abdelmonsef AH. Design and biological evaluation of 3-substituted quinazoline-2, 4 (1 h, 3 h)-dione derivatives as dual c-met/vegfr-2-tk inhibitors. J Enzyme Inhib Med Chem. 2023;38(1):2189578.
  • Elkady H, El-Dardir OA, Elwan A, Taghour MS, Mahdy HA, Dahab MA, Elkaeed EB, Alsfouk BA, Ibrahim IM, Husein DZ, et al. Synthesis, biological evaluation and computer-aided discovery of new thiazolidine-2, 4-dione derivatives as potential antitumor vegfr-2 inhibitors. RSC Adv. 2023;13(40):27801–27827.
  • Abdallah AE, Mabrouk RR, Al Ward MMS, Eissa SI, Elkaeed EB, Mehany AB, Abo-Saif MA, El-Feky OA, Alesawy MS, El-Zahabi M. Synthesis, biological evaluation, and molecular docking of new series of antitumor and apoptosis inducers designed as vegfr-2 inhibitors. J Enzyme Inhib Med Chem. 2022;37(1):573–591.
  • Abdallah AE, Mabrouk RR, Elnagar MR, Farrag AM, Kalaba MH, Sharaf MH, El-Fakharany EM, Bakhotmah DA, Elkaeed EB, Al Ward M. New series of vegfr-2 inhibitors and apoptosis enhancers: design, synthesis and biological evaluation. Drug Des Devel Ther. 2022; 16:587–606.
  • Livak KJ, Schmittgen T. Analysis of relative gene expression data using real-time quantitative pcr and the 2(-Delta Delta C(T)). Method. Methods. 2001;25(4):402–408.
  • Zhou Y, Li L, Mao C, Zhou D. Astragaloside iv ameliorates spinal cord injury through controlling ferroptosis in H2O2-damaged pc12 cells in vitro. Ann Transl Med. 2022;10 (21):1176–1176.
  • Fu J, Zhao J, Zhang H, Fan X, Geng W, Qiao S. Microrna‑451a prevents cutaneous squamous cell carcinoma progression via the 3‑phosphoinositide‑dependent protein kinase‑1‑mediated pi3k/akt signaling pathway. Exp Ther Med. 2021;21(2):1–1.
  • Vilar S, Cozza G, Moro S. Medicinal chemistry and the molecular operating environment (moe): application of qsar and molecular docking to drug discovery. Curr Top Med Chem. 2008;8(18):1555–1572.
  • Scholz C, Knorr S, Hamacher K, Schmidt B. Docktite- a highly versatile step-by-step workflow for covalent docking and virtual screening in the molecular operating environment. J Chem Inf Model. 2015;55(2):398–406.
  • El Hassab MA, Ibrahim TM, Al-Rashood ST, Alharbi A, Eskandrani RO, Eldehna W. In silico identification of novel sars-cov-2 2′-o-methyltransferase (nsp16) inhibitors: structure-based virtual screening, molecular dynamics simulation and mm-pbsa approaches. J Enzyme Inhib Med Chem. 2021;36(1):727–736.
  • Hassab MA, Fares M, Amin MK, Al-Rashood ST, Alharbi A, Eskandrani RO, Alkahtani HM, Eldehna W. Toward the identification of potential α-ketoamide covalent inhibitors for sars-cov-2 main protease: fragment-based drug design and mm-pbsa calculations. Processes. 2021;9(6):1004.
  • Schüttelkopf AW, Van Aalten D. Prodrg: a tool for high-throughput crystallography of protein–ligand complexes. Acta Crystallogr D Biol Crystallogr. 2004;60(Pt 8):1355–1363.
  • El Hassab MA, Ibrahim TM, Shoun AA, Al-Rashood ST, Alkahtani HM, Alharbi A, Eskandrani RO, Eldehna W. In silico identification of potential sars cov-2 2′-o-methyltransferase inhibitor: fragment-based screening approach and mm-pbsa calculations. RSC Adv. 2021;11(26):16026–16033.