389
Views
2
CrossRef citations to date
0
Altmetric
Research Articles

New lead compounds identification against KRas mediated cancers through pharmacophore-based virtual screening and in vitro assays

, , ORCID Icon, , , & show all
Pages 8053-8067 | Received 26 Jul 2022, Accepted 20 Sep 2022, Published online: 02 Oct 2022

References

  • Adams, J. (2004). The development of proteasome inhibitors as anticancer drugs. Cancer Cell, 5(5), 417–421.
  • Adams, J., & Kauffman, M. (2004). Development of the proteasome inhibitor Velcade™(Bortezomib). Cancer Investigation, 22(2), 304–311. https://doi.org/10.1081/CNV-120030218
  • Akbar, S., Subhan, F., Shahid, M., Wadood, A., Shahbaz, N., Farooq, U., Ayaz, M., & Raziq, N. (2020). 6-Methoxyflavanone abates cisplatin-induced neuropathic pain apropos anti-inflammatory mechanisms: A behavioral and molecular simulation study. European Journal of Pharmacology, 872, 172972.
  • Ayaz, M., Junaid, M., Ullah, F., Sadiq, A., Subhan, F., Khan, M. A., Ahmad, W., Ali, G., Imran, M., & Ahmad, S. (2016). Molecularly characterized solvent extracts and saponins from Polygonum hydropiper L. show high anti-angiogenic, anti-tumor, brine shrimp, and fibroblast NIH/3T3 cell line cytotoxicity. Frontiers in Pharmacology, 7, 74. https://doi.org/10.3389/fphar.2016.00074
  • Ayaz, M., Nawaz, A., Ahmad, S., Mosa, O. F., Eisa Hamdoon, A. A., Khalifa, M. A., Sadiq, A., Ullah, F., Wadood, A., & Kabra, A. (2022). Underlying anticancer mechanisms and synergistic combinations of phytochemicals with cancer chemotherapeutics: Potential benefits and risks. Journal of Food Quality, 2022, 1-15.
  • Ayaz, M., Sadiq, A., Mosa, O. F., Zafar, T. A., Eisa Hamdoon, A. A., Elkhalifa, M. E. M., Elawad, M. A., Ahmed, A., Ullah, F., Ghufran, M., & Kabra, A. (2022). Antioxidant, enzyme inhibitory, and molecular docking approaches to the antidiabetic potentials of bioactive compounds from Persicaria hydropiper L. Evidence-Based Complementary and Alternative Medicine, 2022, 1–13.
  • Ayaz, M., Wadood, A., Sadiq, A., Ullah, F., Anichkina, O., & Ghufran, M. (2021). In-silico evaluations of the isolated phytosterols from polygonum hydropiper L against BACE1 and MAO drug targets. Journal of Biomolecular Structure and Dynamics, 1–9. https://doi.org/10.1080/07391102.2021.1940286
  • Baravalia, Y., Vaghasiya, Y., & Chanda, S. (2012). Brine shrimp cytotoxicity, anti-inflammatory and analgesic properties of Woodfordia fruticosa Kurz flowers. Iranian Journal of Pharmaceutical Research: IJPR, 11, 851.
  • Boccadoro, M., Morgan, G., & Cavenagh, J. (2005). Preclinical evaluation of the proteasome inhibitor bortezomib in cancer therapy. Cancer Cell International, 5(1), 1–9. https://doi.org/10.1186/1475-2867-5-18
  • Bourne, H. R., Sanders, D. A., & Mccormick, F. (1990). The GTPase superfamily: A conserved switch for diverse cell functions. Nature, 348(6297), 125–132. https://doi.org/10.1038/348125a0
  • Chen, D., Frezza, M., Schmitt, S., Kanwar, J.P., & Dou, Q. (2011). Bortezomib as the first proteasome inhibitor anticancer drug: Current status and future perspectives. Current Cancer Drug Targets, 11(3), 239–253.
  • Darden, T., York, D., & Pedersen, L. (1993). Particle mesh Ewald: An N⋅ log (N) method for Ewald sums in large systems. The Journal of Chemical Physics, 98(12), 10089–10092. https://doi.org/10.1063/1.464397
  • Drilon, A., Schoenfeld, A. J., Arbour, K. C., Litvak, A., Ni, A., Montecalvo, J., Yu, H. A., Panora, E., Ahn, L., Kennedy, M., Haughney-Siller, A., Miller, V., Ginsberg, M., Ladanyi, M., Arcila, M., Rekhtman, N., Kris, M. G., & Riely, G. J. (2019). Exceptional responders with invasive mucinous adenocarcinomas: A phase 2 trial of bortezomib in patients with KRas G12D-mutant lung cancers. Molecular Case Studies, 5(2), a003665. https://doi.org/10.1101/mcs.a003665
  • Essmann, U., Perera, L., Berkowitz, M. L., Darden, T., Lee, H., & Pedersen, L. G. (1995). A smooth particle mesh Ewald method. The Journal of Chemical Physics, 103(19), 8577–8593. https://doi.org/10.1063/1.470117
  • Ghufran, M., Rehman, A. U., Shah, M., Ayaz, M., Ng, H. L., & Wadood, A. (2020). In-silico design of peptide inhibitors of K-Ras target in cancer disease. Journal of Biomolecular Structure and Dynamics, 38(18), 5488–5499. https://doi.org/10.1080/07391102.2019.1704880
  • Giampieri, R., Lupi, A., Ziranu, P., Bittoni, A., Pretta, A., Pecci, F., Persano, M., Giglio, E., Copparoni, C., Crocetti, S., Mandolesi, A., Faa, G., Coni, P., Scartozzi, M., & Berardi, R. (2021). Retrospective comparative analysis of KRas G12C vs. other KRas mutations in mCRC patients treated with first-line chemotherapy doublet + bevacizumab. Frontiers in Oncology, 11, 1-10. https://doi.org/10.3389/fonc.2021.736104
  • Grant, B. J., Rodrigues, A. P., Elsawy, K. M., Mccammon, J. A., & Caves, L. S. (2006). Bio3d: An R package for the comparative analysis of protein structures. Bioinformatics, 22(21), 2695–2696. https://doi.org/10.1093/bioinformatics/btl461
  • Güner, A., Polatli, E., Akkan, T., Bektaş, H., & Albay, C. (2019). Anticancer and antiangiogenesis activities of novel synthesized 2-substituted benzimidazoles molecules. Turkish Journal of Chemistry, 43(5), 1270–1289. https://doi.org/10.3906/kim-1904-46
  • Hann, M. M., & Oprea, T. I. (2004). Pursuing the leadlikeness concept in pharmaceutical research. Current Opinion in Chemical Biology, 8(3), 255–263. https://doi.org/10.1016/j.cbpa.2004.04.003
  • Harwig, J., & Scott, P. (1971). Brine shrimp (Artemia salina L.) larvae as a screening system for fungal toxins. Applied Microbiology, 21(6), 1011–1016. https://doi.org/10.1128/am.21.6.1011-1016.1971
  • Hidalgo, M. (2010). Pancreatic cancer. The New England Journal of Medicine, 362(17), 1605–1617.
  • Hung, C. L., & Chen, C. C. (2014). Computational approaches for drug discovery. Drug Development Research, 75(6), 412–418.
  • Hussain, S., Ullah, F., Ayaz, M., Ali Shah, S. A., Ali Shah, A.-U.-H., Shah, S. M., Wadood, A., Aman, W., Ullah, R., Shahat, A. A., & Nasr, F. A. (2019). In silico, cytotoxic and antioxidant potential of novel ester, 3-hydroxyoctyl-5-trans-docosenoate isolated from anchusa arvensis (L.) m. bieb. against hepg-2 cancer cells. Drug Design, Development and Therapy, 13, 4195–4205.
  • Iftikhar, F., Yaqoob, F., Tabassum, N., Jan, M. S., Sadiq, A., Tahir, S., Batool, T., Niaz, B., Ansari, F. L., Choudhary, M. I., & Rashid, U. (2018). Design, synthesis, in-vitro thymidine phosphorylase inhibition, in-vivo antiangiogenic and in-silico studies of C-6 substituted dihydropyrimidines. Bioorganic Chemistry, 80, 99–111.
  • Irwin, J. J., Sterling, T., Mysinger, M. M., Bolstad, E. S., & Coleman, R. G. (2012). ZINC: A free tool to discover chemistry for biology. Journal of Chemical Information and Modeling, 52(7), 1757–1768.
  • Karnoub, A. E., & Weinberg, R. A. (2008). Ras oncogenes: Split personalities. Nature Reviews. Molecular Cell Biology, 9(7), 517–531.
  • Khalil, A. T., Ayaz, M., Ovais, M., Wadood, A., Ali, M., Shinwari, Z. K., & Maaza, M. (2018). In vitro cholinesterase enzymes inhibitory potential and in silico molecular docking studies of biogenic metal oxides nanoparticles. Inorganic and Nano-Metal Chemistry, 48(9), 441–448. https://doi.org/10.1080/24701556.2019.1569686
  • Khalil, A. T., Ovais, M., Iqbal, J., Ali, A., Ayaz, M., Abbas, M., Ahmad, I., & Devkota, H. P. (2021). Microbes-mediated synthesis strategies of metal nanoparticles and their potential role in cancer therapeutics. Seminars in Cancer Biology. https://doi.org/10.1016/j.semcancer.2021.06.006
  • Kitchen, D. B., Decornez, H., Furr, J. R., & Bajorath, J. (2004). Docking and scoring in virtual screening for drug discovery: Methods and applications. Nature Reviews Drug Discovery, 3(11), 935–949. https://doi.org/10.1038/nrd1549
  • Leach, A. R., Shoichet, B. K., & Peishoff, C. E. (2006). Prediction of protein − ligand interactions. Docking and scoring: Successes and gaps. Journal of Medicinal Chemistry, 49(20), 5851–5855.
  • Lim, S. M., Westover, K. D., Ficarro, S. B., Harrison, R. A., Choi, H. G., Pacold, M. E., Carrasco, M., Hunter, J., Kim, N. D., Xie, T., Sim, T., Jänne, P. A., Meyerson, M., Marto, J. A., Engen, J. R., & Gray, N. S. (2014). Therapeutic targeting of oncogenic K‐Ras by a covalent catalytic site inhibitor. Angewandte Chemie, 126(1), 203–208. https://doi.org/10.1002/ange.201307387
  • Lipinski, C. A. (2000). Drug-like properties and the causes of poor solubility and poor permeability. Journal of Pharmacological and Toxicological Methods, 44(1), 235–249.
  • Macalino, S. J. Y., Gosu, V., Hong, S., & Choi, S. (2015). Role of computer-aided drug design in modern drug discovery. Archives of Pharmacal Research, 38(9), 1686–1701.
  • Mahnashi, M. H., Alqahtani, Y. S., Alyami, B. A., Alqarni, A. O., Ayaz, M., Ghufran, M., Ullah, F., Sadiq, A., Ullah, I., Haq, I. U., Khalid, M., & Murthy, H. C. A. (2022). Phytochemical analysis, α-glucosidase and amylase inhibitory, and molecular docking studies on Persicaria hydropiper L. leaves essential oils. Evidence-Based Complementary and Alternative Medicine, 2022, 1–11. https://doi.org/10.1155/2022/7924171
  • Mahnashi, M. H., Alqahtani, Y. S., Alyami, B. A., Alqarni, A. O., Ullah, F., Wadood, A., Sadiq, A., Shareef, A., & Ayaz, M. (2021). Cytotoxicity, anti-angiogenic, anti-tumor and molecular docking studies on phytochemicals isolated from Polygonum hydropiper L. BMC Complementary Medicine and Therapies, 21(1), 1–14. https://doi.org/10.1186/s12906-021-03411-1
  • Mao, Z., Xiao, H., Shen, P., Yang, Y., Xue, J., Yang, Y., Shang, Y., Zhang, L., Li, X., Zhang, Y., Du, Y., Chen, C.-C., Guo, R.-T., & Zhang, Y. (2022). KRas (G12D) can be targeted by potent inhibitors via formation of salt bridge. Cell Discovery, 8(1), 1–14. https://doi.org/10.1038/s41421-021-00368-w
  • Meyer, B., Ferrigni, N., Putnam, J., Jacobsen, L., Nichols, D., & Mclaughlin, J. L. (1982). Brine shrimp: A convenient general bioassay for active plant constituents. Planta Medica, 45(5), 31–34.
  • Michael, A., Thompson, C., & Abramovitz, M. (1956). Artemia salina as a test organism for bioassay. Science (New York, N.Y.), 123(3194), 464–464.
  • Morris, G. M., & Lim-Wilby, M. (2008). Molecular docking. In Molecular modeling of proteins (pp. 353-382). Springer. https://doi.org/10.1007/978-1-59745-177-2
  • Muegge, I. (2003). Selection criteria for drug‐like compounds. Medicinal Research Reviews, 23(3), 302–321. https://doi.org/10.1002/med.10041
  • Mutebi, M., Dehar, N., Nogueira, L. M., Shi, K., Yabroff, K. R., & Gyawali, B. (2022). Cancer groundshot: Building a robust cancer control platform in addition to launching the cancer moonshot. American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Annual Meeting, 42, 1–16. https://doi.org/10.1200/EDBK_359521
  • Nasar, M. Q., Shah, M., Khalil, A. T., Kakar, M. Q., Ayaz, M., Dablool, A. S., & Shinwari, Z. K. (2022). Ephedra intermedia mediated synthesis of biogenic silver nanoparticles and their antimicrobial, cytotoxic and hemocompatability evaluations. Inorganic Chemistry Communications, 137, 109252. https://doi.org/10.1016/j.inoche.2022.109252
  • Neves, M. A., Dinis, T. C., Colombo, G., & Sa e Melo, M. L. (2009). Fast three dimensional pharmacophore virtual screening of new potent non-steroid aromatase inhibitors. Journal of Medicinal Chemistry, 52(1), 143–150.
  • Nirmala, M. J., Samundeeswari, A., & Sankar, P. D. (2011). Natural plant resources in anti-cancer therapy-A review. Research in Plant Biology, 1, 01–14.
  • Nobili, S., Lippi, D., Witort, E., Donnini, M., Bausi, L., Mini, E., & Capaccioli, S. (2009). Natural compounds for cancer treatment and prevention. Pharmacological Research, 59(6), 365–378.
  • Nowis, D., Mcconnell, E. J., Dierlam, L., Palamarchuk, A., Lass, A., & Wójcik, C. (2007). TNF potentiates anticancer activity of bortezomib (Velcade®) through reduced expression of proteasome subunits and dysregulation of unfolded protein response. International Journal of Cancer, 121(2), 431–441. https://doi.org/10.1002/ijc.22695
  • Ostrem, J. M., Peters, U., Sos, M. L., Wells, J. A., & Shokat, K. M. (2013). K-Ras (G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature, 503(7477), 548–551.
  • Ovais, M., Hoque, M. Z., Khalil, A. T., Ayaz, M., & Ahmad, I. (2021). Mechanisms underlying the anticancer applications of biosynthesized nanoparticles. In Biogenic nanoparticles for cancer theranostics (pp. 229-241). Elsevier.
  • Ovais, M., Khalil, A. T., Ayaz, M., & Ahmad, I. (2019). Biosynthesized metallic nanoparticles as emerging cancer theranostics agents. In Nanotheranostics (pp. 229-244). Springer. https://doi.org/10.1007/978-3-030-29768-8_11
  • Ovais, M., Khalil, A. T., Ayaz, M., & Ahmad, I. (2020). Metal oxide nanoparticles and plants. In Phytonanotechnology: Challenges and prospects (Vol. 123, pp. 123-141). Elsevier. https://doi.org/10.1016/B978-0-12-822348-2.00007-3
  • Patra, C., Ahmad, I., Ayaz, M., Khalil, A. T., Mukherjee, S., & Ovais, M. (2021). Biogenic nanoparticles for cancer theranostics. Elsevier.
  • Piao, L., Chen, Z., Li, Q., Liu, R., Song, W., Kong, R., & Chang, S. (2019). Molecular dynamics simulations of wild type and mutants of SAPAP in complexed with Shank3. International Journal of Molecular Sciences, 20(1), 224. https://doi.org/10.3390/ijms20010224
  • Rajabi, M., & Mousa, S. A. (2017). The role of angiogenesis in cancer treatment. Biomedicines, 5(4), 34. https://doi.org/10.3390/biomedicines5020034
  • Raphael, B. J., Hruban, R. H., Aguirre, A. J., Moffitt, R. A., Yeh, J. J., Stewart, C., Robertson, A. G., Cherniack, A. D., Gupta, M., Getz, G., Gabriel, S. B., Meyerson, M., Cibulskis, C., Fei, S. S., Hinoue, T., Shen, H., Laird, P. W., Ling, S., Lu, Y., … Zenklusen, J. C. (2017). Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell, 32(2), 185–203. https://doi.org/10.1016/j.ccell.2017.07.007
  • Ryckaert, J.-P., Ciccotti, G., & Berendsen, H. J. (1977). Numerical integration of the cartesian equations of motion of a system with constraints: Molecular dynamics of n-alkanes. Journal of Computational Physics, 23(3), 327–341. https://doi.org/10.1016/0021-9991(77)90098-5
  • Salomon-Ferrer, R., Case, D. A., & Walker, R. C. (2013). An overview of the Amber biomolecular simulation package. Wiley Interdisciplinary Reviews: Computational Molecular Science, 3, 198–210.
  • Salomon-Ferrer, R., Gotz, A. W., Poole, D., Le Grand, S., & Walker, R. C. (2013). Routine microsecond molecular dynamics simulations with AMBER on GPUs. 2. Explicit solvent particle mesh Ewald. Journal of Chemical Theory and Computation, 9(9), 3878–3888.
  • Scannell, J. W., Blanckley, A., Boldon, H., & Warrington, B. (2012). Diagnosing the decline in pharmaceutical R&D efficiency. Nature Reviews Drug Discovery, 11(3), 191–200. https://doi.org/10.1038/nrd3681
  • Seifert, E. (2014). OriginPro 9.1: Scientific data analysis and graphing software-software review. Journal of Chemical Information and Modeling, 54(5), 1552.
  • Sethi, A., Joshi, K., Sasikala, K., & Alvala, M. (2019). Molecular docking in modern drug discovery: Principles and recent applications. Drug Discovery and Development-New Advances, 2, 1–21.
  • Singh, H., Longo, D. L., & Chabner, B. A. (2015). Improving prospects for targeting RAS. Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, 33(31), 3650–3659.
  • Stephen, A. G., Esposito, D., Bagni, R. K., & Mccormick, F. (2014). Dragging ras back in the ring. Cancer Cell, 25(3), 272–281. https://doi.org/10.1016/j.ccr.2014.02.017
  • Stoermer, M. J. (2006). Current status of virtual screening as analysed by target class. Medicinal Chemistry (Shariqah (United Arab Emirates)), 2(1), 89–112.
  • Surabhi, S., & Singh, B. (2018). Computer aided drug design: An overview. Journal of Drug Delivery and Therapeutics, 8(5), 504–509. https://doi.org/10.22270/jddt.v8i5.1894
  • Tong, X., Li, X., Ayaz, M., Ullah, F., Sadiq, A., Ovais, M., Shahid, M., Khayrullin, M., & Hazrat, A. (2020). Neuroprotective studies on Polygonum hydropiper L. essential oils using transgenic animal models. Frontiers in Pharmacology, 11, 580069.
  • Trahey, M., & Mccormick, F. (1987). A cytoplasmic protein stimulates normal N-ras p21 GTPase, but does not affect oncogenic mutants. Science, 238(4826), 542–545. https://doi.org/10.1126/science.2821624
  • Van Walbeek, W., Moodie, C. A., Scott, P. M., Harwig, J., & Grice, H. C. (1971). Toxicity and excretion of ochratoxin A in rats intubated with pure ochratoxin A or fed cultures of Penicillium viridicatum. Toxicology and Applied Pharmacology, 20, 439–441.
  • Vanhaecke, P., Persoone, G., Claus, C., & Sorgeloos, P. (1981). Proposal for a short-term toxicity test with Artemia nauplii. Ecotoxicology and Environmental Safety, 5(3), 382–387.
  • Wang, T., Yin, Z., Zhang, Z., Bender, J. A., Yang, Z., Johnson, G., Yang, Z., Zadjura, L. M., D'Arienzo, C. J., DiGiugno Parker, D., Gesenberg, C., Yamanaka, G. A., Gong, Y.-F., Ho, H.-T., Fang, H., Zhou, N., McAuliffe, B. V., Eggers, B. J., Fan, L., … Kadow, J. F. (2009). Inhibitors of human immunodeficiency virus type 1 (HIV-1) attachment. 5. An evolution from indole to azaindoles leading to the discovery of 1-(4-benzoylpiperazin-1-yl)-2-(4, 7-dimethoxy-1 H-pyrrolo [2, 3-c] pyridin-3-yl) ethane-1, 2-dione (BMS-488043), a drug candidate that demonstrates antiviral activity in HIV-1-infected subjects. Journal of Medicinal Chemistry, 52(23), 7778–7787. https://doi.org/10.1021/jm900843g
  • Wei-Ya, L., Yu-Qing, D., Yang-Chun, M., Xin-Hua, L., Ying, M., & Wang, R.-L. (2019). Exploring the cause of the inhibitor 4AX attaching to binding site disrupting protein tyrosine phosphatase 4A1 trimerization by molecular dynamic simulation. Journal of Biomolecular Structure & Dynamics, 37(18), 4840–4851.
  • Wittinghofer, A., & Pal, E. F. (1991). The structure of Ras protein: A model for a universal molecular switch. Trends in Biochemical Sciences, 16, 382–387. https://doi.org/10.1016/0968-0004(91)90156-P
  • Wolfman, A., & Macara, I. G. (1990). A cytosolic protein catalyzes the release of GDP from p21 ras. Science (New York, N.Y.), 248(4951), 67–69.
  • Yin, D., Zhou, H., Kumagai, T., Liu, G., Ong, J. M., Black, K. L., & Koeffler, H. P. (2005). Proteasome inhibitor PS-341 causes cell growth arrest and apoptosis in human glioblastoma multiforme (GBM). Oncogene, 24(3), 344–354.
  • Zwanzig, R. (1973). Nonlinear generalized Langevin equations. Journal of Statistical Physics, 9(3), 215–220. https://doi.org/10.1007/BF01008729

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.