Generic placeholder image

Letters in Drug Design & Discovery

Editor-in-Chief

ISSN (Print): 1570-1808
ISSN (Online): 1875-628X

Research Article

Design and Synthesis of Tri-substituted Imidazole Derivatives as CD73 Inhibitors for Their Anticancer Activity

Author(s): Abhishek Ghara*, Ganesh Sakharam Andhale, Gurubasavaraja Swamy Purawarga Matada and Prasad Sanjay Dhiwar

Volume 19, Issue 3, 2022

Published on: 04 June, 2021

Page: [242 - 255] Pages: 14

DOI: 10.2174/1570180818666210604113849

Price: $65

Abstract

Background: Monoclonal antibodies licensed by the US Food and Drug Administration (FDA) target diverse biological targets relevant to immuno-oncology, and small compounds in clinical trials target various aspects of immuno-oncology. Several small compounds that target CD73 are at various stages of clinical studies. Several imidazoles are currently being utilized to treat malignancies, including Dacarbazine, Zoledronic acid, Mercaptopurine, and others. As a result, we evaluated the cytotoxicity of modified tri-phenyl imidazoles against breast cancer cell lines, as well as conducted virtual tests.

Methods: We used Accelrys Drug Discovery Studio 3.5 software to undertake molecular docking, ADMET, and molecular properties studies on 68 proposed imidazole derivatives. The synthesized compounds' binding mechanisms were investigated against the CD73 protein (PDB Code: 4H1S). To find the drugs with the best pharmacokinetics, researchers assessed ADMET solubility, BBB penetration, hepatotoxicity, PPB binding, and polar surface area. The MDA-MB-231 breast cancer cell line was treated with these produced compounds, and the MTT test method was used to determine the IC50 values.

Results: The selected 14 compounds showed good binding in the active site of CD73 by forming Hbonds with amino acid residues, according to molecular docking studies. Breast cancer cell lines were treated with substituted tri-phenyl imidazole derivatives, which displayed anticancer activity. Compounds 3a and 3h, which had an electron-donating group at the 2nd and 3rd positions and p-substitutions of the chloro and nitro groups, respectively, showed considerable anticancer action.

Conclusion: Fourteen imidazole derivatives were produced and tested against breast cancer cell lines based on in-silico research. The MDA-MB-231 cell line was strongly suppressed by compounds 3a and 3h. In-vitro enzyme inhibition experiments revealed that only 3h demonstrated considerable inhibition.

Keywords: Imidazole derivatives, in-silico studies, anticancer activity, MDA-MB-231, MTT assay, CD73 inhibition assay.

Graphical Abstract
[1]
Duran, M.; Demirayak, S. Synthesis of 2-[4,5-dimethyl-1-(phenylamino)-1H-imidazol-2-yl thio]-n-(thiazole-2-yl)acetamide derivatives and their anticancer activities. Med. Chem. Res., 2013, 22, 4110-4124.
[http://dx.doi.org/10.1007/s00044-012-0411-5]
[2]
OncoStem. Alarming facts about breast cancer in India., Available from: https://www.oncostem.com/blog/alarming-facts-about-breast-cancer-in-india(Accessed July 30, 2020)
[3]
Smith, R.A.; Andrews, K.S.; Brooks, D.; Fedewa, S.A.; Manassaram-Baptiste, D.; Saslow, D.; Brawley, O.W.; Wender, R.C. Cancer screening in the United States, 2018: A review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J. Clin., 2018, 68(4), 297-316.
[http://dx.doi.org/10.3322/caac.21446] [PMID: 29846940]
[4]
Smith, R.A.; Andrews, K.S.; Brooks, D.; Fedewa, S.A.; Manassaram-Baptiste, D.; Saslow, D.; Wender, R.C. Cancer screening in the United States, 2019: A review of current American Cancer Society guidelines and current issues in cancer screening. CA Cancer J. Clin., 2019, 69(3), 184-210.
[http://dx.doi.org/10.3322/caac.21557] [PMID: 30875085]
[5]
Huck, B.R.; Kötzner, L.; Urbahns, K. Small molecules drive big improvements in immune-oncology therapies. Angew. Chem. Int. Ed. Engl., 2018, 57(16), 4412-4428.
[http://dx.doi.org/10.1002/anie.201707816] [PMID: 28971564]
[6]
van der Zanden, S.Y.; Luimstra, J.J.; Neefjes, J.; Borst, J.; Ovaa, H. Opportunities for small molecules in cancer immunotherapy. Trends Immunol., 2020, 41(6), 493-511.
[http://dx.doi.org/10.1016/j.it.2020.04.004] [PMID: 32381382]
[7]
Antonioli, L.; Yegutkin, G.G.; Pacher, P.; Blandizzi, C.; Hasko, G. Anti-CD73 in cancer immunotherapy: Awakening new opportunities. Trends Cancer, 2016, 2(2), 95-109.
[http://dx.doi.org/10.1016/j.trecan.2016.01.003]
[8]
Ohta, A.; Gorelik, E.; Prasad, S.J.; Ronchese, F.; Lukashev, D.; Wong, M.K.; Huang, X.; Caldwell, S.; Liu, K.; Smith, P.; Chen, J.F.; Jackson, E.K.; Apasov, S.; Abrams, S.; Sitkovsky, M. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl. Acad. Sci. USA, 2006, 103(35), 13132-13137.
[http://dx.doi.org/10.1073/pnas.0605251103] [PMID: 16916931]
[9]
Hoskin, D.W.; Mader, J.S.; Furlong, S.J.; Conrad, D.M.; Blay, J. Inhibition of T cell and natural killer cell function by adenosine and its contribution to immune evasion by tumor cells. (Review) Int. J. Oncol., 2008, 32(3), 527-535.
[http://dx.doi.org/10.3892/ijo.32.3.527] [PMID: 18292929]
[10]
Gao, Z.W.; Dong, K.; Zhang, H.Z. The roles of CD73 in cancer. BioMed Res. Int., 2014, 2014460654
[http://dx.doi.org/10.1155/2014/460654] [PMID: 25126561]
[11]
Gong, Y.P.; Wan, R.Z.; Liu, Z.P. Evaluation of WO2017098421: GSK’s benzothiazine compounds as CD73 inhibitor filings. Expert Opin. Ther. Pat., 2018, 28(2), 167-171.
[http://dx.doi.org/10.1080/13543776.2018.1407756] [PMID: 29166791]
[12]
Gaba, M.; Mohan, C. Development of drugs based on imidazle and benzimidazole bioactive heterocycles: recent advances and future directions. Med. Chem. Res., 2016, 25, 173-210.
[http://dx.doi.org/10.1007/s00044-015-1495-5]
[13]
Sharma, G.; Adepu, R.; Singh, A.; Gourishetty, S.; Vankudoth, J.; Duscharla, D. Imidazole derivatives show anticancer potential by inducing apoptosis and cellular senescence. MedChemComm, 2014, 5, 1751-1760.
[http://dx.doi.org/10.1039/C4MD00277F]
[14]
Verma, A.; Joshi, S.; Singh, D. Imidazole: Having Versatile Biological Activities. J. Chem., 2013, 5, 1-12.
[15]
Sharma, D.; Ashaduzzaman, M.; Golabi, M.; Shriwastav, A.; Bisetty, K.; Tiwari, A. Studies on bacterial proteins corona interaction with saponin imprinted ZnO nano-honeycombs and their toxic responses. ACS Appl. Mater. Interfaces, 2015, 7(43), 23848-23856.
[http://dx.doi.org/10.1021/acsami.5b06617] [PMID: 26439810]
[16]
Zhang, J.; Shan, Y.; Pan, X.; Wang, C.; Xu, W.; He, L. Molecular docking, 3D-QSAR studies, and in silico ADME prediction of p-aminosalicylic acid derivatives as neuraminidase inhibitors. Chem. Biol. Drug Des., 2011, 78(4), 709-717.
[http://dx.doi.org/10.1111/j.1747-0285.2011.01179.x] [PMID: 21752201]
[17]
Usha, T.; Middha, S.K.; Goyal, A.K.; Karthik, M.; Manoj, D.; Faizan, S.; Goyal, P.; Prashanth, H.; Pande, V. Molecular docking studies of anti-cancerous candidates in Hippophae rhamnoides and Hippophae salicifolia. J. Biomed. Res., 2014, 28(5), 406-415.
[PMID: 25332713]
[18]
Chedik, L.; Mias-Lucquin, D.; Bruyere, A.; Fardel, O. In silico prediction for intestinal absorption and brain penetration of chemical pestisides in humans. Int. J. Environ. Res., 2017, 14, 708-724.
[http://dx.doi.org/10.3390/ijerph14070708]
[19]
Richard, L.; Ranjani, V.; Manigandan, K.; Elangovan, N. In silico docking studies to identify potent inhibitors of alpha-synuclein aggregation in parkinson disease. Asian. J. Pharm. Clin. Res., 2013, 6(4), 127-131. Available at: https://innovareacademics.in/journals/index.php/ajpcr/article/view/369
[20]
Ponnan, P.; Gupta, S.; Chopra, M.; Tandon, R.; Baghel, A.; Gupta, G. 2D-QSAR, docking studies and in-silico ADMET prediction of polyphenolic acetates as substrates for protein acetyltransferase function of glutamine synthetase of Mycobacterium tuberculosis. Struc. Bio., 2013, 37, 1-12.
[http://dx.doi.org/10.1155/2013/373516]
[21]
Abdel-Hamid, M.K.; McCluskey, A.; McCluskey, A. In silico docking, molecular dynamics and binding energy insights into the bolinaquinone-clathrin terminal domain binding site. Molecules, 2014, 19(5), 6609-6622.
[http://dx.doi.org/10.3390/molecules19056609] [PMID: 24858095]
[22]
Husain, A.; Drabu, S.; Kumar, N.; Alam, M.M.; Bawa, S. Synthesis and biological evaluation of di- and tri-substituted imidazoles as safer anti-inflammatory-antifungal agents. J. Pharm. Bioallied Sci., 2013, 5(2), 154-161.
[http://dx.doi.org/10.4103/0975-7406.111822] [PMID: 23833522]
[23]
Furnis, B.S.; Hannaford, A.; Smith, P.W.G. Tatchell. Vogel text book of practical organic chemistry, 5th ed; Saurabh Printers PVT Ltd: India, 2004. Available at: https://faculty.ksu.edu.sa/sites/default/files/vogel_-_practical_organic_chemistry_5th_edition.pdf
[24]
Narasimhamurthy, K.H. Novel approach for the synthesis of nitrogen containing heterocycles and their biological studies. Ph.D Thesis, University of Mysore, 2015. Available at: https://www.worldcat.org/title/novel-approaches-to-synthesis-of-nitrogen-containing-heterocycles/oclc/225659730?referer=di&ht=edition
[26]
Antonioli, L.; Yegutkin, G.G.; Pacher, P.; Blandizzi, C.; Haskó, G. Anti-CD73 in cancer immunotherapy: awakening new opportunities. Trends Cancer, 2016, 2(2), 95-109.
[http://dx.doi.org/10.1016/j.trecan.2016.01.003] [PMID: 27014745]
[27]
Asma, S.A.W.; Ashraf, S.H.; Ahmed, M.N. in vitro cytotoxicity and drug likeness of pyrazolines and pyridines bearing benzofuran moiety. J. Appl. Pharm. Sci., 2020, 10(04), 142-148.
[http://dx.doi.org/10.7324/JAPS.2020.104018]
[28]
Vlad, I.M.; Nuta, D.C.; Chirita, C.; Caproiu, M.T.; Draghici, C.; Dumitrascu, F.; Bleotu, C.; Avram, S.; Udrea, A.M.; Missir, A.V.; Marutescu, L.G.; Limban, C. In silico and in vitro experimental studies of new dibenz[b,e]oxepin-11(6H)one O-(arylcarbamoyl)-oximes designed as potential antimicrobial agents. Molecules, 2020, 25(2), 1-18.
[http://dx.doi.org/10.3390/molecules25020321] [PMID: 31941125]
[29]
Lipinski, C.A. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discov. Today. Technol., 2004, 1(4), 337-341.
[http://dx.doi.org/10.1016/j.ddtec.2004.11.007] [PMID: 24981612]
[30]
Anfel, Z.; Salah, B.; Imane, B.; Leena, S.; Samir, C. Virtual screening in drug-likeness and structure/activity relationship of pyridazine derivatives as anti-alzheimer drugs. J. King. Saud. Univ. Sci., 2019, 31, 595-601.
[http://dx.doi.org/10.1016/j.jksus.2018.03.024]
[31]
Vijesh, A.M.; Arun, M.I.; Sandeep, T.; Arulmoli, T.; Hoong-Kun, F. Molecular docking studies of some new imidazole derivatives for antimicrobial properties. Arab. J. Chem., 2013, 6, 197-204.
[http://dx.doi.org/10.1016/j.arabjc.2011.10.007]
[32]
Btissam, B.; Farid, A.; Abdelilah, T.; Manal, E.E.; Mounia, E.E.; Pierre, H.D.; Henri, D.; Rachid, T.; Mohammed, B. Identification of novel antifungal agents: antimicrobial evaluation, SAR, ADME-Tox and molecular docking studies of a series of imidazole derivatives. BMC. Chem, 2019, 13(100), 1-12.
[33]
Alsawalha, M.; Rao Bolla, S.; Kandakatla, N.; Srinivasadesikan, V.; Veeraraghavan, V.P.; Surapaneni, K.M. Molecular docking and ADMET analysis of hydroxamic acids as HDAC2 inhibitors. Bioinformation, 2019, 15(6), 380-387.
[http://dx.doi.org/10.6026/97320630015380] [PMID: 31312074]
[34]
Fiuze, D.A.; Begum, N.S.; Ulviye, A.C.; Ilhan, I.; Yusuf, O.; Hulya, K.G. Novel imidazole derivatives as antifungal agents: Synthesis, biological evaluation, ADME prediction and molecular docking studies. Phosphorus. Sulfur., 2019, 194(9), 887-894.
[35]
Vijayakumar, V.; Radhakrishnan, N.; Rameshkumar, C. Predeicting the biodegradability nature of imidazole and its derivatives by modulating two histidine degradation enzymes (urocanase and formiminoglutamase) activities. Asian. J. Pharm. Clin. Res., 2017, 10(11), 383-386.

Rights & Permissions Print Export Cite as
© 2024 Bentham Science Publishers | Privacy Policy