Generic placeholder image

Anti-Cancer Agents in Medicinal Chemistry

Editor-in-Chief

ISSN (Print): 1871-5206
ISSN (Online): 1875-5992

Research Article

Novel Pyrazolo[3,4-d]pyrimidines as Potential Cytotoxic Agents: Design, Synthesis, Molecular Docking and CDK2 Inhibition

Author(s): Mai Maher , Asmaa E. Kassab*, Ashraf F. Zaher and Zeinab Mahmoud

Volume 19, Issue 11, 2019

Page: [1368 - 1381] Pages: 14

DOI: 10.2174/1871520619666190417153350

Price: $65

conference banner
Abstract

Background: Pyrazolo[3,4-d]pyrimidine scaffold was reported to possess potent cytotoxic and CDK2 inhibitory activity as analogue of roscovitine.

Objective: To design and synthesize novel 1-(4-flourophenyl)pyrazolo[3,4-d]pyrimidine derivatives as bioisosters of roscovitine with potential cytotoxic and CDK2 inhibitory activity.

Methods: A series of novel 1-(4-flourophenyl)pyrazolo[3,4-d]pyrimidines were designed and synthesized. Structural elucidation for all the newly synthesized compounds was achieved through performing MS, 1H NMR, 13C NMR and IR spectral techniques. Eight compounds were screened for their cytotoxic activity by National Cancer Institute (USA) against 60 different human cancer cell lines. Compounds 2a, 4, 6, 7b, 8a and 8b were further studied through the determination of their IC50 values against the most sensitive cell lines. The inhibitory activities of compounds 2a and 4 were evaluated against CDK2 enzyme.

Results: Compound 4 exhibited the most prominent broad-spectrum cytotoxic activity against 42 cell lines representing all human cancer types showing growth inhibition percentages from 53.19 to 99.39. Compound 2a showed promising selectivity against several cell lines. Moreover, all the test compounds exhibited potent cytotoxic activity in nanomolar to micromolar range with IC50 values ranging from 0.58 to 8.32μM. Compound 2a showed significant cytotoxic activity against CNS (SNB-75), lung (NCI-H460) and ovarian (OVCAR-4) cancer cell lines with IC50 values 0.64, 0.78 and 1.9μM, respectively. Compound 4 showed promising potency against leukemia (HL-60) and CNS (SNB-75) cell lines (IC50 = 0.58 and 0.94μM, sequentially). Moreover, the antiproliferative activities of compounds 2a and 4 appeared to correlate well with their ability to inhibit CDK2 at sub-micromolar level (IC50 = 0.69 and 0.67μM, respectively) that were comparable to roscovitine (IC50=0.44μM). The Molecular docking results revealed that compound 4 interacted with the same key amino acids as roscovitine in the active site of CDK2 enzyme with a marked docking score (-14.1031 kcal/mol).

Conclusion: 1-(4-Flourophenyl)pyrazolo[3,4-d]pyrimidine is a promising scaffold for the design and synthesis of potent cytotoxic leads.

Keywords: Pyrazolo[3, 4-d]pyrimidines, design, synthesis, cytotoxicity, CDK2, roscovitine.

Graphical Abstract

[1]
Cancer trends progress report. January 2017. WHO Website http://www.who.int/cancer/en andhttp://progressreport.cancer.gov [Accessed July 11, 2017].
[2]
Peyressatre, M.; Prével, C.; Pellerano, M.; Morris, M. Targeting cyclin-dependent kinases in human cancers: From small molecules to Peptide inhibitors. Cancers, 2015, 7, 179-237.
[3]
Abate, A.A.; Pentimalli, F.; Esposito, L.; Giordano, A. ATP-noncompetitive CDK inhibitors for cancer therapy: An overview. Expert Opin. Investig. Drugs, 2013, 22, 895-906.
[4]
Lim, S.; Kaldis, P. Cdks, cyclins and CKIs: Roles beyond cell cycle regulation. Development, 2013, 140, 3079-3093.
[5]
Chung, J.H.; Bunz, F. CDK2 is required for p53-independent G2/M checkpoint control. PLoS Genet., 2010, 6(2), 1-12.
[6]
Gómez, G.G. Measurement of changes in apoptosis and cell cycle regulatory kinase Cdk2. Methods Mol. Biol., 2004, 282, 131-144.
[7]
Chashoo, G.; Saxena, A.K. Targetting CDKs in cancer : An overview and new insights. J. Cancer Sci. Ther., 2014, 6, 488-496.
[8]
Research and Markets, Global cancer CDK inhibitors market & clinical pipeline outlook 2022 report. Nov. 14. (2017). (Accessed May 24, 2018). https://www.researchandmarkets.com/research/ rx9snc/global_cancer_cdk
[9]
Veselý, J.; Havliček, L.; Strnad, M.; Blow, J.J.; Donella‐Deana, A.; Pinna, L.; Letham, D.S.; Kato, J.; Detivaud, L.; Leclerc, S.; Meijer, L. Inhibition of cyclin-dependent kinases by purine analogues. Eur. J. Biochem., 1994, 224, 771-786.
[10]
De Azevedo, W.F.; Leclerc, S.; Meijer, L.; Havlicek, L.; Strnad, M.; Kim, S.H. Inhibition of cyclin-dependent kinases by purine analogues: crystal structure of human CDK2 complexed with roscovitine. Eur. J. Biochem., 1997, 243, 518-526.
[11]
Chohan, T.A.; Qian, H.; Pan, Y.; Chen, J.Z. Cyclin-dependent kinase 2 as a target for cancer therapy: Progress in the development of CDK2 inhibitors as anti-cancer agents. Curr. Med. Chem., 2015, 22, 237-263.
[12]
Mariaule, G.; Belmont, P. Cyclin-dependent kinase inhibitors as marketed anticancer drugs: Where are we now? A short survey. Molecules, 2014, 19, 14366-14382.
[13]
Cyclacel Pharmaceuticals, Seliciclib. A novel, highly selective inhibitor of cyclin- dependent kinases 2, 7 and 9; in clinical development for hereditary gynecological cancers, Cushing’s Disease and rheumatoid arthritis. (2016) 1-2. http://www.cyclacel.com/ pdf/cyclacel_seliciclib_0216.pdf (accessed November 10, 2017).
[14]
Jorda, R.; Paruch, K.; Krystof, V. Cyclin-dependent kinase inhibitors inspired by Roscovitine: purine bioisosteres. Curr. Pharm. Des., 2012, 18, 2974-2980.
[15]
Heathcote, D.A.; Patel, H.; Kroll, S.H.B.; Hazel, P.; Periyasamy, M.; Alikian, M.; Kanneganti, S.K.; Jogalekar, A.S.; Scheiper, B.; Barbazanges, M.; Blum, A.; Brackow, J.; Siwicka, A.; Pace, R.D.M.; Fuchter, M.J.; Snyder, J.P.; Liotta, D.C.; Freemont, P.S.; Aboagye, E.O.; Coombes, R.C.; Barrett, A.G.M.; Ali, S. A novel pyrazolo[1,5-a]pyrimidine is a potent inhibitor of cyclin-dependent protein kinases 1, 2, and 9, which demonstrates antitumor effects in human tumor xenografts following oral administration. J. Med. Chem., 2010, 53, 8508-8522.
[16]
Bettayeb, K.; Sallam, H.; Ferandin, Y.; Popowycz, F.; Fournet, G.; Hassan, M.; Echalier, A.; Bernard, P.; Endicott, J.; Joseph, B.; Meijer, L. A new class of cell death-inducing kinase inhibitors derived from the purine Roscovitine. Mol. Cancer Ther., 2008, 7, 2713-2724.
[17]
Cherukupalli, S.; Chandrasekaran, B.; Kryštof, V.; Aleti, R.R.; Sayyad, N.; Merugu, S.R.; Kushwaha, N.D.; Karpoormath, R. Synthesis, anticancer evaluation, and molecular docking studies of some novel 4,6-disubstituted pyrazolo[3,4-d]pyrimidines as cyclin-dependent kinase 2 (CDK2) inhibitors. Bioorg. Chem., 2018, 79, 46-59.
[18]
Kim, D.C.; Lee, Y.R.; Yang, B.S.; Shin, K.J.; Kim, D.J.; Chung, B.Y.; Yoo, K.H. Synthesis and biological evaluations of pyrazolo[3,4-d]pyrimidines as cyclin-dependent kinase 2 inhibitors. Eur. J. Med. Chem., 2003, 38, 525-532.
[19]
Zask, A.; Verheijen, J.C.; Curran, K.; Kaplan, J.; Richard, D.J.; Nowak, P.; Malwitz, D.J.; Brooijmans, N.; Bard, J.; Svenson, K.; Lucas, J.; Toral-Barza, L.; Zhang, W.G.; Hollander, I.; Gibbons, J.J.; Abraham, R.T.; Ayral-Kaloustian, S.; Mansour, T.S.; Yu, K. ATP-competitive inhibitors of the mammalian target of rapamycin: Design and synthesis of highly potent and selective pyrazolopyrimidines. J. Med. Chem., 2009, 52, 5013-5016.
[20]
Kassab, A.E.; Gedawy, E.M. Synthesis and anticancer activity of novel 2-pyridyl hexahydrocyclooctathieno[2,3-d]pyrimidine derivatives. Eur. J. Med. Chem., 2013, 63, 224-230.
[21]
Kassab, A.E.; Gedawy, E.M.; El-Malah, A.A.; Abdelghany, T.M.; Abdel-Bakky, M.S. Synthesis, anticancer activity, effect on cell cycle profile, and apoptosis-inducing ability of novel hexahydrocyclooctathieno[2,3-d] pyrimidine derivatives. Chem. Pharm. Bull., 2016, 64, 490-496.
[22]
Kandeel, M.M.; Refaat, H.M.; Kassab, A.E.; Shahin, I.G.; Abdelghany, T.M. Synthesis, anticancer activity and effects on cell cycle profile and apoptosis of novel thieno[2,3-d]pyrimidine and thieno[3,2-e] triazolo[4,3-c]pyrimidine derivatives. Eur. J. Med. Chem., 2015, 90, 620-632.
[23]
Alley, M.C.; Scudiero, D.A.; Monks, P.A.; Hursey, M.L.; Fine, M.J.; Czerwinski, D.L.; Abbott, B.J.; Mayo, J.G.; Shoemaker, R.H.; Boyd, M.R. Feasibility of drug screening with panels of human tumor cell lines using a microculture tetrazolium assay. Cancer Res., 1988, 48, 589-601.
[24]
Grever, M.R.; Schepartz, S.A.; Chabner, B.A. The National Cancer Institute: Cancer drug discovery and development program. Semin. Oncol., 1992, 19, 622-638.
[25]
Boyd, M.R.; Paull, K.D. Some practical considerations and applications of the national cancer institute in vitro anticancer drug discovery screen. Drug Dev. Res., 1995, 34, 91-109.
[26]
Jiufu, L. Preparation of 1-aryl-3-substitution-5-substitution amino- 4- pyrazolecarboxamide derivatives as antitumor agent From Faming Zhuanli Shenqing. CN Patent 102,558,058 2012.
[27]
Hiroyuki, K.O.U.; Seigo, I.; Youichiro, N.; Naoki, S.; Takafumi, K.; Makio, C.Y. Naka. Preparation of pyrazolecarboxamides and pyrrolecarboxamides as inhibitors of the proliferation of activated lymphocytes and as remedies for autoimmune disease. WO Patent 2,000,047,558 2000.
[28]
Jorda, R.; Havlícek, L.; McNae, I.W.; Walkinshaw, M.D.; Voller, J.; Sturc, A.; Navrátilová, J.; Kuzma, M.; Mistrík, M.; Bártek, J.; Strnad, M.; Krystof, V. Pyrazolo[4,3-d]pyrimidine bioisostere of Roscovitine: evaluation of a novel selective inhibitor of cyclin-dependent kinases with antiproliferative activity. J. Med. Chem., 2011, 54, 2980-2993.
[29]
“RCSB Protein Data Bank - RCSB PDB. Available at. http://www. rcsb.org/pdb

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