Antitumor Activity of s-Triazine Derivatives: A Systematic Review
Abstract
:1. Introduction
2. The Antitumor Activity of s-Triazine Derivatives
2.1. Topoisomerases Inhibition
2.2. Tyrosine Kinases Inhibition
2.2.1. BTK-TKIs
2.2.2. EGFR-TKIs
2.2.3. FAK-TKIs
2.3. Phosphoinositide 3-Kinase Inhibition
2.4. NADP+-Dependent Isocitrate Dehydrogenases Inhibition
2.5. Cyclin-Dependent Kinases Inhibiton
2.6. Others
3. Conclusions and Prospect
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Singh, S.; Mandal, M.K.; Masih, A.; Saha, A.; Ghosh, S.K.; Bhat, H.R.; Singh, U.P. 1,3,5-Triazine: A versatile pharmacophore with diverse biological activities. Arch. Pharm. 2021, 354, 2000363–2000386. [Google Scholar] [CrossRef] [PubMed]
- Marin-Ocampo, L.; Angela Veloza, L.; Abonia, R.; Sepulveda-Arias, J.C. Anti-inflammatory activity of triazine derivatives: A systematic review. Eur. J. Med. Chem. 2019, 162, 435–447. [Google Scholar] [CrossRef] [PubMed]
- Keldsen, N.; Havsteen, H.; Vergote, I.; Bertelsen, K.; Jakobsen, A. Altretamine (hexamethylmelamine) in the treatment of platinum-resistant ovarian cancer: A phase II study. Gynecol. Oncol. 2003, 88, 118–122. [Google Scholar] [CrossRef] [PubMed]
- Champoux, J.J. DNA topoisomerases: Structure, function, and mechanism. Annu. Rev. Biochem. 2003, 88, 118–122. [Google Scholar] [CrossRef]
- Hu, W.; Huang, X.-S.; Wu, J.-F.; Yang, L.; Zheng, Y.-T.; Shen, Y.-M.; Li, Z.-Y.; Li, X. Discovery of novel topoisomerase II inhibitors by medicinal chemistry approaches. J. Med. Chem. 2018, 61, 8947–8980. [Google Scholar] [CrossRef]
- Furet, P.; Schoepfer, J.; Radimerski, T.; Chene, P. Discovery of a new class of catalytic topoisomerase II inhibitors targeting the ATP-binding site by structure based design. Part I. Bioorg. Med. Chem. Lett. 2009, 19, 4014–4017. [Google Scholar] [CrossRef]
- Pogorelcnik, B.; Brvar, M.; Zajc, I.; Filipic, M.; Solmajer, T.; Perdih, A. Monocyclic 4-amino-6-(phenylamino)-1,3,5-triazines as inhibitors of human DNA topoisomerase II alpha. Bioorg. Med. Chem. Lett. 2014, 24, 5762–5768. [Google Scholar] [CrossRef]
- Pogorelcnik, B.; Janezic, M.; Sosic, I.; Gobec, S.; Solmajer, T.; Perdih, A. 4,6-Substituted-1,3,5-triazin-2(H-1)-ones as monocyclic catalytic inhibitors of human DNA topoisomerase II alpha targeting the ATP binding site. Bioorg. Med. Chem. 2015, 23, 4218–4229. [Google Scholar] [CrossRef]
- Bergant, K.; Janezic, M.; Valjavec, K.; Sosic, I.; Pajk, S.; Stampar, M.; Zegura, B.; Gobec, S.; Filipic, M.; Perdih, A. Structure-guided optimization of 4,6-substituted-1,3,5-triazin-2(1H)-ones as catalytic inhibitors of human DNA topoisomerase II alpha. Eur. J. Med. Chem. 2019, 175, 330–348. [Google Scholar] [CrossRef]
- Liang, C.Y.; Tian, D.N.; Ren, X.D.; Ding, S.J.; Jia, M.Y.; Xin, M.H.; Thareja, S. The development of Bruton’s tyrosine kinase (BTK) inhibitors from 2012 to 2017: A mini-review. Eur. J. Med. Chem. 2018, 151, 315–326. [Google Scholar] [CrossRef]
- Teng, Y.; Lu, X.; Xiao, M.X.; Li, Z.B.; Zou, Y.M.; Ren, S.N.; Cheng, Y.; Luo, G.S.; Xiang, H. Discovery of potent and highly selective covalent inhibitors of Bruton’s tyrosine kinase bearing triazine scaffold. Eur. J. Med. Chem. 2020, 199, 112339. [Google Scholar] [CrossRef] [PubMed]
- Xiao, M.; Zhu, M.; Wu, S.; Ma, L.; Qi, L.; Ha, S.; Xiong, S.; Chen, M.; Chen, D.; Luo, G.; et al. Novel 6-amino-1,3,5-triazine derivatives as potent BTK inhibitors: Structure-activity relationship (SAR) analysis and preliminary mechanism investigation. Bioorg. Chem. 2023, 130, 106263. [Google Scholar] [CrossRef] [PubMed]
- Bai, F.; Liu, H.; Tong, L.; Zhou, W.; Liu, L.; Zhao, Z.; Liu, X.; Jiang, H.; Wang, X.; Xie, H.; et al. Discovery of novel selective inhibitors for EGFR-T790M/L858R. Bioorg. Med. Chem. Lett. 2012, 22, 1365–1370. [Google Scholar] [CrossRef] [PubMed]
- Srivastava, J.K.; Pillai, G.G.; Bhat, H.R.; Verma, A.; Singh, U.P. Design and discovery of novel monastrol-1,3,5-triazines as potent anti-breast cancer agent via attenuating Epidermal Growth Factor Receptor tyrosine kinase. Sci. Rep. 2017, 7, 1038–1045. [Google Scholar] [CrossRef]
- Bhat, H.R.; Singh, U.P.; Thakur, A.; Ghosh, S.K.; Gogoi, K.; Prakash, A.; Singh, R.K. Design, synthesis, anticancer, antibacterial, and antifungal evaluation of 4-aminoquinoline-1,3,5-triazine derivatives. J. Heterocycl. Chem. 2019, 57, 390–399. [Google Scholar] [CrossRef]
- Pathak, P.; Rimac, H.; Grishina, M.; Verma, A.; Potemkin, V. Hybrid quinazoline 1,3,5-triazines as Epidermal Growth Factor Receptor (EGFR) Inhibitors with anticancer activity: Design, synthesis, and computational study. ChemMedChem 2021, 16, 822–838. [Google Scholar] [CrossRef]
- Yan, W.; Zhao, Y.; He, J. Anti-breast cancer activity of selected 1,3,5-triazines via modulation of EGFR-TK. Mol. Med. Rep. 2018, 18, 4175–4184. [Google Scholar] [CrossRef]
- Raghu, M.S.; Pradeep Kumar, C.B.; Prashanth, M.K.; Yogesh Kumar, K.; Prathibha, B.S.; Kanthimathi, G.; Alissa, S.A.; Alghulikah, H.A.; Osman, S.M. Novel 1,3,5-triazine-based pyrazole derivatives as potential antitumor agents and EFGR kinase inhibitors: Synthesis, cytotoxicity, DNA binding, molecular docking and DFT studies. New J. Chem. 2021, 45, 13909–13924. [Google Scholar] [CrossRef]
- Shawish, I.; Barakat, A.; Aldalbahi, A.; Malebari, A.M.; Nafie, M.S.; Bekhit, A.A.; Albohy, A.; Khan, A.; Ul-Haq, Z.; Haukka, M.; et al. Synthesis and Antiproliferative Activity of a New Series of Mono- and Bis(dimethylpyrazolyl)-s-triazine Derivatives Targeting EGFR/ PI3K/AKT/mTOR Cascades. Acs Omega 2022, 7, 24858–24870. [Google Scholar] [CrossRef]
- Shawish, I.; Barakat, A.; Aldalbahi, A.; Alshaer, W.; Daoud, F.; Alqudah, D.A.; Al Zoubi, M.; Hatmal, M.m.M.; Nafie, M.S.; Haukka, M.; et al. Acetic Acid Mediated for One-Pot Synthesis of Novel Pyrazolyl s-Triazine Derivatives for the Targeted Therapy of Triple-Negative Breast Tumor Cells (MDA-MB-231) via EGFR/PI3K/AKT/mTOR Signaling Cascades. Pharmaceutics 2022, 14, 1558. [Google Scholar] [CrossRef]
- Hashem, H.E.; Amr, A.E.-G.E.; Nossier, E.S.; Anwar, M.M.; Azmy, E.M. New Benzimidazole-, 1,2,4-Triazole-, and 1,3,5-Triazine-Based Derivatives as Potential EGFRWT and EGFRT790M Inhibitors: Microwave-Assisted Synthesis, Anticancer Evaluation, and Molecular Docking Study. Acs Omega 2022, 7, 7155–7171. [Google Scholar] [CrossRef] [PubMed]
- Pang, X.-J.; Liu, X.-J.; Liu, Y.; Liu, W.-B.; Li, Y.-R.; Yu, G.-X.; Tian, X.-Y.; Zhang, Y.-B.; Song, J.; Jin, C.-Y.; et al. Drug Discovery Targeting Focal Adhesion Kinase (FAK) as a Promising Cancer Therapy. Molecules 2021, 26, 4250. [Google Scholar] [CrossRef] [PubMed]
- Dao, P.; Jarray, R.; Le Coq, J.; Lietha, D.; Loukaci, A.; Lepelletier, Y.; Hadj-Slimane, R.; Garbay, C.; Raynaud, F.; Chen, H. Synthesis of novel diarylamino-1,3,5-triazine derivatives as FAK inhibitors with anti-angiogenic activity. Bioorg. Med. Chem. Lett. 2013, 23, 4552–4556. [Google Scholar] [CrossRef]
- Dao, P.; Smith, N.; Tornkiewicz-Raulet, C.; Yen-Pon, E.; Carnacho-Artacho, M.; Lietha, D.; Herbeuval, J.-P.; Commoul, X.; Garbay, C.; Chen, H. Design, Synthesis, and Evaluation of Novel Imidazo 1,2-a 1,3,5 triazines and Their Derivatives as Focal Adhesion Kinase Inhibitors with Antitumor Activity. J. Med. Chem. 2015, 58, 237–251. [Google Scholar] [CrossRef] [PubMed]
- Tong, G.; Peng, T.; Chen, Y.; Sha, L.; Dai, H.; Xiang, Y.; Zou, Z.; He, H.; Wang, S. Effects of GLP-1 Receptor Agonists on Biological Behavior of Colorectal Cancer Cells by Regulating PI3K/AKT/mTOR Signaling Pathway. Front. Pharmacol. 2022, 13, 901559. [Google Scholar] [CrossRef]
- Bertucci, A.; Bertucci, F.; Goncalves, A. Phosphoinositide 3-Kinase (PI3K) Inhibitors and Breast Cancer: An Overview of Current Achievements. Cancers 2023, 15, 1416. [Google Scholar] [CrossRef]
- Yaguchi, S.; Izumisawa, Y.; Sato, M.; Nakagane, T.; Koshimizu, I.; Sakita, K.; Kato, M.; Yoshioka, K.; Sakato, M.; Kawashima, S. In vitro cytotoxicity of imidazolyl-1,3,5-triazine derivatives. Biol. Pharm. Bull. 1997, 20, 698–700. [Google Scholar] [CrossRef]
- Kong, D.; Yamori, T. ZSTK474 is an ATP-competitive inhibitor of class I phosphatidylinositol 3 kinase isoforms. Cancer Sci. 2007, 98, 1638–1642. [Google Scholar] [CrossRef]
- Vanhaesebroeck, B.; Perry, M.W.D.; Brown, J.R.; Andre, F.; Okkenhaug, K. PI3K inhibitors are finally coming of age. Nat. Rev. Drug Discov. 2021, 20, 741–769. [Google Scholar] [CrossRef]
- Van Dort, M.E.; Galban, S.; Nino, C.A.; Hong, H.; Apfelbaum, A.A.; Luker, G.D.; Thurber, G.M.; Atangcho, L.; Besirli, C.G.; Ross, B.D. Structure-Guided Design and Initial Studies of a Bifunctional MEK/PI3K Inhibitor(ST-168). Acs Med. Chem. Lett. 2017, 8, 808–813. [Google Scholar] [CrossRef]
- Van Dort, M.E.; Jang, Y.; Bonham, C.A.; Heist, K.; McDonald, L.; Zhang, E.Z.; Chenevert, T.L.; Luker, G.D.; Ross, B.D. Structural effects of morpholine replacement in ZSTK474 on Class I PI3K isoform inhibition: Development of novel MEK/PI3K bifunctional inhibitors. Eur. J. Med. Chem. 2022, 229, 113996. [Google Scholar] [CrossRef] [PubMed]
- Rewcastle, G.W.; Gamage, S.A.; Flanagan, J.U.; Frederick, R.; Denny, W.A.; Baguley, B.C.; Kestell, P.; Singh, R.; Kendall, J.D.; Marshall, E.S.; et al. Synthesis and Biological Evaluation of Novel Analogues of the Pan Class I Phosphatidylinositol 3-Kinase (PI3K) Inhibitor 2-(Difluoromethyl)-1- 4,6-di(4-morpholinyl)-1,3,5-triazin-2-yl -1H-benzi midazole (ZSTK474). J. Med. Chem. 2011, 54, 7105–7126. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Liu, Y.; Ge, T.; Tang, J.; Wang, S.; Gao, Z.; Chen, J.; Xu, J.; Gong, P.; Zhao, Y.; et al. Based on 2-(difluoromethyl)-1- 4,6-di (4-morpholinyl)-1,3,5-triazin-2-yl -1H-benzimidazole (ZSTK474), design, synthesis and biological evaluation of novel PI3K alpha selective inhibitors. Bioorg. Chem. 2023, 130, 106211. [Google Scholar] [CrossRef] [PubMed]
- Venkatesan, A.M.; Dehnhardt, C.M.; Delos Santos, E.; Chen, Z.; Dos Santos, O.; Ayral-Kaloustian, S.; Khafizova, G.; Brooijmans, N.; Mallon, R.; Hollander, I.; et al. Bis(morpholino-1,3,5-triazine) Derivatives: Potent Adenosine 5′-Triphosphate Competitive Phosphatidylinositol-3-kinase/Mammalian Target of Rapamycin Inhibitors: Discovery of Compound 26 (PKI-587), a Highly Efficacious Dual Inhibitor. J. Med. Chem. 2010, 53, 2636–2645. [Google Scholar] [CrossRef] [PubMed]
- Venkatesan, A.M.; Chen, Z.; Dos Santos, O.; Dehnhardt, C.; Delos Santos, E.; Ayral-Kaloustian, S.; Mallon, R.; Hollander, I.; Feldberg, L.; Lucas, J.; et al. PKI-179: An orally efficacious dual phosphatidylinositol-3-kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitor. Bioorg. Med. Chem. Lett. 2010, 20, 5869–5873. [Google Scholar] [CrossRef]
- Chen, Z.; Venkatesan, A.M.; Dos Santos, O.; Delos Santos, E.; Dehnhardt, C.M.; Ayral-Kaloustian, S.; Ashcroft, J.; McDonald, L.A.; Mansour, T.S. Stereoselective Synthesis of an Active Metabolite of the Potent PI3 Kinase Inhibitor PKI-179. J. Org. Chem. 2010, 75, 1643–1651. [Google Scholar] [CrossRef]
- Wu, T.-T.; Guo, Q.-Q.; Chen, Z.-L.; Wang, L.-L.; Du, Y.; Chen, R.; Mao, Y.-H.; Yang, S.-G.; Huang, J.; Wang, J.-T.; et al. Design, synthesis and bioevaluation of novel substituted triazines as potential dual PI3K/mTOR inhibitors. Eur. J. Med. Chem. 2020, 204, 112637. [Google Scholar] [CrossRef]
- Bohnacker, T.; Prota, A.E.; Beaufils, F.; Burke, J.E.; Melone, A.; Inglis, A.J.; Rageot, D.; Sele, A.M.; Cmiljanovic, V.; Cmiljanovic, N.; et al. Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat. Commun. 2017, 8, 14683. [Google Scholar] [CrossRef]
- Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C.; et al. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-ami ne (PQR309), a Potent, Brain-Penetrant, Orally Bioavailable, Pan-Class I PI3K/mTOR Inhibitor as Clinical Candidate in Oncology. J. Med. Chem. 2017, 60, 7524–7538. [Google Scholar] [CrossRef]
- Tarantelli, C.; Gaudio, E.; Arribas, A.J.; Kwee, I.; Hillmann, P.; Rinaldi, A.; Cascione, L.; Spriano, F.; Bernasconi, E.; Guidetti, F.; et al. PQR309 Is a Novel Dual PI3K/mTOR Inhibitor with Preclinical Antitumor Activity in Lymphomas as a Single Agent and in Combination Therapy. Clin. Cancer Res. 2018, 24, 120–129. [Google Scholar] [CrossRef]
- Wicki, A.; Brown, N.; Xyrafas, A.; Bize, V.; Hawle, H.; Berardi, S.; Cmiljanovic, N.; Cmiljanovic, V.; Stumm, M.; Dimitrijevic, S.; et al. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13). Eur. J. Cancer 2018, 96, 6–16. [Google Scholar] [CrossRef] [PubMed]
- Borsari, C.; Rageot, D.; Beaufils, F.; Bohnacker, T.; Keles, E.; Buslov, I.; Melone, A.; Sele, A.M.; Hebeisen, P.; Fabbro, D.; et al. Preclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl-Pyrimidine Moiety. Acs Med. Chem. Lett. 2019, 10, 1473–1479. [Google Scholar] [CrossRef] [PubMed]
- Borsari, C.; Keles, E.; McPhail, J.A.; Schaefer, A.; Sriramaratnam, R.; Goch, W.; Schaefer, T.; De Pascale, M.; Bal, W.; Gstaiger, M.; et al. Covalent Proximity Scanning of a Distal Cysteine to Target PI3K alpha. J. Am. Chem. Soc. 2022, 144, 6326–6342. [Google Scholar] [CrossRef]
- Li, W.; Sun, Q.; Song, L.; Gao, C.; Liu, F.; Chen, Y.; Jiang, Y. Discovery of 1-(3-aryl-4-chloropheny1)-3-(p-aryl)urea derivatives against breast cancer by inhibiting PI3K/Akt/mTOR and Hedgehog signalings. Eur. J. Med. Chem. 2017, 141, 721–733. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Gao, C.; Zhao, L.; Yuan, Z.; Chen, Y.; Jiang, Y. Phthalimide conjugations for the degradation of oncogenic PI3K. Eur. J. Med. Chem. 2018, 151, 237–247. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, Y.H.; Garcia-Garcia, C.; Serra, V.; He, L.; Torres-Lockhart, K.; Prat, A.; Anton, P.; Cozar, P.; Guzman, M.; Grueso, J.; et al. PI3K Inhibition Impairs BRCA1/2 Expression and Sensitizes BRCA-Proficient Triple-Negative Breast Cancer to PARP Inhibition. Cancer Discov. 2012, 2, 1036–1047. [Google Scholar] [CrossRef]
- Juvekar, A.; Burga, L.N.; Hu, H.; Lunsford, E.P.; Ibrahim, Y.H.; Balmana, J.; Rajendran, A.; Papa, A.; Spencer, K.; Lyssiotis, C.A.; et al. Combining a PI3K Inhibitor with a PARP Inhibitor Provides an Effective Therapy for BRCA1-Related Breast Cancer. Cancer Discov. 2012, 2, 1048–1063. [Google Scholar] [CrossRef]
- Wang, J.; Li, H.; He, G.; Chu, Z.; Peng, K.; Ge, Y.; Zhu, Q.; Xu, Y. Discovery of Novel Dual Poly(ADP-ribose)polymerase and Phosphoinositide 3-Kinase Inhibitors as a Promising Strategy for Cancer Therapy. J. Med. Chem. 2020, 63, 122–139. [Google Scholar] [CrossRef]
- Wang, J.; He, G.; Li, H.; Ge, Y.; Wang, S.; Xu, Y.; Zhu, Q. Discovery of novel PARP/PI3K dual inhibitors with high efficiency against BRCA-proficient triple negative breast cancer. Eur. J. Med. Chem. 2021, 213, 113054. [Google Scholar] [CrossRef]
- Van Dort, M.E.; Galban, S.; Wang, H.; Sebolt-Leopold, J.; Whitehead, C.; Hong, H.; Rehemtulla, A.; Ross, B.D. Dual inhibition of allosteric mitogen-activated protein kinase (MEK) and phosphatidylinositol 3-kinase (PI3K) oncogenic targets with a bifunctional inhibitor. Bioorg. Med. Chem. 2015, 23, 1386–1394. [Google Scholar] [CrossRef]
- Van Dort, M.E.; Hong, H.; Wang, H.; Nino, C.A.; Lombardi, R.L.; Blanks, A.E.; Galban, S.; Ross, B.D. Discovery of Bifunctional Oncogenic Target Inhibitors against Allosteric Mitogen-Activated Protein Kinase (MEK1) and Phosphatidylinositol 3-Kinase (PI3K). J. Med. Chem. 2016, 59, 2512–2522. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Zhang, Q.; Xiao, Z.; Sun, X.; Yang, Z.; Gu, Q.; Liu, Z.; Xie, T.; Jin, Q.; Zheng, P.; et al. Design, synthesis and biological evaluation of substituted 2-(thiophen-2-yl)-1,3,5-triazine derivatives as potential dual PI3K alpha/mTOR inhibitors. Bioorg. Chem. 2020, 95, 103525. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Zhang, B.; Luo, L.; Yang, Y.; He, B.; Zhang, Q.; Wang, L.; Xu, S.; Zheng, P.; Zhu, W. Design, synthesis and pharmacological evaluation of 2-arylurea-1,3,5-tria-zine derivative (XIN-9): A novel potent dual PI3K/mTOR inhibitor for cancer therapy. Bioorg. Chem. 2022, 129, 106157. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Luo, L.; Sun, X.; Yang, Y.; Guo, Q.; Jiang, Z.; Wu, Y. Design, synthesis and antitumor activity of novel thiophene- triazine derivatives bearing arylurea unit as potent PI3K/mTOR inhibitorss. Bioorganic Med. Chem. 2023, 78, 117133. [Google Scholar] [CrossRef] [PubMed]
- Kim, E.S. Enasidenib: First Global Approval. Drugs 2017, 77, 1705–1711. [Google Scholar] [CrossRef]
- Yen, K.; Travins, J.; Wang, F.; David, M.D.; Artin, E.; Straley, K.; Padyana, A.; Gross, S.; DeLaBarre, B.; Tobin, E.; et al. AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations. Cancer Discov. 2017, 7, 478–493. [Google Scholar] [CrossRef]
- Teng, Y.; Lu, K.; Zhang, Q.; Zhao, L.; Huang, Y.; Ingarra, A.M.; Galons, H.; Li, T.; Cui, S.; Yu, P.; et al. Recent advances in the development of cyclin-dependent kinase 7 inhibitors. Eur. J. Med. Chem. 2019, 183, 111641. [Google Scholar] [CrossRef]
- Kuo, G.H.; DeAngelis, A.; Emanuel, S.; Wang, A.H.; Zhang, Y.; Connolly, P.J.; Chen, X.; Gruninger, R.H.; Rugg, C.; Fuentes-Pesquera, A.; et al. Synthesis and identification of 1,3,5 triazine-pyridine biheteroaryl as a novel series of potent cyclin-dependent kinase inhibitors. J. Med. Chem. 2005, 48, 4535–4546. [Google Scholar] [CrossRef]
- Luecking, U.; Scholz, A.; Lienau, P.; Siemeister, G.; Kosemund, D.; Bohlmann, R.; Briem, H.; Terebesi, I.; Meyer, K.; Prelle, K.; et al. Identification of Atuveciclib (BAY 1143572), the First Highly Selective, Clinical PTEFb/CDK9 Inhibitor for the Treatment of Cancer. ChemMedChem 2017, 12, 1776–1793. [Google Scholar] [CrossRef]
- Masih, A.; Singh, S.; Agnihotri, A.K.; Giri, S.; Shrivastava, J.K.; Pandey, N.; Bhat, H.R.; Singh, U.P. Design and development of 1,3,5-triazine-thiadiazole hybrids as potent adenosine A(2)A receptor (A(2)AR) antagonist for benefit in Parkinson’s disease. Neurosci. Lett. 2020, 735, 135222. [Google Scholar] [CrossRef]
- Masih, A.; Agnihotri, A.K.; Srivastava, J.K.; Pandey, N.; Bhat, H.R.; Singh, U.P. Discovery of novel 1,3,5-triazine as adenosine A(2A) receptor antagonist for benefit in Parkinson’s disease. J. Biochem. Mol. Toxicol. 2021, 35, e22659. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Ahn, Y.; Kim, Y.; Roh, E.J.; Lee, Y.; Han, C.; Yoo, H.M.; Yu, J. Design, Synthesis and Biological Evaluation of 1,3,5-Triazine Derivatives Targeting hA1 and hA3 Adenosine Receptor. Molecules 2022, 27, 4016. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.-l.; Liu, P.; Zhu, W.-l.; Lou, L.-g. DCZ5248, a novel dual inhibitor of Hsp90 and autophagy, exerts antitumor activity against colon cancer. Acta Pharmacol. Sin. 2021, 42, 132–141. [Google Scholar] [CrossRef] [PubMed]
- Zarganes-Tzitzikas, T.; Konstantinidou, M.; Gao, Y.; Krzemien, D.; Zak, K.; Dubin, G.; Holak, T.A.; Domling, A. Inhibitors of programmed cell death 1 (PD-1): A patent review (2010–2015). Expert Opin. Ther. Pat. 2016, 26, 973–977. [Google Scholar] [CrossRef]
- Russomanno, P.; Assoni, G.; Amato, J.; D’Amore, V.M.; Scaglia, R.; Brancaccio, D.; Pedrini, M.; Polcaro, G.; La Pietra, V.; Orlando, P.; et al. Interfering with the Tumor-Immune Interface: Making Way for Triazine-Based Small Molecules as Novel PD-L1 Inhibitors. J. Med. Chem. 2021, 64, 16020–16045. [Google Scholar] [CrossRef]
- Haiba, N.S.; Khalil, H.H.; Bergas, A.; Abu-Serie, M.M.; Khattab, S.N.; Teleb, M. First-in-Class Star-Shaped Triazine Dendrimers Endowed with MMP-9 Inhibition and VEGF Suppression Capacity: Design, Synthesis, and Anticancer Evaluation. Acs Omega 2022, 7, 21131–21144. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Dai, Q.; Sun, Q.; Ouyang, X.; Liu, J.; Jin, L.; Liu, A.; He, B.; Fan, T.; Jiang, Y. Antitumor Activity of s-Triazine Derivatives: A Systematic Review. Molecules 2023, 28, 4278. https://doi.org/10.3390/molecules28114278
Dai Q, Sun Q, Ouyang X, Liu J, Jin L, Liu A, He B, Fan T, Jiang Y. Antitumor Activity of s-Triazine Derivatives: A Systematic Review. Molecules. 2023; 28(11):4278. https://doi.org/10.3390/molecules28114278
Chicago/Turabian StyleDai, Qiuzi, Qinsheng Sun, Xiaorong Ouyang, Jinyang Liu, Liye Jin, Ahao Liu, Binsheng He, Tingting Fan, and Yuyang Jiang. 2023. "Antitumor Activity of s-Triazine Derivatives: A Systematic Review" Molecules 28, no. 11: 4278. https://doi.org/10.3390/molecules28114278
APA StyleDai, Q., Sun, Q., Ouyang, X., Liu, J., Jin, L., Liu, A., He, B., Fan, T., & Jiang, Y. (2023). Antitumor Activity of s-Triazine Derivatives: A Systematic Review. Molecules, 28(11), 4278. https://doi.org/10.3390/molecules28114278