CC BY 4.0 · Pharmaceutical Fronts 2021; 03(01): e1-e7
DOI: 10.1055/s-0041-1731081
Original Article

Discovery of SIPI6473, a New, Potent, and Orally Bioavailable Multikinase Inhibitor for the Treatment of Non-small Cell Lung Cancer

Xiu Gu
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
2   School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai, People's Republic of China
,
Zi-Xue Zhang
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Min-Ru Jiao
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Xin-Yan Peng
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Jian-Qi Li
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
,
Qing-Wei Zhang
1   Novel Technology Center of Pharmaceutical Chemistry, Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
› Author Affiliations
Funding This work was financially supported by the National Science and Technology Major Project (Grant No. 2018ZX09711002-002-009), the National Natural Science Foundation of China (Grant No. 81703358), and Science and Technology Commission of Shanghai Municipality (Grant No. 17431903900, 18QB1404200,18ZR1437700).


Abstract

A novel series of quinazoline derivatives were designed, synthesized, and evaluated as multikinase inhibitors. Most of these compounds showed antiproliferation activities of several human cancer cell lines and exhibited inhibition efficacy against the estimated glomerular filtration rate (EGFR) in the nanomolar level. Among those compounds, compound B5 (also named SIPI6473) displayed the maximum effect, and thus was chosen for further study. Our data revealed that B5 inhibited the activity of several kinases (such as EGFR, VEGFR2, and PDGFRα) that contributed to the development of non-small cell lung cancer (NSCLC). Besides, an in vivo study also showed that B5 inhibited tumor growth without signs of adverse effects in the A549 xenograft model. In conclusion, B5 may represent a new and promising drug for the treatment of NSCLC.



Publication History

Received: 09 March 2021

Accepted: 08 May 2021

Article published online:
23 June 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Shagufta AI, Ahmad I. An insight into the therapeutic potential of quinazoline derivatives as anticancer agents. Med Chem Commun 2017; 8 (05) 871-885
  • 2 Ravez S, Castillo-Aguilera O, Depreux P, Goossens L. Quinazoline derivatives as anticancer drugs: a patent review (2011 - present). Expert Opin Ther Pat 2015; 25 (07) 789-804
  • 3 Marzaro G, Guiotto A, Chilin A. Quinazoline derivatives as potential anticancer agents: a patent review (2007 - 2010). Expert Opin Ther Pat 2012; 22 (03) 223-252
  • 4 Das D, Hong J. Recent advancements of 4-aminoquinazoline derivatives as kinase inhibitors and their applications in medicinal chemistry. Eur J Med Chem 2019; 170: 55-72
  • 5 Chandregowda V, Kush AK, Chandrasekara Reddy G. Synthesis and in vitro antitumor activities of novel 4-anilinoquinazoline derivatives. Eur J Med Chem 2009; 44 (07) 3046-3055
  • 6 Ding C, Li D, Wang YW. et al. Discovery of ErbB/HDAC inhibitors by combining the core pharmacophores of HDAC inhibitor vorinostat and kinase inhibitors vandetanib, BMS-690514, neratinib, and TAK-285. Chin Chem Lett 2017; 28 (06) 1220-1227
  • 7 Kobayashi S, Boggon TJ, Dayaram T. et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med 2005; 352 (08) 786-792
  • 8 Gainor JF, Varghese AM, Ou SH. et al. ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an analysis of 1,683 patients with non-small cell lung cancer. Clin Cancer Res 2013; 19 (15) 4273-4281
  • 9 Naumov GN, Nilsson MB, Cascone T. et al. Combined vascular endothelial growth factor receptor and epidermal growth factor receptor (EGFR) blockade inhibits tumor growth in xenograft models of EGFR inhibitor resistance. Clin Cancer Res 2009; 15 (10) 3484-3494
  • 10 Akhavan D, Pourzia AL, Nourian AA. et al. De-repression of PDGFRβ transcription promotes acquired resistance to EGFR tyrosine kinase inhibitors in glioblastoma patients. Cancer Discov 2013; 3 (05) 534-547
  • 11 Ware KE, Hinz TK, Kleczko E. et al. A mechanism of resistance to gefitinib mediated by cellular reprogramming and the acquisition of an FGF2-FGFR1 autocrine growth loop. Oncogenesis 2013; 2 (03) e39
  • 12 Bean J, Brennan C, Shih JY. et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci U S A 2007; 104 (52) 20932-20937
  • 13 Gentile C, Martorana A, Lauria A, Bonsignore R. Kinase inhibitors in multitargeted cancer therapy. Curr Med Chem 2017; 24 (16) 1671-1686
  • 14 Medinger M, Drevs J. Receptor tyrosine kinases and anticancer therapy. Curr Pharm Des 2005; 11 (09) 1139-1149
  • 15 Markowitz JN, Fancher KM. Cabozantinib: a multitargeted oral tyrosine kinase inhibitor. Pharmacotherapy 2018; 38 (03) 357-369
  • 16 Skouras VS, Maragkos C, Grapsa D, Syrigos KN. Targeting neovasculature with multitargeted antiangiogenesis tyrosine kinase inhibitors in non-small cell lung cancer. BioDrugs 2016; 30 (05) 421-439
  • 17 Sun HP, Zhu J, Chen YD. et al. Docking study and three‐dimensional quantitative structure‐activity relationship (3D‐QSAR) analyses and novel molecular design of a series of 4‐aminoquinazolines as inhibitors of Aurora B kinase. Chin J Chem 2011; 29 (09) 1785-1799
  • 18 Reddy PS, Lokhande KB, Nagar S, Reddy VD, Murthy PS, Swamy KV. Molecular modeling, docking, dynamics and simulation of gefitinib and its derivatives with EGFR in non-small cell lung cancer. Curr Comput Aided Drug Des 2018; 14 (03) 246-252
  • 19 Dömötör O, Pelivan K, Borics A, Keppler BK, Kowol CR, Enyedy ÉA. Comparative studies on the human serum albumin binding of the clinically approved EGFR inhibitors gefitinib, erlotinib, afatinib, osimertinib and the investigational inhibitor KP2187. J Pharm Biomed Anal 2018; 154: 321-331
  • 20 Smaill JB, Gonzales AJ, Spicer JA. et al. Tyrosine kinase inhibitors. 20. Optimization of substituted quinazoline and pyrido[3,4-d]pyrimidine derivatives as orally active, irreversible inhibitors of the epidermal growth factor receptor family. J Med Chem 2016; 59 (17) 8103-8124
  • 21 Smaill JB, Palmer BD, Rewcastle GW. et al. Tyrosine kinase inhibitors. 15. 4-(Phenylamino)quinazoline and 4-(phenylamino)pyrido[d]pyrimidine acrylamides as irreversible inhibitors of the ATP binding site of the epidermal growth factor receptor. J Med Chem 1999; 42 (10) 1803-1815
  • 22 Chilin A, Conconi MT, Marzaro G. et al. Exploring epidermal growth factor receptor (EGFR) inhibitor features: the role of fused dioxygenated rings on the quinazoline scaffold. J Med Chem 2010; 53 (04) 1862-1866
  • 23 Chang J, Ren H, Zhao M. et al. Development of a series of novel 4-anlinoquinazoline derivatives possessing quinazoline skeleton: design, synthesis, EGFR kinase inhibitory efficacy, and evaluation of anticancer activities in vitro. Eur J Med Chem 2017; 138: 669-688
  • 24 Li DD, Fang F, Li JR. et al. Discovery of 6-substituted 4-anilinoquinazolines with dioxygenated rings as novel EGFR tyrosine kinase inhibitors. Bioorg Med Chem Lett 2012; 22 (18) 5870-5875
  • 25 Zhang HQ, Gong FH, Li CG. et al. Design and discovery of 4-anilinoquinazoline-acylamino derivatives as EGFR and VEGFR-2 dual TK inhibitors. Eur J Med Chem 2016; 109: 371-379
  • 26 Abouzid K, Shouman S. Design, synthesis and in vitro antitumor activity of 4-aminoquinoline and 4-aminoquinazoline derivatives targeting EGFR tyrosine kinase. Bioorg Med Chem 2008; 16 (16) 7543-7551
  • 27 Dubin AE, Nasser N, Rohrbacher J. et al. Identifying modulators of hERG channel activity using the PatchXpress planar patch clamp. J Biomol Screen 2005; 10 (02) 168-181