98%
921
2 minutes
20
Aurora kinases have emerged as important anticancer targets so that there are several inhibitors have advanced into clinical study. Herein, we identified novel indazole derivatives as potent Aurora kinases inhibitors by utilizing in silico fragment-based approach and knowledge-based drug design. After intensive hit-to-lead optimization, compounds 17 (dual Aurora A and B), 21 (Aurora B selective) and 30 (Aurora A selective) possessed indazole privileged scaffold with different substituents, which provide sub-type kinase selectivity. Computational modeling helps in understanding that the isoform selectivity could be targeted specific residue in the Aurora kinase binding pocket in particular targeting residues Arg220, Thr217 or Glu177.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1016/j.ejmech.2016.08.026 | DOI Listing |
Elife
September 2025
State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
Innate immune cells can acquire a memory phenotype, termed trained immunity, but the mechanism underlying the regulation of trained immunity remains largely elusive. Here, we demonstrate that inhibition of Aurora kinase A (AurA) dampens trained immunity induced by β-glucan. ATAC-seq and RNA-seq analysis reveal that AurA inhibition restricts chromatin accessibility of genes associated with inflammatory pathways such as JAK-STAT, TNF, and NF-κB pathways.
View Article and Find Full Text PDFFront Biosci (Landmark Ed)
August 2025
Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107 Shenzhen, Guangdong, China.
Background: Adenocarcinoma of Lung (LUAD) remains a leading cause of cancer-related deaths across the globe, and patients harboring epidermal growth factor receptor (EGFR) mutations frequently develop resistance to targeted therapies. While aurora kinase A (AURKA) has been implicated in tumorigenesis, its involvement in regulating ferroptosis via the kelch-like ECH-associated protein 1 (KEAP1)/NF-E2-related factor 2 (NRF2)/heme oxygenase 1 (HO‑1) signaling axis in EGFR-mutant LUAD remains poorly understood.
Methods: We analyzed RNA-seq and clinical data from 594 LUAD samples from The Cancer Genome Atlas (TCGA) to explore associations between AURKA expression, EGFR mutation status, and immune cell infiltration.
Mitosis in spp., the causative agent of malaria, is fundamentally different from model eukaryotes, proceeding via a bipartite microtubule organising centre (MTOC) and lacking canonical regulators such as Polo and Bub1 kinases. During schizogony, asynchronous nuclear replication produces a multinucleate schizont, while rapid male gametogony generates an octaploid nucleus before gamete formation.
View Article and Find Full Text PDFBioorg Chem
September 2025
Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran. Electronic address:
Multi-targeted agents can sequentially act on two or more targets, leading to synergistic and more effective therapeutic effects against several complicated disorders, containing cancer, even with relatively modest action. The TRKs (tropomyosin receptor kinases) are confirmed as promising targets in anti-tumor drug discovery. Over the past 20 years, many small molecules TRK inhibitors have been identified, that some of them are being investigated in various clinical phases.
View Article and Find Full Text PDFSci Signal
September 2025
Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
In -mutant melanoma cells treated with inhibitors of the kinases BRAF and MEK, a subset of cells rapidly and nongenetically adapts to escape drug-induced quiescence and reenters the cell cycle. Here, we investigated the mechanisms enabling this drug escape by computationally reconstructing single-cell lineages from time-lapse imaging data, linking dynamic signaling pathways to distinct cell-cycle fate outcomes. We found that reactivation of the MEK substrate ERK was necessary but not sufficient to drive escape; rather, the activity of the protein complex mTORC1 was also required to promote cell growth and protein synthesis in drug-treated cells destined for cell-cycle reentry.
View Article and Find Full Text PDF