Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Chem ; 215: 113288, 2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33640763

ABSTRACT

Kinesin spindle protein (KSP) is expressed only in cells undergoing cell division, and hence represents an attractive target for the treatment of cancer. Several KSP inhibitors have been developed and undergone clinical trial, but their clinical use is limited by their toxicity to rapidly proliferating non-cancerous cells. To create new KSP inhibitors that are highly selective for cancer cells, we optimized the amino acid moiety of S-trityl-l-cysteine (STLC) derivative 1 using in silico modeling. Molecular docking and molecular dynamics simulation were performed to investigate the binding mode of 1 with KSP. Consistent with the structure activity relationship studies, we found that a cysteine amino moiety plays an important role in stabilizing the interaction. Based on these findings and the structure of GSH, a substrate of γ-glutamyltransferase (GGT), we designed and synthesized the prodrug N-γ-glutamylated STLC derivative 9, which could be hydrolyzed by GGT to produce 1. The KSP ATPase inhibitory activity of 9 was lower than that of 1, and LC-MS analysis indicated that 9 was converted to 1 only in the presence of GGT in vitro. In addition, the cytotoxic activity of 9 was significantly attenuated in GGT-knockdown A549 cells. Since GGT is overexpressed on the cell membrane of various cancer cells, these results suggest that compound 9 could be a promising prodrug that selectively inhibits the proliferation of GGT-expressing cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cysteine/pharmacology , Dibenzocycloheptenes/pharmacology , Kinesins/antagonists & inhibitors , Prodrugs/pharmacology , Trityl Compounds/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cattle , Cell Line, Tumor , Cysteine/chemical synthesis , Cysteine/metabolism , Dibenzocycloheptenes/chemical synthesis , Dibenzocycloheptenes/metabolism , Humans , Kinesins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/metabolism , Protein Binding , Structure-Activity Relationship , Thermodynamics , Trityl Compounds/chemical synthesis , Trityl Compounds/metabolism , gamma-Glutamyltransferase/metabolism
2.
Biochem Biophys Res Commun ; 519(3): 505-511, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31530389

ABSTRACT

Kinesin centromere-associated protein E (CENP-E) has emerged as a potential target for the development of anticancer drugs due to its involvement in the mitotic progression of the cell cycle. Although several CENP-E inhibitors have been reported, more knowledge of chemical structures and inhibitory mechanisms is necessary for developing CENP-E inhibitors. Here, we describe the identification of new CENP-E inhibitors. Screening of a small-molecule chemical library identified benzo[d]pyrrolo[2,1-b]thiazole derivatives, including 1, as compounds with inhibitory activity against the microtubule-stimulated ATPase of the CENP-E motor domain. Among the mitotic kinesins examined, 1 selectively inhibited the kinesin ATPase activity of CENP-E. In a steady-state ATPase assay, 1 exhibited ATP-competitive behavior, which was different from the CENP-E inhibitor GSK923295. Compound 1 inhibited the proliferation of tumor-derived HeLa and HCT116 cells more efficiently than that of non-cancerous WI-38 cells. The inhibition of cell proliferation was attributed to the ability of 1 to induce apoptotic cell death. The compound showed antimitotic activity, which caused cell cycle arrest at mitosis via interference with proper chromosome alignment. We identified 1 and its derivatives as the lead compounds that target CENP-E, thus providing a new opportunity for the development of anticancer agents targeting kinesins.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Sarcosine/analogs & derivatives , Antineoplastic Agents/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , HeLa Cells , Humans , Molecular Structure , Sarcosine/chemistry , Sarcosine/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
3.
ACS Med Chem Lett ; 5(5): 566-71, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24900881

ABSTRACT

Diaryl amine derivatives have been designed and synthesized as novel kinesin spindle protein (KSP) inhibitors based on planar carbazole-type KSP inhibitors with poor aqueous solubility. The new generation of inhibitors was found to show comparable inhibitory activity and high selectivity for KSP, and this was accompanied with improved solubility. Kinetic analysis and molecular modeling studies suggested that these inhibitors work in an ATP-competitive manner via binding to the secondary allosteric site formed by α4 and α6 helices of KSP. Comparative structural investigations on a series of compounds revealed that the higher solubility of diaryl amine-type inhibitors was attributed to fewer van der Waals interactions in the crystal packing and the hydrogen-bond acceptor nitrogen of the aniline moiety for favorable solvation.

SELECTION OF CITATIONS
SEARCH DETAIL
...