Three-kinase inhibitor combination recreates multipathway effects of a geldanamycin analogue on hepatocellular carcinoma cell death.

TitleThree-kinase inhibitor combination recreates multipathway effects of a geldanamycin analogue on hepatocellular carcinoma cell death.
Publication TypeJournal Article
Year of Publication2009
AuthorsPritchard, JR, Cosgrove, BD, Hemann, MT, Griffith, LG, Wands, JR, Lauffenburger, DA
JournalMol Cancer Ther
Volume8
Issue8
Pagination2183-92
Date Published2009 Aug
ISSN1538-8514
KeywordsApoptosis, Benzoquinones, Carcinoma, Hepatocellular, Cell Death, Cell Line, Tumor, Combined Modality Therapy, Humans, Lactams, Macrocyclic, Liver Neoplasms, Principal Component Analysis, Protein Kinase Inhibitors
Abstract

Multitarget compounds that act on a diverse set of regulatory pathways are emerging as a therapeutic approach for a variety of cancers. Toward a more specified use of this approach, we hypothesize that the desired efficacy can be recreated in terms of a particular combination of relatively more specific (i.e., ostensibly single target) compounds. We test this hypothesis for the geldanamycin analogue 17-Allylamino-17-demethoxygeldanamycin (17AAG) in hepatocellular carcinoma cells, measuring critical phosphorylation levels that indicate the kinase pathway effects correlating with apoptotic responsiveness of the Hep3B cell line in contrast to the apoptotic resistance of the Huh7 cell line. A principal components analysis (PCA) constructed from time course measurements of seven phosphoprotein signaling levels identified modulation of the AKT, IkappaB kinase, and signal transducer and activator of transcription 3 pathways by 17AAG treatment as most important for distinguishing these cell-specific death responses. The analysis correctly suggested from 17AAG-induced effects on these phosphoprotein levels that the FOCUS cell line would show apoptotic responsiveness similarly to Hep3B. The PCA also guided the inhibition of three critical pathways and rendered Huh7 cells responsive to 17AAG. Strikingly, in all three hepatocellular carcinoma lines, the three-inhibitor combination alone exhibited similar or greater efficacy to 17AAG. We conclude that (a) the PCA captures and clusters the multipathway phosphoprotein time courses with respect to their 17AAG-induced apoptotic responsiveness and (b) we can recreate, in a more specified manner, the cellular responses of a prospective multitarget cancer therapeutic.

DOI10.1158/1535-7163.MCT-08-1203
Alternate JournalMol. Cancer Ther.
PubMed ID19671754
PubMed Central IDPMC2779718
Grant ListU54 CA112967-05 / CA / NCI NIH HHS / United States