Fischer-Mertens, Janina, Otte, Felix, Roderwieser, Andrea, Rosswog, Carolina, Kahlert, Yvonne, Werr, Lisa, Hellmann, Anna-Maria, Berding, Maya, Chiu, Bill, Bartenhagen, Christoph and Fischer, Matthias (2022). Telomerase-targeting compounds Imetelstat and 6-thio-dG act synergistically with chemotherapy in high-risk neuroblastoma models. Cell. Oncol., 45 (5). S. 991 - 1004. DORDRECHT: SPRINGER. ISSN 2211-3436

Full text not available from this repository.

Abstract

Background The majority of high-risk neuroblastomas harbor telomerase activity, and telomerase-interacting compounds, such as 6-thio-2'-deoxyguanosine (6-thio-dG), have been found to impair the growth of telomerase-positive neuroblastoma cell lines. It has remained unclear, however, how such drugs can be combined with other compounds used in current treatment concepts for neuroblastoma patients. Methods Growth-inhibitory effects of varying concentrations of 6-thio-dG in combination with etoposide, doxorubicin or ceritinib were determined in eight telomerase-positive neuroblastoma cell lines with distinct genetic backgrounds. Tumor growth inhibition of subcutaneous xenografts from three different cell lines was assessed upon treatment with 6-thio-dG, the competitive telomerase inhibitor imetelstat, etoposide, or combinations of these compounds. Results Robust synergistic anti-tumor effects were observed for combinations of 6-thio-dG and etoposide or doxorubicin, but not for 6-thio-dG and ceritinib, in telomerase-positive neuroblastoma cell lines in vitro. Treatment of mouse xenografts with combinations of 6-thio-dG and etoposide significantly attenuated tumor growth and improved mouse survival over etoposide alone in two of three cell line models. Treatment of xenograft tumors by imetelstat monotherapy decreased telomerase activity by roughly 50% and significantly improved survival over control in all three models, whereas treatment with imetelstat plus etoposide led to enhanced survival over etoposide monotherapy in one model. Mechanistically, the synergistic effect was found to be due to both increased apoptosis and cell cycle arrest. Conclusion Our study indicates that telomerase is an actionable target in telomerase-positive neuroblastoma, and demonstrates that combination therapies including telomerase-interacting compounds may improve the efficacy of established cytotoxic drugs. Targeting telomerase may thus represent a therapeutic option in high-risk neuroblastoma patients.

Item Type: Journal Article
Creators:
CreatorsEmailORCIDORCID Put Code
Fischer-Mertens, JaninaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Otte, FelixUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Roderwieser, AndreaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Rosswog, CarolinaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Kahlert, YvonneUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Werr, LisaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Hellmann, Anna-MariaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Berding, MayaUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Chiu, BillUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Bartenhagen, ChristophUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
Fischer, MatthiasUNSPECIFIEDUNSPECIFIEDUNSPECIFIED
URN: urn:nbn:de:hbz:38-671058
DOI: 10.1007/s13402-022-00702-8
Journal or Publication Title: Cell. Oncol.
Volume: 45
Number: 5
Page Range: S. 991 - 1004
Date: 2022
Publisher: SPRINGER
Place of Publication: DORDRECHT
ISSN: 2211-3436
Language: English
Faculty: Unspecified
Divisions: Unspecified
Subjects: no entry
Uncontrolled Keywords:
KeywordsLanguage
ACTIVATING MUTATIONS; CANCER; ALK; INHIBITION; REARRANGEMENTS; COMBINATION; DIAGNOSIS; THERAPYMultiple languages
Oncology; Cell Biology; PathologyMultiple languages
URI: http://kups.ub.uni-koeln.de/id/eprint/67105

Downloads

Downloads per month over past year

Altmetric

Export

Actions (login required)

View Item View Item