Back to Search Start Over

The addition of arginine deiminase potentiates Mithramycin A-induced cell death in patient-derived glioblastoma cells via ATF4 and cytochrome C.

Authors :
Linke, Charlotte
Freitag, Thomas
Riess, Christin
Scheffler, Jana Vanessa
del Moral, Katharina
Schoenwaelder, Nina
Fiedler, Tomas
Fiebig, Adina
Kaps, Philipp
Dubinski, Daniel
Schneider, Björn
Bergmann, Wendy
Classen, Carl Friedrich
Maletzki, Claudia
Source :
Cancer Cell International; 2/27/2023, Vol. 23 Issue 1, p1-18, 18p
Publication Year :
2023

Abstract

Background: Arginine auxotrophy constitutes a shortcoming for ~ 30% of glioblastoma multiforme (GBM). Indeed, arginine-depleting therapy using arginine deiminase from Streptococcus pyogenes (SpyADI) has proven activity against GBM in preclinical studies. The good safety profile of SpyADI renders this agent an ideal combination partner for cytostatic therapy. Methods: In this study, we combined the antineoplastic antibiotic Mithramycin A (MitA) with SpyADI to boost single-agent activity and analyzed underlying response mechanisms in-depth. Results: MitA monotherapy induced a time- and dose-dependent cytotoxicity in eight patient-derived GBM cell lines and had a radiosensitizing effect in all but one cell line. Combination treatment boosted the effects of the monotherapy in 2D- and 3D models. The simultaneous approach was superior to the sequential application and significantly impaired colony formation after repetitive treatment. MitA monotherapy significantly inhibited GBM invasiveness. However, this effect was not enhanced in the combination. Functional analysis identified SpyADI-triggered senescence induction accompanied by increased mitochondrial membrane polarization upon mono- and combination therapy. In HROG63, induction of lysosomes was seen after both monotherapies, indicative of autophagy. These cells seemed swollen and had a more pronounced cortically formed cytoskeleton. Also, cytochrome C and endoplasmatic reticulum-stress-associated proteins ATF4 and Calnexin were enhanced in the combination, contributing to apoptosis. Notably, no significant increases in glioma-stemness marker were seen. Conclusions: Therapeutic utilization of a metabolic defect in GBM along with cytostatic therapy provides a novel combination approach. Whether this SpyADI/MitA regimen will provide a safe alternative to combat GBM, will have to be addressed in subsequent (pre-)clinical trials. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
14752867
Volume :
23
Issue :
1
Database :
Complementary Index
Journal :
Cancer Cell International
Publication Type :
Academic Journal
Accession number :
162076142
Full Text :
https://doi.org/10.1186/s12935-023-02873-2