Unknown

Dataset Information

0

The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in IDH1-Mutant Cancers and Potentiates NAD+ Depletion-Mediated Cytotoxicity.


ABSTRACT: IDH1-mutant gliomas are dependent upon the canonical coenzyme NAD+ for survival. It is known that PARP activation consumes NAD+ during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD+ biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD+ depletion-mediated cytotoxicity in mutant IDH1 cancer cells. To investigate the impact of temozolomide on NAD+ metabolism, patient-derived xenografts and engineered mutant IDH1-expressing cell lines were exposed to temozolomide, in vitro and in vivo, both alone and in combination with nicotinamide phosphoribosyltransferase (NAMPT) inhibitors, which block NAD+ biosynthesis. The acute time period (<3 hours) after temozolomide treatment displayed a burst of NAD+ consumption driven by PARP activation. In IDH1-mutant-expressing cells, this consumption reduced further the abnormally lowered basal steady-state levels of NAD+, introducing a window of hypervulnerability to NAD+ biosynthesis inhibitors. This effect was selective for IDH1-mutant cells and independent of methylguanine methyltransferase or mismatch repair status, which are known rate-limiting mediators of adjuvant temozolomide genotoxic sensitivity. Combined temozolomide and NAMPT inhibition in an in vivo IDH1-mutant cancer model exhibited enhanced efficacy compared with each agent alone. Thus, we find IDH1-mutant cancers have distinct metabolic stress responses to chemotherapy-induced DNA damage and that combination regimens targeting nonredundant NAD+ pathways yield potent anticancer efficacy in vivo Such targeting of convergent metabolic pathways in genetically selected cancers could minimize treatment toxicity and improve durability of response to therapy. Cancer Res; 77(15); 4102-15. ©2017 AACR.

SUBMITTER: Tateishi K 

PROVIDER: S-EPMC5783559 | biostudies-literature | 2017 Aug

REPOSITORIES: biostudies-literature

altmetric image

Publications

The Alkylating Chemotherapeutic Temozolomide Induces Metabolic Stress in <i>IDH1</i>-Mutant Cancers and Potentiates NAD<sup>+</sup> Depletion-Mediated Cytotoxicity.

Tateishi Kensuke K   Higuchi Fumi F   Miller Julie J JJ   Koerner Mara V A MVA   Lelic Nina N   Shankar Ganesh M GM   Tanaka Shota S   Fisher David E DE   Batchelor Tracy T TT   Iafrate A John AJ   Wakimoto Hiroaki H   Chi Andrew S AS   Cahill Daniel P DP  

Cancer research 20170616 15


<i>IDH1</i>-mutant gliomas are dependent upon the canonical coenzyme NAD<sup>+</sup> for survival. It is known that PARP activation consumes NAD<sup>+</sup> during base excision repair (BER) of chemotherapy-induced DNA damage. We therefore hypothesized that a strategy combining NAD<sup>+</sup> biosynthesis inhibitors with the alkylating chemotherapeutic agent temozolomide could potentiate NAD<sup>+</sup> depletion-mediated cytotoxicity in mutant <i>IDH1</i> cancer cells. To investigate the impac  ...[more]

Similar Datasets

| S-EPMC4684594 | biostudies-literature
| S-EPMC5362526 | biostudies-literature
| S-EPMC7017040 | biostudies-literature
| S-EPMC7669718 | biostudies-literature
| S-EPMC8127964 | biostudies-literature
| S-EPMC6856069 | biostudies-literature
2023-03-10 | MSV000091468 | MassIVE