RT Journal Article SR Electronic T1 Eradication of ENO1-deleted Glioblastoma through Collateral Lethality JF bioRxiv FD Cold Spring Harbor Laboratory SP 331538 DO 10.1101/331538 A1 Yu-Hsi Lin A1 Nikunj Satani A1 Naima Hammoudi A1 Jeffrey J. Ackroyd A1 Sunada Khadka A1 Victoria C. Yan A1 Dimitra K. Georgiou A1 Yuting Sun A1 Rafal Zielinski A1 Theresa Tran A1 Susana Castro Pando A1 Xiaobo Wang A1 David Maxwell A1 Zhenghong Peng A1 Federica Pisaneschi A1 Pijus Mandal A1 Paul G. Leonard A1 Quanyu Xu A1 Qi Wu A1 Yongying Jiang A1 Barbara Czako A1 Zhijun Kang A1 John M. Asara A1 Waldemar Priebe A1 William Bornmann A1 Joseph R. Marszalek A1 Ronald A. DePinho A1 Florian L. Muller YR 2018 UL http://biorxiv.org/content/early/2018/05/25/331538.abstract AB Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We recently demonstrated that SF2312, a natural product phosphonate antibiotic, is a potent inhibitor of the glycolytic enzyme Enolase with potential utility for the collateral lethality-based treatment of Enolase-deficient glioblastoma (GBM). However, phosphonates are anionic at physiological pH, limiting cell and tissue permeability. Here, we show that addition of pivaloyloxymethyl (POM) groups to SF2312 (POMSF) dramatically increases potency, leading to inhibition of glycolysis and killing of ENO1-deleted glioma cells in the low nM range. But the utility of POMSF in vivo is dose-limited by severe hemolytic anemia. A derivative, POMHEX, shows equipotency to POMSF without inducing hemolytic anemia. POMHEX can eradicate intracranial orthotopic ENO1-deleted tumors, despite sub-optimal pharmacokinetic properties. Taken together, our data provide in vivo proof-of-principal for collateral lethality in precision oncology and showcase POMHEX as a useful molecule for the study of glycolysis in cancer metabolism.