Your browser doesn't support javascript.
loading
Exploiting Metabolic Defects in Glioma with Nanoparticle Encapsulated NAMPT Inhibitors.
Murray, Matthew A; Noronha, Katelyn J; Wang, Yazhe; Friedman, Anna P; Paradkar, Sateja; Suh, Hee-Won; Sundaram, Ranjini K; Brenner, Charles; Saltzman, W Mark; Bindra, Ranjit S.
Afiliação
  • Murray MA; Yale University, New Haven, CT, United States.
  • Noronha KJ; Yale University, New Haven, CT, United States.
  • Wang Y; Yale University, New Haven, CT, United States.
  • Friedman AP; Yale School of Medicine, New Haven, CT, United States.
  • Paradkar S; Yale University, United States.
  • Suh HW; Dartmouth College, Hanover, NH, United States.
  • Sundaram RK; Yale School of Medicine, New Haven, United States.
  • Brenner C; University of Iowa, Iowa City, Iowa, United States.
  • Saltzman WM; Yale University, New Haven, CT, United States.
  • Bindra RS; Yale School of Medicine, New Haven, CT, United States.
Mol Cancer Ther ; 2024 May 01.
Article em En | MEDLINE | ID: mdl-38691846
ABSTRACT
The treatment of primary central nervous system (CNS) tumors is challenging due to the blood-brain barrier and complex mutational profiles, which is associated with low survival rates. However, recent studies have identified common mutations in gliomas (IDH-WT and mutant, WHO grades II-IV; with grade IV tumors referred to as glioblastomas; GBMs). These mutations drive epigenetic changes, leading to promoter methylation at the NAPRT gene locus, which encodes an enzyme involved in generating NAD+. Importantly, NAPRT-silencing introduces a therapeutic vulnerability to inhibitors targeting another NAD+ biogenesis enzyme, NAMPT, rationalizing a treatment for these malignancies. Multiple systemically-administered NAMPTis have been developed and tested in clinical trials, but dose-limiting toxicities-including bone marrow suppression and retinal toxicity-have limited their efficacy. Here, we report a novel approach for the treatment of NAPRT-silenced GBMs using nanoparticle-encapsulated (NP) NAMPT inhibitors (NAMPTis) administered by convection-enhanced delivery (CED). We demonstrate that GMX1778 (a NAMPTi) can be formulated in degradable polymer NPs with retention of potency for NAMPT inhibition and anticancer activity in vitro, plus sustained drug release in vitro and in vivo. Direct injection of these drugs via CED into the brain is associated with reduced retinal toxicity compared with systemic administration. Finally, we show that CED of NP-encapsulated GMX1778 to NAPRT-silenced intracranial GBM xenografts in mice exhibit significant tumor growth delay and extends survival. These data support an approach to treat gliomas harboring defects in NAD+ metabolism using CED of NP-encapsulated NAMPTis to greatly improve the therapeutic index and treatment efficacy for this class of drugs.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Mol Cancer Ther Assunto da revista: ANTINEOPLASICOS Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Mol Cancer Ther Assunto da revista: ANTINEOPLASICOS Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos