Your browser doesn't support javascript.
loading
Triptolide and its prodrug Minnelide target high-risk MYC-amplified medulloblastoma in preclinical models.
Rodriguez-Blanco, Jezabel; Salvador, April D; Suter, Robert K; Swiderska-Syn, Marzena; Palomo-Caturla, Isabel; Kliebe, Valentin; Shahani, Pritika; Peterson, Kendell; Turos-Cabal, Maria; Vieira, Megan E; Wynn, Daniel T; Howell, Ashley J; Yang, Fan; Ban, Yuguang; McCrea, Heather J; Zindy, Frederique; Danis, Etienne; Vibhakar, Rajeev; Jermakowicz, Anna; Martin, Vanesa; Coss, Christopher C; Harris, Brent T; de Cubas, Aguirre; Chen, X Steven; Barnoud, Thibaut; Roussel, Martine F; Ayad, Nagi G; Robbins, David J.
Affiliation
  • Rodriguez-Blanco J; Darby Children's Research Institute, Department of Pediatrics, and.
  • Salvador AD; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA.
  • Suter RK; Darby Children's Research Institute, Department of Pediatrics, and.
  • Swiderska-Syn M; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA.
  • Palomo-Caturla I; Darby Children's Research Institute, Department of Pediatrics, and.
  • Kliebe V; Darby Children's Research Institute, Department of Pediatrics, and.
  • Shahani P; Darby Children's Research Institute, Department of Pediatrics, and.
  • Peterson K; Darby Children's Research Institute, Department of Pediatrics, and.
  • Turos-Cabal M; Darby Children's Research Institute, Department of Pediatrics, and.
  • Vieira ME; Darby Children's Research Institute, Department of Pediatrics, and.
  • Wynn DT; Darby Children's Research Institute, Department of Pediatrics, and.
  • Howell AJ; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA.
  • Yang F; Darby Children's Research Institute, Department of Pediatrics, and.
  • Ban Y; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA.
  • McCrea HJ; Department of Public Health Sciences, and.
  • Zindy F; Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, USA.
  • Danis E; Departments of Neurological Surgery and Pediatrics, University of Miami, Jackson Health System, Miller School of Medicine, Miami, Florida, USA.
  • Vibhakar R; Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
  • Jermakowicz A; University of Colorado Cancer Center.
  • Martin V; Department of Biomedical Informatics, and.
  • Coss CC; Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
  • Harris BT; Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, USA.
  • de Cubas A; Department of Morphology and Cell Biology, University of Oviedo, Oviedo, Asturias, Spain.
  • Chen XS; College of Pharmacy, The Ohio State University, Columbus, Ohio, USA.
  • Barnoud T; Departments of Neurology and Pathology, Georgetown University Medical Center, Washington DC, USA.
  • Roussel MF; Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA.
  • Ayad NG; Department of Microbiology and Immunology, and.
  • Robbins DJ; Department of Public Health Sciences, and.
J Clin Invest ; 134(15)2024 Jun 17.
Article in En | MEDLINE | ID: mdl-38885332
ABSTRACT
Most children with medulloblastoma (MB) achieve remission, but some face very aggressive metastatic tumors. Their dismal outcome highlights the critical need to advance therapeutic approaches that benefit such high-risk patients. Minnelide, a clinically relevant analog of the natural product triptolide, has oncostatic activity in both preclinical and early clinical settings. Despite its efficacy and tolerable toxicity, this compound has not been evaluated in MB. Utilizing a bioinformatic data set that integrates cellular drug response data with gene expression, we predicted that Group 3 (G3) MB, which has a poor 5-year survival, would be sensitive to triptolide/Minnelide. We subsequently showed that both triptolide and Minnelide attenuate the viability of G3 MB cells ex vivo. Transcriptomic analyses identified MYC signaling, a pathologically relevant driver of G3 MB, as a downstream target of this class of drugs. We validated this MYC dependency in G3 MB cells and showed that triptolide exerts its efficacy by reducing both MYC transcription and MYC protein stability. Importantly, Minnelide acted on MYC to reduce tumor growth and leptomeningeal spread, which resulted in improved survival of G3 MB animal models. Moreover, Minnelide improved the efficacy of adjuvant chemotherapy, further highlighting its potential for the treatment of MYC-driven G3 MB.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Phenanthrenes / Proto-Oncogene Proteins c-myc / Xenograft Model Antitumor Assays / Diterpenes / Epoxy Compounds / Medulloblastoma Limits: Animals / Female / Humans Language: En Journal: J Clin Invest Year: 2024 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Phenanthrenes / Proto-Oncogene Proteins c-myc / Xenograft Model Antitumor Assays / Diterpenes / Epoxy Compounds / Medulloblastoma Limits: Animals / Female / Humans Language: En Journal: J Clin Invest Year: 2024 Document type: Article