Your browser doesn't support javascript.
loading
KP372-1-Induced AKT Hyperactivation Blocks DNA Repair to Synergize With PARP Inhibitor Rucaparib via Inhibiting FOXO3a/GADD45α Pathway.
Jiang, Lingxiang; Liu, Yingchun; Su, Xiaolin; Wang, Jiangwei; Zhao, Ye; Tumbath, Soumya; Kilgore, Jessica A; Williams, Noelle S; Chen, Yaomin; Wang, Xiaolei; Mendonca, Marc S; Lu, Tao; Fu, Yang-Xin; Huang, Xiumei.
Afiliación
  • Jiang L; Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Liu Y; Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Su X; Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Sciences, Fujian Medical University, Fujian, China.
  • Wang J; Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Zhao Y; Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Tumbath S; Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Kilgore JA; Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Williams NS; Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States.
  • Chen Y; Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States.
  • Wang X; Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Mendonca MS; State Key Laboratory of Applied Organic Chemistry, Department of Chemistry, Lanzhou University, Lanzhou, China.
  • Lu T; Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Fu YX; Department of Pharmacology and Toxicology, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
  • Huang X; Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
Front Oncol ; 12: 976292, 2022.
Article en En | MEDLINE | ID: mdl-36203459
ABSTRACT
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) have exhibited great promise in the treatment of tumors with homologous recombination (HR) deficiency, however, PARPi resistance, which ultimately recovers DNA repair and cell progress, has become an enormous clinical challenge. Recently, KP372-1 was identified as a novel potential anticancer agent that targeted the redox enzyme, NAD(P)Hquinone oxidoreductase 1 (NQO1), to induce extensive reactive oxygen species (ROS) generation that amplified DNA damage, leading to cancer cell death. To overcome PARPi resistance and expand its therapeutic utility, we investigated whether a combination therapy of a sublethal dose of KP372-1 with a nontoxic dose of PARPi rucaparib would synergize and enhance lethality in NQO1 over-expressing cancers. We reported that the combination treatment of KP372-1 and rucaparib induced a transient and dramatic AKT hyperactivation that inhibited DNA repair by regulating FOXO3a/GADD45α pathway, which enhanced PARPi lethality and overcame PARPi resistance. We further found that PARP inhibition blocked KP372-1-induced PARP1 hyperactivation to reverse NAD+/ATP loss that promoted Ca2+-dependent autophagy and apoptosis. Moreover, pretreatment of cells with BAPTA-AM, a cytosolic Ca2+ chelator, dramatically rescued KP372-1- or combination treatment-induced lethality and significantly suppressed PAR formation and γH2AX activation. Finally, we demonstrated that this combination therapy enhanced accumulation of both agents in mouse tumor tissues and synergistically suppressed tumor growth in orthotopic pancreatic and non-small-cell lung cancer xenograft models. Together, our study provides novel preclinical evidence for new combination therapy in NQO1+ solid tumors that may broaden the clinical utility of PARPi.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Front Oncol Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Idioma: En Revista: Front Oncol Año: 2022 Tipo del documento: Article País de afiliación: Estados Unidos