Your browser doesn't support javascript.
loading
U1 Adaptors Suppress the KRAS-MYC Oncogenic Axis in Human Pancreatic Cancer Xenografts.
Tsang, Ashley T; Dudgeon, Crissy; Yi, Lan; Yu, Xin; Goraczniak, Rafal; Donohue, Kristen; Kogan, Samuel; Brenneman, Mark A; Ho, Eric S; Gunderson, Samuel I; Carpizo, Darren R.
Afiliação
  • Tsang AT; Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
  • Dudgeon C; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
  • Yi L; Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
  • Yu X; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
  • Goraczniak R; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
  • Donohue K; Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
  • Kogan S; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
  • Brenneman MA; Silagene Inc., Hillsborough, New Jersey.
  • Ho ES; Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
  • Gunderson SI; Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey.
  • Carpizo DR; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.
Mol Cancer Ther ; 16(8): 1445-1455, 2017 08.
Article em En | MEDLINE | ID: mdl-28377488
ABSTRACT
Targeting KRAS and MYC has been a tremendous challenge in cancer drug development. Genetic studies in mouse models have validated the efficacy of silencing expression of both KRAS and MYC in mutant KRAS-driven tumors. We investigated the therapeutic potential of a new oligonucleotide-mediated gene silencing technology (U1 Adaptor) targeting KRAS and MYC in pancreatic cancer. Nanoparticles in complex with anti-KRAS U1 Adaptors (U1-KRAS) showed remarkable inhibition of KRAS in different human pancreatic cancer cell lines in vitro and in vivo As a nanoparticle-free approach is far easier to develop into a drug, we refined the formulation of U1 Adaptors by conjugating them to tumor-targeting peptides (iRGD and cRGD). Peptides coupled to fluorescently tagged U1 Adaptors showed selective tumor localization in vivo Efficacy experiments in pancreatic cancer xenograft models showed highly potent (>90%) antitumor activity of both iRGD and (cRGD)2-KRAS Adaptors. U1 Adaptors targeting MYC inhibited pancreatic cancer cell proliferation caused by apoptosis in vitro (40%-70%) and tumor regressions in vivo Comparison of iRGD-conjugated U1 KRAS and U1 MYC Adaptors in vivo revealed a significantly greater degree of cleaved caspase-3 staining and decreased Ki67 staining as compared with controls. There was no significant difference in efficacy between the U1 KRAS and U1 MYC Adaptor groups. Our results validate the value in targeting both KRAS and MYC in pancreatic cancer therapeutics and provide evidence that the U1 Adaptor technology can be successfully translated using a nanoparticle-free delivery system to target two undruggable genes in cancer. Mol Cancer Ther; 16(8); 1445-55. ©2017 AACR.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Oligonucleotídeos / Oncogenes / Neoplasias Pancreáticas / Proteínas Proto-Oncogênicas p21(ras) / Proteínas Proto-Oncogênicas c-myc / Ensaios Antitumorais Modelo de Xenoenxerto Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Oligonucleotídeos / Oncogenes / Neoplasias Pancreáticas / Proteínas Proto-Oncogênicas p21(ras) / Proteínas Proto-Oncogênicas c-myc / Ensaios Antitumorais Modelo de Xenoenxerto Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2017 Tipo de documento: Article