Your browser doesn't support javascript.
loading
Antitumor Synergism and Enhanced Survival with a Tumor Vasculature-Targeted Enzyme Prodrug System, Rapamycin, and Cyclophosphamide.
Krais, John J; Virani, Needa; McKernan, Patrick H; Nguyen, Quang; Fung, Kar-Ming; Sikavitsas, Vassilios I; Kurkjian, Carla; Harrison, Roger G.
Afiliación
  • Krais JJ; School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma.
  • Virani N; School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma.
  • McKernan PH; School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma.
  • Nguyen Q; School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma.
  • Fung KM; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
  • Sikavitsas VI; School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma.
  • Kurkjian C; Oncology/Hematology Section, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
  • Harrison RG; School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma. rharrison@ou.edu.
Mol Cancer Ther ; 16(9): 1855-1865, 2017 09.
Article en En | MEDLINE | ID: mdl-28522586
ABSTRACT
Mutant cystathionine gamma-lyase was targeted to phosphatidylserine exposed on tumor vasculature through fusion with Annexin A1 or Annexin A5. Cystathionine gamma-lyase E58N, R118L, and E338N mutations impart nonnative methionine gamma-lyase activity, resulting in tumor-localized generation of highly toxic methylselenol upon systemic administration of nontoxic selenomethionine. The described therapeutic system circumvents systemic toxicity issues using a novel drug delivery/generation approach and avoids the administration of nonnative proteins and/or DNA required with other enzyme prodrug systems. The enzyme fusion exhibits strong and stable in vitro binding with dissociation constants in the nanomolar range for both human and mouse breast cancer cells and in a cell model of tumor vascular endothelium. Daily administration of the therapy suppressed growth of highly aggressive triple-negative murine 4T1 mammary tumors in immunocompetent BALB/cJ mice and MDA-MB-231 tumors in SCID mice. Treatment did not result in the occurrence of negative side effects or the elicitation of neutralizing antibodies. On the basis of the vasculature-targeted nature of the therapy, combinations with rapamycin and cyclophosphamide were evaluated. Rapamycin, an mTOR inhibitor, reduces the prosurvival signaling of cells in a hypoxic environment potentially exacerbated by a vasculature-targeted therapy. IHC revealed, unsurprisingly, a significant hypoxic response (increase in hypoxia-inducible factor 1 α subunit, HIF1A) in the enzyme prodrug-treated tumors and a dramatic reduction of HIF1A upon rapamycin treatment. Cyclophosphamide, an immunomodulator at low doses, was combined with the enzyme prodrug therapy and rapamycin; this combination synergistically reduced tumor volumes, inhibited metastatic progression, and enhanced survival. Mol Cancer Ther; 16(9); 1855-65. ©2017 AACR.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Profármacos / Sirolimus / Ciclofosfamida / Neoplasias / Neovascularización Patológica Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: Mol Cancer Ther Asunto de la revista: ANTINEOPLASICOS Año: 2017 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Profármacos / Sirolimus / Ciclofosfamida / Neoplasias / Neovascularización Patológica Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans Idioma: En Revista: Mol Cancer Ther Asunto de la revista: ANTINEOPLASICOS Año: 2017 Tipo del documento: Article