Your browser doesn't support javascript.
loading
Targeted Drug Delivery with an Integrin-Binding Knottin-Fc-MMAF Conjugate Produced by Cell-Free Protein Synthesis.
Currier, Nicolas V; Ackerman, Shelley E; Kintzing, James R; Chen, Rishard; Filsinger Interrante, Maria; Steiner, Alexander; Sato, Aaron K; Cochran, Jennifer R.
Affiliation
  • Currier NV; Division of Pediatric Hematology/Oncology, Stanford Medical School, Stanford, California.
  • Ackerman SE; Department of Bioengineering, Stanford University, Stanford, California.
  • Kintzing JR; Department of Bioengineering, Stanford University, Stanford, California.
  • Chen R; Sutro Biopharma, Inc., South San Francisco, California.
  • Filsinger Interrante M; Department of Bioengineering, Stanford University, Stanford, California.
  • Steiner A; Sutro Biopharma, Inc., South San Francisco, California.
  • Sato AK; Sutro Biopharma, Inc., South San Francisco, California.
  • Cochran JR; Department of Bioengineering, Stanford University, Stanford, California. Department of Chemical Engineering, Stanford University, Stanford, California. jennifer.cochran@stanford.edu.
Mol Cancer Ther ; 15(6): 1291-300, 2016 06.
Article in En | MEDLINE | ID: mdl-27197305
ABSTRACT
Antibody-drug conjugates (ADC) have generated significant interest as targeted therapeutics for cancer treatment, demonstrating improved clinical efficacy and safety compared with systemic chemotherapy. To extend this concept to other tumor-targeting proteins, we conjugated the tubulin inhibitor monomethyl-auristatin-F (MMAF) to 2.5F-Fc, a fusion protein composed of a human Fc domain and a cystine knot (knottin) miniprotein engineered to bind with high affinity to tumor-associated integrin receptors. The broad expression of integrins (including αvß3, αvß5, and α5ß1) on tumor cells and their vasculature makes 2.5F-Fc an attractive tumor-targeting protein for drug delivery. We show that 2.5F-Fc can be expressed by cell-free protein synthesis, during which a non-natural amino acid was introduced into the Fc domain and subsequently used for site-specific conjugation of MMAF through a noncleavable linker. The resulting knottin-Fc-drug conjugate (KFDC), termed 2.5F-Fc-MMAF, had approximately 2 drugs attached per KFDC. 2.5F-Fc-MMAF inhibited proliferation in human glioblastoma (U87MG), ovarian (A2780), and breast (MB-468) cancer cells to a greater extent than 2.5F-Fc or MMAF alone or added in combination. As a single agent, 2.5F-Fc-MMAF was effective at inducing regression and prolonged survival in U87MG tumor xenograft models when administered at 10 mg/kg two times per week. In comparison, tumors treated with 2.5F-Fc or MMAF were nonresponsive, and treatment with a nontargeted control, CTRL-Fc-MMAF, showed a modest but not significant therapeutic effect. These studies provide proof-of-concept for further development of KFDCs as alternatives to ADCs for tumor targeting and drug delivery applications. Mol Cancer Ther; 15(6); 1291-300. ©2016 AACR.
Subject(s)

Full text: 1 Database: MEDLINE Main subject: Oligopeptides / Integrins / Immunoconjugates / Cystine-Knot Miniproteins / Neoplasms Limits: Animals / Humans Language: En Journal: Mol Cancer Ther Journal subject: ANTINEOPLASICOS Year: 2016 Type: Article

Full text: 1 Database: MEDLINE Main subject: Oligopeptides / Integrins / Immunoconjugates / Cystine-Knot Miniproteins / Neoplasms Limits: Animals / Humans Language: En Journal: Mol Cancer Ther Journal subject: ANTINEOPLASICOS Year: 2016 Type: Article