1. Academic Validation
  2. Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery

Tumor radiosensitization by monomethyl auristatin E: mechanism of action and targeted delivery

  • Cancer Res. 2015 Apr 1;75(7):1376-1387. doi: 10.1158/0008-5472.CAN-14-1931.
Lisa Buckel  # 1 Elamprakash N Savariar  # 2 Jessica L Crisp 2 Karra A Jones 3 Angel M Hicks 1 Daniel J Scanderbeg 1 Quyen T Nguyen 4 Jason K Sicklick 5 Andrew M Lowy 5 Roger Y Tsien 2 6 Sunil J Advani 1 7
Affiliations

Affiliations

  • 1 Department of Radiation Medicine and Applied Sciences.
  • 2 Department of Pharmacology.
  • 3 Department of Pathology Department of Surgery.
  • 4 Division of Head and Neck Surgery.
  • 5 Division of Surgical Oncology.
  • 6 Howard Hughes Medical Institute.
  • 7 Center for Advanced Radiotherapy Technologies University of California San Diego.
  • # Contributed equally.
Abstract

Intrinsic tumor resistance to radiotherapy limits the efficacy of ionizing radiation (IR). Sensitizing Cancer cells specifically to IR would improve tumor control and decrease normal tissue toxicity. The development of tumor-targeting technologies allows for developing potent radiosensitizing drugs. We hypothesized that the anti-tubulin agent monomethyl Auristatin E (MMAE), a component of a clinically approved antibody-directed conjugate, could function as a potent radiosensitizer and be selectively delivered to tumors using an activatable cell-penetrating peptide targeting Matrix Metalloproteinases and RGD-binding integrins (ACPP-cRGD-MMAE). We evaluated the ability of MMAE to radiosensitize both established Cancer cells and a low-passage cultured human pancreatic tumor cell line using clonogenic and DNA damage assays. MMAE sensitized colorectal and pancreatic Cancer cells to IR in a schedule- and dose-dependent manner, correlating with mitotic arrest. Radiosensitization was evidenced by decreased clonogenic survival and increased DNA double-strand breaks in irradiated cells treated with MMAE. MMAE in combination with IR resulted in increased DNA damage signaling and activation of Chk1. To test a therapeutic strategy of MMAE and IR, PANC-1 or HCT-116 murine tumor xenografts were treated with nontargeted free MMAE or tumor-targeted MMAE (ACPP-cRGD-MMAE). While free MMAE in combination with IR resulted in tumor growth delay, tumor-targeted ACPP-cRGD-MMAE with IR produced a more robust and significantly prolonged tumor regression in xenograft models. Our studies identify MMAE as a potent radiosensitizer. Importantly, MMAE radiosensitization can be localized to tumors by targeted activatable cell-penetrating Peptides.

Figures
Products