The goal of this project is to develop new prodrugs that become activated to cytotoxic anticancer drugs exclusively in solid tumors. These nontoxic prodrugs will be enzymatically activated to cytotoxic agents by the nonpathogenic obligate anaerobe C. sporogenes genetically engineered to express the prodrug-activating enzymes. When spores of these genetically engineered C. sporogenes are injected intravenously the spores germinate exclusively in the hypoxic necrotic areas of tumors and produce high levels of the enzyme exclusively in these regions. Such hypoxic/necrotic regions are extremely common in human solid tumors and provide the basis for the selective expression of the prodrug-activating enzymes in the tumors. When a nontoxic prodrug is administered systemically following the intravenous injection of the genetically engineered bacterial spores, conversion to the cytotoxic metabolite occurs exclusively in the tumor. We have demonstrated proof-of-principle of this CDEPT (clostridia-directed enzyme prodrug therapy) approach with the E. coli derived enzyme cytosine deaminase which converts the nontoxic drug 5-fluorocytosine to the anticancer drug 5-fluorouracil, and with E. coli derived nitroreductase which converts the dinitrobenzamide prodrug CB 1954 to a toxic bifunctional alkylating agent. We propose to evaluate newly synthesized prodrugs with three bacterial enzymes: nitroreductase, beta-glucuronidase, and beta-galactosidase in this system. For each of these enzymes we will test prodrugs designed and synthesized for their conversion to the toxic drug, for their ability to diffuse through a multicellular layer and for their antitumor efficacy, both alone and in combination with single doses and fractionated irradiation. The combination with radiation will test the hypothesis that there will be greater cell kill by the activated prodrugs of the hypoxic compared to the aerobic tumor cells. We will also combine this approach with two vascular-targeting drugs, the TNF-inducing drug DMXAA and the tubulin-binding agent ZD6126, both of which decrease blood flow and increase necrosis in experimental and human tumors. We hypothesize that these vascular targeting agents will sensitize very small tumors (lung metastases) to CDEPT because of their ability to induce necrosis in such tumors.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
2P01CA082566-04A1
Application #
6985592
Study Section
Subcommittee G - Education (NCI)
Project Start
2004-04-01
Project End
2009-03-31
Budget Start
2004-04-01
Budget End
2005-03-31
Support Year
4
Fiscal Year
2004
Total Cost
$125,742
Indirect Cost
Name
Stanford University
Department
Type
DUNS #
009214214
City
Stanford
State
CA
Country
United States
Zip Code
94305
Brown, Martin; Bernhard, Eric; Mitchel, James et al. (2016) Fractionated Radiation for Newly Diagnosed Supratentorial Glioblastoma Multiforme: In Regard to Brachman et al. Int J Radiat Oncol Biol Phys 94:210-211
Bonnet, Muriel; Flanagan, Jack U; Chan, Denise A et al. (2014) Identifying novel targets in renal cell carcinoma: design and synthesis of affinity chromatography reagents. Bioorg Med Chem 22:711-20
Chan, Denise A; Sutphin, Patrick D; Nguyen, Phuong et al. (2011) Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci Transl Med 3:94ra70
Bonnet, Muriel; Flanagan, Jack U; Chan, Denise A et al. (2011) SAR studies of 4-pyridyl heterocyclic anilines that selectively induce autophagic cell death in von Hippel-Lindau-deficient renal cell carcinoma cells. Bioorg Med Chem 19:3347-56
Hicks, Kevin O; Siim, Bronwyn G; Jaiswal, Jagdish K et al. (2010) Pharmacokinetic/pharmacodynamic modeling identifies SN30000 and SN29751 as tirapazamine analogues with improved tissue penetration and hypoxic cell killing in tumors. Clin Cancer Res 16:4946-57
Hay, Michael P; Turcotte, Sandra; Flanagan, Jack U et al. (2010) 4-Pyridylanilinothiazoles that selectively target von Hippel-Lindau deficient renal cell carcinoma cells by inducing autophagic cell death. J Med Chem 53:787-97
Turcotte, Sandra; Giaccia, Amato J (2010) Targeting cancer cells through autophagy for anticancer therapy. Curr Opin Cell Biol 22:246-51
Brown, Martin (2010) Henry S. Kaplan Distinguished Scientist Award Lecture 2007. The remarkable yin and yang of tumour hypoxia. Int J Radiat Biol 86:907-17
Shinde, Sujata S; Maroz, Andrej; Hay, Michael P et al. (2009) One-electron reduction potential of the neutral guanyl radical in the GC base pair of duplex DNA. J Am Chem Soc 131:5203-7
Turcotte, Sandra; Sutphin, Patrick D; Giaccia, Amato J (2008) Targeted therapy for the loss of von Hippel-Lindau in renal cell carcinoma: a novel molecule that induces autophagic cell death. Autophagy 4:944-6

Showing the most recent 10 out of 41 publications