The overall goal in this PO1 renewal is to develop new anticancer drugs that are active specifically in solid tumors. In this application we propose to accomplish this by exploiting a major difference between normal and malignant tissues, namely, the presence in human solid tumors of regions of very low oxygenation, which are often associated with necrosis. Although this lower oxygenation is currently a negative prognostic factor for radiotherapy, for chemotherapy, and for malignant progression, it also provides a potentially powerful way to activate cytotoxic anticancer drugs exclusively in tumors. We propose three different strategies for exploiting these hypoxic regions in solid tumors to develop tumor-specific anticancer drugs. All three depend on the activation within hypoxic regions of nontoxic prodrugs into toxic drugs but by different mechanisms. One project will design, synthesize, and provide the necessary physicochemical evaluation of the drugs for each of the three strategies. A second project will evaluate prodrugs activated to cytotoxic agents by the nonpathogenic obligate anaerobe C. sporogenes genetically engineered to express specific prodrug-activating enzymes. These anaerobes colonize the necrotic areas of solid tumors, express the prodrug-activating enzymes in these regions, and consequently activate systemically delivered prodrugs to the cytotoxic drugs specifically in the tumors. A third project will continue our mechanism-based development of a superior analog of the clinically active hypoxic cytotoxin tirapazamine (TPZ), which was developed by us and which is showing success in clinical trials. The rational development of an improved TPZ analog depends on the extensive knowledge we have gained in understanding the properties of drugs affecting their in vivo activity. A fourth project is designed to exploit the fact that hypoxic cells in tumors have elevated levels and activity of the transcription factor HIF-1, which transactivates many genes that become specifically expressed in the hypoxic regions. We will screen for drugs that are conditionally lethal to cells withhigh HIF-1 activity. An important advantage of our strategy is that though we are targeting tumor cells in general, we expect the maximum cell killing to be against the hypoxic cells of the tumor, which are known to be those resistant to standard therapy.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
2P01CA082566-04A1
Application #
6765648
Study Section
Subcommittee G - Education (NCI)
Program Officer
Stone, Helen B
Project Start
2000-04-25
Project End
2009-03-31
Budget Start
2004-04-01
Budget End
2005-03-31
Support Year
4
Fiscal Year
2004
Total Cost
$1,049,012
Indirect Cost
Name
Stanford University
Department
Radiation-Diagnostic/Oncology
Type
Schools of Medicine
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