The overall objective of this project is to design, synthesize and further develop several series of small-molecule drugs that have the common feature of being selectively toxic to hypoxic cells. This approach is based on the fact that hypoxia is much more severe in solid tumors than normal tissue. We will explore several approaches to the development of such drugs, which have the promise of being less toxic than current cytotoxic agents to normal tissue, and therefore having better therapeutic ratios. We will follow up progress made in the current program in defining optimal physicochemical properties (solubility, lipophilicity, reduction potential, radical stability, DNA targeting) of analogs of the hypoxia-activated prodrug tirapazamine now in Phase III clinical trials. In particular, we will prepare and explore the utility of 3-substituted analogs to optimize many of these properties. We will prepare and characterize novel prodrugs of potent alkylating agents, designed for activation by nitroreductase, beta-glucuronidase and beta-galactosidase enzymes isolated from the bacteria Escherischia coli and from Clostridium species. The genes for these enzymes will be delivered selectively to hypoxic tumor cells by the anaerobic bacterium Clostridia sporogenes, in a therapy approach called Clostridia-directed enzyme-prodrug therapy (CDEPT). We will explore the positioning of the cytotoxin and trigger units within the prodrug to optimize prodrug/enzyme interactions, and will determine the appropriate lipophilicities of the prodrugs and their released effectors for optimum extravascular diffusion, in developing prodrugs for this exciting new vector delivery system. We will study compounds that are lethal to cells with upregulated activity of hypoxia-induced transcription factor 1alpha (HIF-1alpha). This is an important component of the pathway by which cells adapt to low-oxygen environments, and drugs that are preferentially cytotoxic in its presence or in the presence of downstream enzymes upregulated by it, are expected to be selectively toxic to hypoxic cells as well as to aerobic cells over-expressing HIF-1alpha. We will evaluate hits in terms of their potential development as therapeutic drugs (e.g., solubility, diffusional properties, ease of synthesis, novelty). We will then prepare small series of analogs of selected lead small molecules, to identify classes suitable for advanced development.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
2P01CA082566-04A1
Application #
6985585
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
$141,732
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
Chan, Denise A; Giaccia, Amato J (2008) Targeting cancer cells by synthetic lethality: autophagy and VHL in cancer therapeutics. Cell Cycle 7:2987-90

Showing the most recent 10 out of 41 publications