This Project is one of three that together constitute a coordinated and comprehensive Program for the development of new bioreductive drugs for the exploitation of hypoxic cells in tumors. The approaches focuses on analogs of tirapazamine (TPZ), a drug with proven clinical ability but which does not fully exploit the potential of this new class of anti-cancer agents. The broad objective of the present Project is to develop a detailed understanding of the features of TPZ-like drugs that determine delivery to hypoxic tumor cells (and normal tissues) in vivo, and to use this information to develop analogs with superior utility. The hypothesis and approaches are: (1) The rate of overall bioreductive drug metabolism, and extent of entrapment in cells, determine the ability of TPZ analogs to penetrate hypoxic tumor tissue. Drug metabolism and uptake will be quantified in single cell suspensions, at a range of O2 concentrations. Tissue diffusion properties will be investigated using a novel tissue culture model, multi- cellular layers (MCL), developed in this laboratory. (2) Optimization of the ability of TPZ analogs to penetrate hypoxic tumor tissue will improve therapeutic efficacy of combining the analogs with radiation and cisplatin. Plasma pharmacokinetic parameters will be measured, and used along with in vitro cytotoxicity (Project 2) and penetration data (MCL studies) to predict activity against hypoxic cells. The model will be tested against measured cell killing in combination with radiation and cisplatin. (3) Physiology-based, high-throughput screens, designed to evaluate delivery to hypoxic cells, will facilitate optimization of bioreductive drug design. These studies will exploit the selectivity of liquid chromatography/mass spectroscopy (LC/MS) for simultaneous evaluation of MCL penetration of multiple compounds. (4) Alteration of physicochemical properties (especially log P) and/or reductase specificity can be used to reduce retinal toxicity of TPZ analogs relative to anti-tumor efficacy. We will measure retinal damage with compounds having a wide range of lipophilicities and enzyme substrate specificities. We will perform experiments to understand the mechanism of this toxicity.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
1P01CA082566-01A1
Application #
6404069
Study Section
Subcommittee E - Prevention &Control (NCI)
Project Start
2000-04-25
Project End
2003-03-31
Budget Start
Budget End
Support Year
1
Fiscal Year
2000
Total Cost
Indirect Cost
Name
Stanford University
Department
Type
DUNS #
800771545
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
Song, Suisui; McCann, Kelly E; Brown, J Martin (2008) Radiosensitization of yeast cells by inhibition of histone h4 acetylation. Radiat Res 170:618-27

Showing the most recent 10 out of 41 publications