The overall objective of this project are to design, synthesize, and determine relevant physicochemical properties for, new analogs of the benzotriazine-di-N-oxides (tirapazamine; TPZ) and related quinoxaline- di-N- oxides (QNOs), seeking compounds in this class with superior anti- tumor efficacy than the clinical drug tirapazamine itself. An initial set of analogs with widely varying lipophilic, electronic and steric properties will be designed and prepared (by carefully varying the ring substituents). In addition to substituent parameters, we will also measure a number of global physicochemical properties, including lipophilicity, one-electron reduction potential and the lifetime of the critical radical species (the latter two by pulse radiolysis). The activities of these analogs for the nuclear matrix enzyme complex (that results in drug activation) and the mitochondrial enzyme complex (considered to give rise to the major toxic side effects of tirapazamine) will be determined by Project 2. The results of these assays will be used to determine (by techniques such as multiple linear regression, principal component analysis, etc) structure-activity relationships (SAR) that will be used in subsequent syntheses to develop compounds with optimal patterns of enzyme substrate properties to minimize toxicity and maximize activity. The analogs will also be screened in Project 2 for hypoxic cell selectivity. These additional biological assays in Projects 2 and 3. The results from these assays will decide, through a detailed decision tree, whether compounds progress to further testing, and will also provide direct feedback for new drug design. A second major hypothesis to be tested is that DNA-targeting of TPZ analogs (by the attachment of DNA-affinic intercalating or minor groove binding carriers) will provide compounds with superior in vivo activity because of targeting the reactive unit to the activating nuclear enzyme complex ( and away from the mitochondrial enzyme complex). Project 3 will use selected of these compounds to determine the features required to ensure efficient delivery to hypoxic tumor cells in vivo. A third hypothesis to be tested is that N-oxide pro-drug forms of the DNA- targeting units will provide analogs with good distribution properties. We will also evaluate known methods of preparing BTO compounds, and will adapt these to the preparation of analogs of the above types. In both cases, we will explore the use of parallel array synthesis from common precursors from suitable.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Program Projects (P01)
Project #
1P01CA082566-01A1
Application #
6404065
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
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