Fatty acid synthase (FAS), the enzyme that synthesizes the fatty acid palmitate, is overexpressed in prostate cancer and many other cancers of epithelial origin. We have discovered that orlistat, an FDA approved drug, is a novel inhibitor of the thioesterase (TE) domain of FAS. Inhibition of FAS by orlistat results in the selective killing of tumor cells in vitro and in vivo. The goal of this project is to determine the basic cellular and biochemical mechanisms of the anti-tumor effects of orlistat and other FAS inhibitors. To this end we have demonstrated that the endoplasmic reticulum (ER) stress response is activated in tumor cells upon FAS inhibitor treatment. Activation of the ER stress response appears to be upstream of apoptosis, perhaps engaging the cell death program. Moreover, the combination of FAS inhibitors with thapsigargin, another agent known to activate ER stress, yields a synergistic decrease in tumor cell survival. We have also determined the crystal structure of the TE domain (FAS-TE) in complex with orlistat. Orlistat binds to the active site in a manner contrary to a previously proposed model of substrate binding.
Three specific aims are proposed to further explore these observations and to provide a critical foundation for the design and optimization of FAS inhibitors for cancer therapy.
In Specific Aim 1 we will use tumor cell lines and transformed mouse embryonic fibroblasts (MEFs) with pathway specific mutations to determine the contribution of the PERK, IRE1 and ATF6 signaling pathways to the ER stress response when FAS is inhibited.
In Specific Aim 2 the ability of thapsigargin and hypoxia to enhance the cytotoxic effects of FAS inhibitors will be determined. An in vivo imaging system will also be used to monitor ER stress-driven luciferase activity in tumor xenografts of mice treated with FAS inhibitors.
In Specific Aim 3 we will use X-ray crystallography to determine the structural basis for the interactions between FAS-TE and orlistat and the orlistat analog Ebelactone B. We will also test whether substrate binds in a similar manner to orlistat by determining the crystal structures of substrate and product and analyzing the activity of site-directed mutants of conserved residues within the binding groove using a new, continuous assay. These studies will provide invaluable insights into how orlistat and other FAS inhibitors initiate apoptosis and how these compounds may be useful to increase the efficacy of current cancer drugs, particularly those where resistance has occurred. The proposed crystal structures will be instrumental to the rational design of analogs of orlistat with improved target specificity, bioavailability, and pharmacokinetics for the treatment of a variety of cancers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA114104-05
Application #
7895851
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Forry, Suzanne L
Project Start
2006-09-01
Project End
2011-07-31
Budget Start
2010-08-01
Budget End
2011-07-31
Support Year
5
Fiscal Year
2010
Total Cost
$247,327
Indirect Cost
Name
Wake Forest University Health Sciences
Department
Biology
Type
Schools of Medicine
DUNS #
937727907
City
Winston-Salem
State
NC
Country
United States
Zip Code
27157
Ritchie, Melissa K; Johnson, Lynnette C; Clodfelter, Jill E et al. (2016) Crystal Structure and Substrate Specificity of Human Thioesterase 2: INSIGHTS INTO THE MOLECULAR BASIS FOR THE MODULATION OF FATTY ACID SYNTHASE. J Biol Chem 291:3520-30
Deford-Watts, Laura M; Mintz, Akiva; Kridel, Steven J (2013) The potential of ¹¹C-acetate PET for monitoring the Fatty acid synthesis pathway in Tumors. Curr Pharm Biotechnol 14:300-12
Scott, Kristen E N; Wheeler, Frances B; Davis, Amanda L et al. (2012) Metabolic regulation of invadopodia and invasion by acetyl-CoA carboxylase 1 and de novo lipogenesis. PLoS One 7:e29761
Bowlby, Sarah C; Thomas, Michael J; D'Agostino Jr, Ralph B et al. (2012) Nicotinamide phosphoribosyl transferase (Nampt) is required for de novo lipogenesis in tumor cells. PLoS One 7:e40195
Little, Joy L; Wheeler, Frances B; Koumenis, Constantinos et al. (2008) Disruption of crosstalk between the fatty acid synthesis and proteasome pathways enhances unfolded protein response signaling and cell death. Mol Cancer Ther 7:3816-24
Vavere, Amy L; Kridel, Steven J; Wheeler, Frances B et al. (2008) 1-11C-acetate as a PET radiopharmaceutical for imaging fatty acid synthase expression in prostate cancer. J Nucl Med 49:327-34
Little, Joy L; Kridel, Steven J (2008) Fatty acid synthase activity in tumor cells. Subcell Biochem 49:169-94
Kridel, Steven J; Lowther, W Todd; Pemble 4th, Charles W (2007) Fatty acid synthase inhibitors: new directions for oncology. Expert Opin Investig Drugs 16:1817-29
Chen, Yong Q; Edwards, Iris J; Kridel, Steven J et al. (2007) Dietary fat-gene interactions in cancer. Cancer Metastasis Rev 26:535-51
Pemble 4th, Charles W; Johnson, Lynnette C; Kridel, Steven J et al. (2007) Crystal structure of the thioesterase domain of human fatty acid synthase inhibited by Orlistat. Nat Struct Mol Biol 14:704-9