Matrix Metalloproteinases (MMPs) are a family of over 20 types of enzymes, both secreted and membrane-bound zinc endopeptidases, which collectively are capable of degrading all the components of the extracellular matrix. Studies suggest that MMPs are utilized in cancer, and facilitate both local tumor invasion and metastasis. MMP-2 and MMP-9 (also known as gelatinases), in particular, are thought to play critical roles in tumor cell invasion and are frequently coexpressed in human cancers. The goal of this study is to test a hypothesis regarding the ability to image by positron emission tomography (PET) the expression of MMP-2 and MMP-9 in a metastatic breast cancer tumor model using radiolabeled MMP-2 and MMP-9 peptide inhibitors. The hypothesis to be tested is that the accumulation of a radiolabeled gelatinase inhibitor will correlate with gelatinase enzyme activity determined in the tumor in an ex vivo assay. It has been demonstrated by Koivunen et al. that the synthetic cyclic peptide, CTTHWGFTLC (CTT), suppressed migration of breast cancer tumor cells in vitro and prevented the growth and invasion of MDA-MB-435 human breast cancer tumors in mice, suggesting that this peptide has potential as an anticancer agent. In preliminary experiments relating to this proposal, we conjugated CTT with the chelator DOTA (1,4,7, 10-tetraazacyclotetradecane-N,N',N"""""""",N'""""""""- tetraacetic acid) and initial in vitro assays demonstrated that DOTA-CTT inhibits MMP-2 activity comparably to the broad-range MMP inhibitor, Ilomastat. Preliminary microPET imaging and biodistribution studies in tumor-bearing mice show 64Cu-DOTA-CTT is taken up in three histologically different tumor types. Our preliminary data show that a 64Cu-labeled negative control peptide, DOTA-D-Trp-CTT does not show significant tumor uptake.
Our specific aims are as follows: 1) to optimize fluorogenic assays for determining inhibitory activity of MMP-2 and MMP-9 inhibitors and for determining MMP-2 and MMP-9 concentrations in tumors grown in vivo; 2) To confirm specific tumor uptake of 64Cu-DOTA-CTT vs a 64Cu-DOTA-conjngated control peptide in gelatinase-expressing tumor-bearing mouse models from biodistribution studies and/or microPET imaging data and to correlate uptake of 64Cu-DOTA-CTT with gelatinase expression in the tumor using an ex vivo assay; and 3) to synthesis a series of gelatinase inhibitors based upon the lead structure CTT that are conjugated to either DOTA or a cross-bridged macrocyclic chelator, as well as synthesize negative control peptides. If successful, the outcome of this study may be a PET tracer for determining the metastatic potential of various types of cancer.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Exploratory/Developmental Grants (R21)
Project #
5R21CA098698-02
Application #
6750629
Study Section
Diagnostic Radiology Study Section (RNM)
Program Officer
Menkens, Anne E
Project Start
2003-06-01
Project End
2006-05-31
Budget Start
2004-06-01
Budget End
2006-05-31
Support Year
2
Fiscal Year
2004
Total Cost
$191,250
Indirect Cost
Name
Washington University
Department
Radiation-Diagnostic/Oncology
Type
Schools of Medicine
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Wadas, Thaddeus J; Deng, Hongju; Sprague, Jennifer E et al. (2009) Targeting the alphavbeta3 integrin for small-animal PET/CT of osteolytic bone metastases. J Nucl Med 50:1873-80
Shokeen, Monica; Anderson, Carolyn J (2009) Molecular imaging of cancer with copper-64 radiopharmaceuticals and positron emission tomography (PET). Acc Chem Res 42:832-41
Sprague, Jennifer E; Kitaura, Hideki; Zou, Wei et al. (2007) Noninvasive imaging of osteoclasts in parathyroid hormone-induced osteolysis using a 64Cu-labeled RGD peptide. J Nucl Med 48:311-8
Sprague, Jennifer E; Li, Wen Ping; Liang, Kexian et al. (2006) In vitro and in vivo investigation of matrix metalloproteinase expression in metastatic tumor models. Nucl Med Biol 33:227-37
Li, W P; Anderson, C J (2003) Imaging matrix metalloproteinase expression in tumors. Q J Nucl Med 47:201-8