We have identified calcium influx as a molecular target in angiogenesis, in part using CAI, an inhibitor of nonvoltage-gated calcium influx. CAI inhibits endothelial cell proliferation, motility, collagenase expression, and regulates adhesion and spreading on matrix. We previously implicated the FAK, RhoA, and RhoGAP pathways in this function. We have now demonstrated that CAI regulates VEGF production. CAI regulates VEGF expression and protein production under normoxic and relative hypoxic conditions with a greater effect when stressed by acidosis in melanoma cells. One mechanisms for this may be through recently observed CAI regulation of HIF-1a expression. Reduction in VEGF expression was also observed in small cell lung cancer; CAI was both anti-tumor and anti-proliferative in a small cell lung cancer xenograft model. CAI also regulates response to VEGF by reducing endothelial migration to VEGF as an attractant. These results indicate that as a signal transduction regulator, CAI targets both tumor and stromal elements. In order to further dissect the pathways regulating angiogenesis, two new approaches have been initiated: cDNA microarray and microproteomic pathway profiling. In vitro models of vascular development have been used. cDNA microarray analysis showed little gene expression differences occurred in angiogenesis-related genes during vascular tube formation, including genes in the categories of invasion, proliferation, and signaling. Caldesmon was found to be reduced in gene expression early in tube formation, and reduced at the protein level later in tube formation. It interacts with the RhoA pathway and may be a downstream target of calcium influx regulation. Microproteomics is being applied to profile the signaling pathways driving neovascular events. Wound healing, tubulogenesis, and in vivo vascularization assays are being optimized for application to microdissection and protein array analysis. The effect of signal modulatory agents, such as CAI and 2-methoxyestradiol will be applied. Translational application of these assays will be investigated in upcoming clinical trials of 2-methoxyestradiol, STI-571, and an EGF receptor inhibitor.

Agency
National Institute of Health (NIH)
Institute
Division of Clinical Sciences - NCI (NCI)
Type
Intramural Research (Z01)
Project #
1Z01SC009374-10
Application #
6558536
Study Section
(LP)
Project Start
Project End
Budget Start
Budget End
Support Year
10
Fiscal Year
2001
Total Cost
Indirect Cost
Name
Clinical Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Hoskins, Ebony; Rodriguez-Canales, Jaime; Hewitt, Stephen M et al. (2011) Paracrine SLPI secretion upregulates MMP-9 transcription and secretion in ovarian cancer cells. Gynecol Oncol 122:656-62
Balkwill, Frances R; Ashworth, Alan; Bast, Robert C et al. (2006) 10th Biennial Helene Harris Memorial Trust meeting. Cancer Res 66:2904-6
Alessandro, Riccardo; Di Bella, Maria Antonietta; Flugy, Anna Maria et al. (2006) Comparative study of T84 and T84SF human colon carcinoma cells: in vitro and in vivo ultrastructural and functional characterization of cell culture and metastasis. Virchows Arch 449:48-61
Davidson, Ben; Espina, Virginia; Steinberg, Seth M et al. (2006) Proteomic analysis of malignant ovarian cancer effusions as a tool for biologic and prognostic profiling. Clin Cancer Res 12:791-9
Azad, Nilofer S; Rasool, Nabila; Annunziata, Christina M et al. (2006) Proteomics in clinical trials and practice: present uses and future promise. Mol Cell Proteomics 5:1819-29
Kassis, Jareer N; Guancial, Elizabeth A; Doong, Howard et al. (2006) CAIR-1/BAG-3 modulates cell adhesion and migration by downregulating activity of focal adhesion proteins. Exp Cell Res 312:2962-71
Stevens, Ellen V; Raffeld, Mark; Espina, Virginia et al. (2005) Expression of xeroderma pigmentosum A protein predicts improved outcome in metastatic ovarian carcinoma. Cancer 103:2313-9
Kassis, Jareer; Klominek, Julius; Kohn, Elise C (2005) Tumor microenvironment: what can effusions teach us? Diagn Cytopathol 33:316-9
Perabo, Frank G E; Demant, Andre W; Wirger, Andreas et al. (2005) Carboxyamido-triazole (CAI) reverses the balance between proliferation and apoptosis in a rat bladder cancer model. Anticancer Res 25:725-9
Winters, Mary E; Mehta, Arpita I; Petricoin 3rd, Emanuel F et al. (2005) Supra-additive growth inhibition by a celecoxib analogue and carboxyamido-triazole is primarily mediated through apoptosis. Cancer Res 65:3853-60

Showing the most recent 10 out of 22 publications