Distinct role of fibroblast growth factor-2 and vascular endothelial growth factor on tumor growth and angiogenesis

R Giavazzi, B Sennino, D Coltrini, A Garofalo… - The American journal of …, 2003 - Elsevier
R Giavazzi, B Sennino, D Coltrini, A Garofalo, R Dossi, R Ronca, MPM Tosatti, M Presta
The American journal of pathology, 2003Elsevier
Tumors express more than a single angiogenic growth factor. To investigate the relative
impact of fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF)
on tumor growth and neovascularization, we generated tumor cell transfectants differing for
VEGF and/or FGF-2 expression. Human endometrial adenocarcinoma HEC-1-B-derived Tet-
FGF-2 cells that express FGF-2 under the control of the tetracycline-responsive promoter
(Tet-off system) were further transfected with a VEGF121 anti-sense (AS-VEGF) cDNA. Next …
Tumors express more than a single angiogenic growth factor. To investigate the relative impact of fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF) on tumor growth and neovascularization, we generated tumor cell transfectants differing for VEGF and/or FGF-2 expression. Human endometrial adenocarcinoma HEC-1-B-derived Tet-FGF-2 cells that express FGF-2 under the control of the tetracycline-responsive promoter (Tet-off system) were further transfected with a VEGF121 anti-sense (AS-VEGF) cDNA. Next, Tet-FGF-2 and AS-VEGF/Tet-FGF-2 cells were transplanted subcutaneously in nude mice that received tetracycline or not in the drinking water. Simultaneous expression of FGF-2 and VEGF in Tet-FGF-2 cells resulted in fast-growing lesions characterized by high blood vessel density, patency and permeability, and limited necrosis. Blood vessels were highly heterogeneous in size and frequently associated with pericytes. Inhibition of FGF-2 production by tetracycline caused a significant decrease in tumor burden paralleled by a decrease in blood vessel density and size. AS-VEGF expression resulted in a similar reduction in blood vessel density associated with a significant decrease in pericyte organization, vascular patency, and permeability. The consequent decrease in tumor burden was paralleled by increased tumor hypoxia and necrosis. A limited additional inhibitory effect was exerted by simultaneous down-regulation of FGF-2 and VEGF expression. These findings demonstrate that FGF-2 and VEGF stimulate vascularization synergistically but with distinctive effects on vessel functionality and tumor survival. Blockade of either one of the two growth factors results in a decrease in blood vessel density and, consequently, in tumor burden. However, inhibition of the expression of VEGF, but not of FGF-2, affects also vessel maturation and functionality, leading to tumor hypoxia and necrosis. Our experimental model represents an unique tool to investigate anti-neoplastic therapies in different angiogenic environments.
Elsevier