PPARα agonist fenofibrate suppresses tumor growth through direct and indirect angiogenesis inhibition

D Panigrahy, A Kaipainen, S Huang… - Proceedings of the …, 2008 - National Acad Sciences
D Panigrahy, A Kaipainen, S Huang, CE Butterfield, CM Barnés, M Fannon, AM Laforme…
Proceedings of the National Academy of Sciences, 2008National Acad Sciences
Angiogenesis and inflammation are central processes through which the tumor
microenvironment influences tumor growth. We have demonstrated recently that peroxisome
proliferator-activated receptor (PPAR) α deficiency in the host leads to overt inflammation
that suppresses angiogenesis via excess production of thrombospondin (TSP)-1 and
prevents tumor growth. Hence, we speculated that pharmacologic activation of PPARα
would promote tumor growth. Surprisingly, the PPARα agonist fenofibrate potently …
Angiogenesis and inflammation are central processes through which the tumor microenvironment influences tumor growth. We have demonstrated recently that peroxisome proliferator-activated receptor (PPAR)α deficiency in the host leads to overt inflammation that suppresses angiogenesis via excess production of thrombospondin (TSP)-1 and prevents tumor growth. Hence, we speculated that pharmacologic activation of PPARα would promote tumor growth. Surprisingly, the PPARα agonist fenofibrate potently suppressed primary tumor growth in mice. This effect was not mediated by cancer-cell-autonomous antiproliferative mechanisms but by the inhibition of angiogenesis and inflammation in the host tissue. Although PPARα-deficient tumors were still susceptible to fenofibrate, absence of PPARα in the host animal abrogated the potent antitumor effect of fenofibrate. In addition, fenofibrate suppressed endothelial cell proliferation and VEGF production, increased TSP-1 and endostatin, and inhibited corneal neovascularization. Thus, both genetic abrogation of PPARα as well as its activation by ligands cause tumor suppression via overlapping antiangiogenic pathways. These findings reveal the potential utility of the well tolerated PPARα agonists beyond their use as lipid-lowering drugs in anticancer therapy. Our results provide a mechanistic rationale for evaluating the clinical benefits of PPARα agonists in cancer treatment, alone and in combination with other therapies.
National Acad Sciences