You are on page 1of 2

Cancer and Metastasis Reviews 19: 19–27, 2000.

© 2000 Kluwer Academic Publishers. Printed in the Netherlands.

The contributions of cyclooxygenase-2 to tumor angiogenesis

Stephen Gately
Division of Hematology/Oncology, Department of Medicine, Northwestern University Medical School,
Chicago, IL, USA

Key words: angiogenesis, apoptosis, cyclooxygenase-2, prostaglandins, vascular endothelial growth factor

Abstract

Cyclooxygenase-2 (COX-2) is an immediate early response gene that can be induced by a variety of tumor promoters,
cytokines, growth factors and hypoxia. COX-2 overexpression is linked to all stages of carcinogenesis with the
enzyme localized to the neoplastic cells, microvascular endothelial cells, and stromal fibroblasts. The contributions
of COX-2 in tumor angiogenesis include: (a) the increased expression of the proangiogenic growth factor VEGF;
(b) the production of the eicosanoid products thromboxane A2 , PGE2 and PGI2 that can directly stimulate endothelial
cell migration and growth factor-induced angiogenesis; and potentially, (c) the inhibition of endothelial cell apoptosis
by stimulation of Bcl-2 or Akt activation. Selective pharmacological inhibitors of COX-2 as angiosuppressive agents
could have therapeutic benefit in the treatment of neoplastic disease from prevention through treatment of advanced
metastatic disease. These agents are safe and well tolerated and can be added to chemotherapy and radiation therapy
where angiogenesis inhibitors appear to provide at least additive therapeutic benefit.

Classical therapeutic strategies for neoplastic diseases [20–22]. Vasculogenesis was previously thought to be
focused on the cancer cell. These approaches achieved restricted to the embryo [23,24], however, circulating
only limited success with nearly half of all diagnosed stem cells in peripheral blood have been detected that
cancer patients dying as a result of their disease despite can differentiate into endothelial cells and contribute
aggressive treatment with highly toxic regimens [1]. to angiogenesis in the adult [25–27].
Because tumor growth and metastases are depen-
dent upon angiogenesis [2–5], therapeutic strategies
that target the microvasculature are being evaluated Angiogenesis and cancer
clinically [6].
Tumor angiogenesis results from a cascade of molec- It is generally accepted that solid tumor growth and
ular and cellular events [4], often initiated by the release metastases are dependent upon the acquisition of
of angiogenic growth factors [7,8]. Tumor cells express an adequate blood supply [2,4,28–31]. Pharmaco-
a variety of proangiogenic growth factors that diffuse logical targeting of the microvasculature in patients
in the direction of pre-existing blood vessels [2,8,9]. with neoplasms represents an attractive therapeutic
These growth factors activate the normally quiescent approach because the inhibition of angiogenesis has
vascular cells inducing: (i) proteolytic degradation been shown to prevent growth [32], and can induce
of the basement membrane [10,11]; (ii) migration regression of experimental solid tumors [33]. Further-
of endothelial cells towards the angiogenic stimulus more, antiangiogenic therapy could be utilized as adju-
[12,13]; (iii) endothelial cell proliferation [12,14,15]; vant treatment for microscopic metastases, preventing
(iv) lumen formation [10,12,13,16]; (v) pericyte cap- the emergence of a vascular supply and maintaining
ping [12,17–19]; and (vi) production of a new base- tumor dormancy [34,35]. Because of the normally slow
ment membrane. Tumors can also acquire vasculature renewal of vascular stroma, even in tissues with high
through vasculogenesis, the formation of blood ves- epithelial turnover such as skin and jejunum, antiangio-
sels from progenitor endothelial cells, or angioblasts genic therapies promise to be free of the hematopoetic,
20

gastrointestinal and other toxicities of standard antipro- polyp formation was equal to that achieved by treat-
liferative therapies [33,36], and appear not to induce ing the polyp forming mice with a selective COX-2
acquired drug resistance [33]. Recent data suggest that inhibitor [54]. The pharmacologic application of selec-
selective inhibitors of cyclooxygenase-2 (COX-2) are tive COX-2 inhibitors also has been demonstrated to
potent inhibitors of angiogenesis [37,38] suggesting significantly reduce the growth of a variety of experi-
the potential utility of these agents in oncology. The mental tumors including head and neck [55], skin [56],
remainder of this review will focus on the contribution colon [57] and bladder [58].
of COX-2 in tumor angiogenesis.

Cellular expression of COX-2


COX-1 and COX-2
The elevated expression of COX-2 in the cancer setting
COX-1 and COX-2 are the two enzymes that convert has been localized to the neoplastic epithelium [59],
arachidonic acid to prostaglandins [39–41]. COX-1 within the microvasculature [37] and stromal cells [60].
is constitutively expressed and is responsible for
normal kidney and platelet function as well as the Tumor associated COX-2 expression. The potential
maintenance of the gastrointestinal mucosa [41]. In importance of tumor-associated COX-2 expression for
contrast the COX-2 enzyme can be induced by a vari- angiogenesis and tumor growth was demonstrated
ety of proinflammatory cytokines such as IL-1 [42], when COX-2 expressing tumor cells were found to
growth factors such as epidermal growth factor (EGF) grow larger and were more angiogenic in comparison
[43], transforming growth factor-beta (TGF-b) [44,45], to implanted tumor cells that lacked COX-2 expres-
tumor necrosis factor-alpha (TNF-α) [45,46], inducible sion [59]. Additionally, tumor growth and angiogenesis
nitric oxide synthase (iNOS) and ultraviolet B light could be suppressed by selective COX-2 inhibitors only
(UVB) [40]. Indomethacin, a non-selective inhibitor of if the tumor cells expressed COX-2 [61]. Together
cyclooxygenases has been shown to inhibit angiogen- these data demonstrate that tumor-associated COX-2 is
esis in experimental models of angiogenesis induced required for angiogenesis and tumor growth, and sug-
by tumors [47], thermal burns [48], copper salts [49] gest that the potential effectiveness of selective COX-2
or prostaglandins [50]. Indomethacin also suppressed inhibitors is determined by expression of COX-2 by
lymphocyte-mediated angiogenesis in vascularized the tumor cell. One mechanism for the elevated tumor-
fascia [51]. More recently, indomethacin was shown associated COX-2 expression could be related to the
to inhibit angiogenesis alone [52], and when com- marked repression of transcription of the COX-2 gene
bined with interleukin-12 [53]. Because non-selective by wild-type p53 [62]. Loss of wild-type p53 would
cyclooxygenase inhibitors are often associated with then be associated with the increased expression of
renal and gastrointestinal toxicities, selective inhibitors COX-2 mRNA and protein [62], increased secretion
of the COX-2 enzyme have been developed that may of VEGF and decreased production of an endoge-
be ideal in chronic administration for angiogenesis nous inhibitor of angiogenesis, thrombospondin-1
inhibition in the oncology setting. [63,64] resulting in the acquisition of a proangiogenic
phenotype.

COX-2 Microvascular COX-2 expression. Previous studies


suggested a role for endothelial cell-derived COX-1 in
The significant contribution of COX-2 in cancer pro- angiogenesis [59,61], however, a recent study demon-
motion was elegantly demonstrated experimentally in strated that selective COX-2 inhibitors could suppress
a model of human familial adenomatous polyposis, growth factor-induced angiogenesis, and suggested
where mice, genetically predisposed for polyp for- that endothelial-derived COX-2 is essential in directly
mation by a targeted truncation deletion in the APC regulating angiogenesis [52]. A recent histochemical
tumor suppressor gene, were crossed with COX-2−/− evaluation of human colon, breast, prostate and lung
mice [54]. The tumor burden of the double mutant tumor biopsies demonstrated that COX-1 immunore-
offspring was significantly reduced by the genetic activity is broadly distributed within normal and neo-
knockout of COX-2, furthermore the reduction in plastic tissues, whereas COX-2 immunoreactivity is

You might also like