Anda di halaman 1dari 15

Biochimica et Biophysica Acta 1654 (2004) 23 37 www.bba-direct.

com

Review

Stimulation of angiogenesis by Ras proteins


Onno Kranenburg a,*, Martijn F.B.G. Gebbink b, Emile E. Voest b
b

Department of Surgery, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands Received 7 April 2003; accepted 3 September 2003

Abstract Cells that have acquired a proliferative advantage form islets of hyperplasia during the initial stages of tumor development. Like normal cells, they require oxygen and nutrients to survive and proliferate. The centre of the islets is characterized by low oxygen pressure and low pH, conditions that stimulate the sprouting of new capillaries from nearby vascular beds. It is now well established that neovascularisation (angiogenesis) of the hyperplasias is essential for further development of the tumor. The family of ras oncogenes promotes the initiation of tumor growth by stimulating tumor cell proliferation, but also ensures tumor progression by stimulating tumor-associated angiogenesis. Oncogenic Ras proteins stimulate a number of effector pathways that culminate in the transcriptional activation of genes that control angiogenesis. Moreover, Ras signaling leads to stabilization of the produced mRNAs and, possibly, to enhanced initiation of their translation. In this review we describe the mechanisms that underlie Ras regulation of vascular endothelial growth factor (VEGF), cyclooxygenases (COX-1/-2), thrombospondins (TSP-1/-2), urokinase plasminogen activator (uPA) and matrix metalloproteases-2 and -9 (MMP-2/-9). As a result of these Ras-regulated changes in gene expression, the tumor cells cause stimulation of endothelial cells in nearby vascular beds (directly via VEGF, and indirectly via COX-produced prostaglandins) and promote remodeling of the extracellular matrix (by lowering TSP and increasing uPA/MMPs). The latter effect makes growth factors available for endothelial cell activation and migration. In addition, tumor cell-activated stromal cells also contribute to the stimulation of angiogenesis by further enhancing the production and secretion of proangiogenic factors into the tumor stroma. D 2004 Elsevier B.V. All rights reserved.
Keywords: Ras; Angiogenesis; VEGF; Cyclooxygenase; Thrombospondin; Metalloprotease; Urokinase

1. Introduction A series of genetic alterations that turn proto-oncogenes into oncogenes and active tumor suppressor genes into inactive ones underlies the complex process of tumor development. Early studies focused on identifying the transforming genes in the genomic DNA of human tumor cells. Some of these transforming oncogenes were found to be the human homologues of oncogenes in the Harvey and Kirsten Rat sarcoma viruses (v-H-ras, v-K-ras) [1 3]. A

Abbreviations: TSP, thrombospondin; COX, cyclooxygenase; NSAID, nonsteroidal anti-inflammatory drug; VEGF, vascular endothelial growth factor; MMP, matrix metalloprotease; uPA, urokinase-type plasminogen activator; FTI, farnesyltransferase inhibitor * Corresponding author. Department of Medical Oncology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands. Tel.: +31-30-2506681; fax: +31-30-2523741. E-mail address: o.kranenburg@azu.nl (O. Kranenburg). 0304-419X/$ - see front matter D 2004 Elsevier B.V. All rights reserved. doi:10.1016/j.bbcan.2003.09.004

related transforming gene was subsequently isolated from a human neuroblastoma cell line (SK-N-SH) and was designated N-ras [4,5]. Activating mutations in all three ras genes, c-H-ras, c-K-ras2 (c-K-ras1 is a pseudogene) and c-N-ras, are found in a large variety of human tumors [6]. Distinct types of tumors are associated with mutations in distinct Ras isoforms. K-ras2 mutations are frequently found in carcinomas of the colon, the lung (non-small cell lung carcinoma) and the pancreas, whereas N-ras mutations are primarily found in melanomas and hematologic cancers and H-ras mutations are uncommon in human tumors [6]. The reason(s) for these differences are still largely unclear. Wild-type Ras proteins control signal transduction pathways downstream from transiently activated (ligand-bound) tyrosine kinase receptors and G protein-coupled receptors [7,8]. Ras activation occurs through the stimulated exchange of bound GDP for GTP. GTP-bound Ras activates

24

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

several signal transduction cascades that lead to changes in gene expression and cell motility. Thereby, Ras signaling controls fundamental cell biological processes like proliferation, differentiation, survival and invasion. Mutations in the ras genes that are found in tumors (commonly in codons 12, 13 or 61) greatly impair the ability of Ras to hydrolyze bound GTP and thereby its intrinsic ability to switch off. As a result, mutant (GTPase-deficient) Ras proteins constitutively stimulate signal transduction through their effector pathways. Mutational activation of ras genes has been detected in pre-malignant hyperplasias in the colon, thyroid, lung and bone marrow [6,9 13]. Thus, Ras protein activation occurs early during tumor development in man. Several observations suggest that mutational activation of one of the ras genes is, in fact, an initiating event in tumor development. First, many of the tumors induced by mutagenic compounds in mice harbor activating mutations in one of the three ras genes and these mutations can be detected at the very early stages of tumor development [14 20]. Second, studies in mice that are genetically modified to (conditionally) express K-ras2 or H-ras oncogenes have further and unequivocally established that activated Ras promotes the initiation and progression of several types of tumors [21 26]. Moreover, the expression of activated H-Ras is also required for the maintenance of experimental (melanoma) tumor growth [27]. Constitutive Ras signaling promotes tumor cell proliferation, survival and invasiveness but also causes modulation of the behavior of non-tumor cells in the microenvironment. Of particular relevance to this review is the capacity of mutant Ras-containing tumor cells to stimulate endothelial cells in nearby capillary beds to grow out and form new blood vessels (angiogenesis) [28]. Growing tumors depend on angiogenesis for sufficient supply of oxygen and nutrients [29]. This is true for most solid tumors, but also for hematologic cancers that grow in the bone marrow [30]. The first observation that activated Ras stimulates tumorassociated angiogenesis came from a study in which the contribution of different oncogenes to tumorigenesis was studied in vivo, in a reconstituted mouse prostate gland infected with distinct oncogene-expressing retroviruses [31]. The authors found that activated Ras not only caused local hyperplasia but also hypervascularisation of the gland with more than a 10-fold increase in cytologically normal blood vessels. Interestingly, both the mutational activation of Ras and the stimulation of angiogenesis are early pre-malignant events when examined in several clinical and experimental settings [14,21,31 37]. In this review, the mechanisms underlying Ras-stimulated angiogenesis will be discussed. In particular, we have attempted to clarify the causal relationship between Rasstimulated signal transduction pathways and the angiogenesis-related phenomena that are observed in tumors and cell lines expressing the distinct activated Ras isoforms.

2. Vascular endothelial growth factor (VEGF) in Rasstimulated angiogenesis 2.1. Oncogenic Ras promotes VEGF expression VEGF-A is one of the most potent angiogenesis-stimulating growth factors, and has become a key target in antitumor therapy [38 40]. Tumors harboring mutant ras often express high levels of VEGF. Retrospective studies have shown a correlation between the presence of oncogenic Kras2 and high VEGF levels in patients with pancreas carcinoma and non-small cell lung carcinoma [41,42]. Furthermore, elevated VEGF levels and increased bone marrow vascularisation are associated with poor prognosis in hematologic malignancies [30,43 45], although in these cases the correlation with N-ras gene mutations (frequently observed in these cancers) has not yet been evaluated. Tumor-derived cell lines expressing activated K-Ras as well as cell lines transfected with an activated H-ras gene show increased synthesis and production of VEGF-A [46 48], but not VEGF-B or VEGF-C [49,50]. Conversely, inhibition of mutant K-ras2 gene expression in human colon cancer cells, either through disruption of the gene itself (by homologous recombination), through expression of a hammerhead ribozyme (specifically cleaving mutant K-Ras mRNA) or through antisense oligonucleotides, reversed the up-regulation of VEGF expression [48,51,52]. Thus, activated K-Ras is instrumental in elevating VEGF expression in colon cancer cells. What is the mechanism of Ras-stimulated VEGF production? The best-known VEGF-regulating transcription factor is hypoxia-inducible factor 1 (HIF-1), which is involved in the activation of VEGF synthesis under hypoxic conditions [53]. HIF-1 a is normally bound by the Von Hippel Lindau (VHL) tumor suppressor protein and is thereby targeted for degradation. HIF-1 a hydroxylation, as it occurs under normoxic conditions, is required for the interaction with VHL. Hypoxia leads to diminished HIF-1 a hydroxylation, release of HIF-1 a from VHL, and to expression of HIF-1 target genes, including VEGF [54 56]. Thus, HIF-1-dependent transcription is activated under hypoxic conditions. Interestingly, there is evidence to suggest that Ras activation modulates this oxygen-sensing signal transduction cascade. Two Ras-activated signal transduction pathways have been implicated in the control of VEGF expression. The classical Ras>Raf >MEK>ERK1/2 kinase cascade and the Ras >PI(3)K>PDK>PKB pathway independently control VEGF transcription in cells expressing oncogenic Ras. HIF-1 a is directly phosphorylated by ERK1/2, resulting in enhanced transactivation potential and stimulation of the VEGF promoter [50,57 60]. In addition, Ras signaling is also instrumental in hypoxia-induced stabilization of HIF1 a protein [61], and, in line with this, elevated levels of HIF-1 a are found in fibroblasts and breast cancer cells expressing activated Ras, even under normoxic conditions [62,63]. The mechanism underlying

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

25

this phenomenon is presently unknown but may involve PI(3)-kinase-regulated modulation of HIF degradation [59,62 64]. By regulating HIF-1 protein stability and activity, oncogenic Ras can stimulate VEGF gene transcription through the HIF-binding region in the VEGF promoter. However, this is not the only mechanism. Several other transcription factors on the VEGF promoter are affected by Ras signaling. A critical element in the promoter is a GC-rich stretch ( 88/ 50) that contains consensus binding sites for Sp1, Sp3 and AP2 [60,65]. Stimulation of the classical Ras> Raf >MEK>ERK1/2 pathway, either by expression of oncogenic H-Ras, inducible Raf, or constitutively active MEK, results in enhanced VEGF promoter activity that requires the Sp1/AP2 binding region. Indeed, binding of Sp1 and AP2 to this promoter element is greatly enhanced following expression of Raf [50]. This effect is presumably caused by direct phosphorylation of Sp1 by ERK1/2, as this greatly increases Sp1 binding to DNA [66,67]. VEGF mRNA levels are not solely determined by changes in gene transcription, but also by modulation of mRNA stabilization, presumably via the binding of cellular proteins to the 3VUTR [68,69]. Activation of stress-activated protein kinases (like Jun-kinase (JNK)) may modulate binding of these factors to the VEGF mRNA [69]. Oncogenic Ras stimulates these kinases through the Ras >Rac > MEKK1>JNKK>JNK pathway. VEGF mRNA is indeed three- to fivefold more stable in human tumor cells harboring activated H-Ras (EJ bladder carcinoma) and N-Ras (HT 1080 fibrosarcoma) as well as in murine fibroblasts expressing oncogenic H-ras [70], but it has not been evaluated whether Ras-mediated activation of JNK is responsible for this effect. Tumorigenicity is not only associated with changes in gene expression, but also with a general increase in mRNA translation [71]. The translation initiation factor eIF-4E is under negative control of the 4E-binding protein 1 (4E-BP1). Phosphorylation of 4E-BP1 results in decreased eIF-4E binding and enhanced translation initiation [72]. Interestingly, the kinase cascades that control 4EBP1 phosphorylation, PI(3)K>PDK>PKB>FRAP and Raf >MEK>ERK1/2, are stimulated by Ras [73,74]. Stimulated eIF-4E-dependent protein translation is essential for tumor growth of Rastransformed fibroblasts through inhibition of tumor cell apoptosis [75]. Furthermore, increased VEGF levels in head and neck, breast and bladder cancer correlate with high levels of eIF-4E expression [76 80], and VEGF protein is highly up-regulated in eIF-4E-overexpressing cells [81]. Further work is still required to assess whether the initiation of VEGF mRNA translation is indeed enhanced in tumor cells harboring oncogenic Ras and, if so, whether this is caused by increased 4E-BP phosphorylation. Taken together, Ras regulation of VEGF synthesis is very complex and occurs at multiple levels of transcription and translation (Fig. 1). Furthermore, many different cell lines have been used to study the distinct phenomena outlined

Fig. 1. Regulation of VEGF synthesis by Ras. Activated Ras controls VEGF production at multiple levels of transcription and translation. Rasactivated signal transduction pathways culminate in the phosphorylation of the transcription factors HIF-1, Sp1/3 and AP-2, resulting in enhanced binding to DNA and/or to enhanced transactivation potential. Ras regulation of stress-activated protein kinases may also control mRNA stability by stimulating the binding of proteins to the 3VUTR. In addition, two Ras-regulated pathways stimulate phosphorylation of 4E-BP, leading to release of eIF-4E and to eIF-4E-stimulated initiation of mRNA translation. Finally, wild-type Ras plays a critical role during hypoxia- and acidosisinduced regulation of VEGF synthesis. See text for references and details.

above, making it impossible to present a universal model of Ras-regulated VEGF synthesis and secretion. One or more pathways may predominate under specific conditions in specific cell types and tumors (see for instance Ref. [82]). VEGF synthesis is not only regulated by activated Ras, but also by intratumoral pH and pO2, and by cell density as well as by a number of growth factors. The overall control of VEGF synthesis is therefore determined by the combined signaling pathways activated under these conditions and those that are activated by Ras [82 84]. Is the stimulation of VEGF synthesis essential for Rasstimulated angiogenesis and tumor growth? Grunstein et al. [85] have indeed provided proof of this principle by showing that H-ras-transformed fibroblasts with a targeted deletion of the VEGF gene failed to grow out as fibrosarcomas in mice due to massively reduced vascularity and vascular permeability. In a separate study, Okada et al. [47] showed that down-regulation of VEGF transcription by antisense technology in human colon cancer cells harboring an activated K-ras2 gene greatly reduces their tumorigenic potential in vivo. Conversely, reexpression of VEGF in cells with a deleted K-ras2 oncogene is not sufficient to fully restore tumorigenic potential [47]. Likewise, enforced expression of VEGF in melanomas, initiated by an inducible H-ras oncogene, is not sufficient for maintenance of tumor growth following discontinuation of H-ras expression [27]. In this model system the sustained expression of oncogenic H-ras in the tumor cells is required for maintaining endothelial cell survival and integrity of the tumor vasculature. Surprisingly, endothelial cell apoptosis was observed well before a decline in VEGF synthesis and additional H-ras-

26

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

regulated factors may therefore be critical in maintaining tumor vessel integrity [27]. In conclusion, oncogenic H- and K-Ras stimulate VEGF production and thereby promote tumor vascularisation and tumor growth. Additional Ras-regulated factors are likely to cooperate with VEGF during these processes. Conversely, enhanced VEGF synthesis is not sufficient for either initiation or maintenance of tumor growth in the absence of activated Ras. 2.2. Endogenous Ras controls hypoxia-driven VEGF expression Due to a lack of proper vascularisation and irregular blood flow in the tumor vessels, large areas inside tumors may become hypoxic. Under hypoxic conditions, VEGF production is greatly increased and this is essential for neovascularisation of the hypoxic tissue [40,86]. Interestingly, hypoxia also leads to the activation of Ras [61,87]. Thus, also in tumor types that do not harbor ras oncogenes, wild-type Ras may be activated in hypoxic tumor areas. These findings raise two questions. First, what is the mechanism of hypoxia-induced Ras activation? And second, to what extent does Ras activation contribute to hypoxia-driven VEGF production and angiogenesis? Under hypoxic conditions, reactive oxygen and nitrogen species (ROS and RNS) are formed [88]. Ras can be directly modified and activated by radicals, in particular by SNO [89,90]. Under hypoxic conditions, SNO is synthesized by inducible NO synthase (iNOS), one of the HIFresponsive genes during hypoxia [91]. Through direct Ras activation, SNO may contribute to hypoxia-induced VEGF production. Conversely, SNO has been implicated in the negative regulation of VEGF synthesis through its effects on cGMP signaling [92]. Moreover, whereas Ras activation is robust during hypoxia and modest during reoxygenation [87], radical formation is modest during hypoxia and robust during reoxygenation [88]. These paradoxes need to be resolved in order to assess whether radical modification of Ras can cause or contribute to hypoxia-driven Ras activation. An alternative pathway of Ras activation is that induced by tyrosine kinase signaling, as it occurs after classical stimulation of tyrosine kinase or G protein-coupled receptors. Hypoxia leads to activation of the tyrosine kinase c-Src [93], tyrosine phosphorylation of the adapter protein Shc (a good c-Src substrate), activation of Ras and stimulation of ERK1/2 [61,87]. Interestingly, this signal transduction pathway is similar to the pathway that operates downstream of some (though not all) G-protein-coupled receptors [8]. More work is required to assess whether Src activation is causative in the activation of Ras and ERK1/2 following hypoxia. In addition, it remains unclear how c-Src itself is activated under hypoxic conditions. Is the transient activation of wild-type Ras under hypoxic conditions sufficient to promote VEGF production? Ras

signaling through ERK-mediated activation of the transcription factors Sp1 and Sp3 (presumably through direct phosphorylation [67]) is critical for stimulation of VEGF expression in hypoxic gastric cells [94]. Furthermore, inhibition of Ras signaling (either by using dominant negative Ras or by farnesyltransferase inhibitors (FTIs)) prevents efficient hypoxia-induced VEGF mRNA synthesis and protein secretion in malignant human astrocytoma cells that do not harbor ras mutations [95]. Thus, the induction of VEGF synthesis by hypoxia requires activation of the Ras pathway in addition to stabilization of HIF-1 a (via liberation from VHL). 2.3. Endogenous Ras controls acidosis-driven VEGF expression The intratumoral microenvironment is not only characterized by lower oxygen pressure, but also by a lower pH (acidosis). Recently, it was shown that tumor acidosis induces activation of Ras-dependent signaling to promote VEGF production [96,97]. Thus, two distinct changes in the tumor microenvironment (low pO2 and low pH) independently cause stimulation of Ras signaling and consequent VEGF production.

3. Releasing a break on angiogenesis: Ras represses thrombospondin (TSP) expression Whereas tumor cells expressing activated Ras show elevated levels of VEGF, the levels of thrombospondin-1 (TSP-1) [98 101] and thrombospondin-2 (TSP-2) [100] are dramatically reduced. TSP-1 and TSP-2 are extracellular matrix glycoproteins that are negative regulators of angiogenesis [102 105]. Several mechanism(s) may underlie the anti-angiogenic effects of TSP-1 and TSP-2. Their ability to interact with a variety of ECM components, ECM proteases, cytokines (in particular TGF-h) and growth factors and their direct effects on endothelial cell viability and migration indicate that TSP modulation of angiogenesis is very complex [103,106]. Furthermore, TSP-1 and TSP-2, though sharing their potential to act as anti-angiogenic molecules, do not share complete functional overlap as evidenced by their distinct expression patterns and by the distinct phenotypes of the respective knockout mice [103,107]. Repression of thrombospondin expression was found in cells expressing either one of the three ras isoforms, H-, Kor N-ras [99]. One of the signaling pathways that is switched on by oncogenic Ras (Ras>Rac>MEKK1>JNKK>JNK) leads to phosphorylation of the transcription factor c-Jun, by Jun kinase (JNK). The same pathway causes stimulation of c-jun gene transcription in cells expressing activated Ras [108]. Activated c-Jun, like activated Ras, strongly represses TSP-1 gene expression [98,109]. It seems likely therefore that c-Jun activation by Ras mediates, or at least contributes to, TSP-1 gene repression in Ras-transformed cells.

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

27

In a mouse model for the development of intestinal adenomas, endogenous TSP-1 expression is down-regulated in regions of dysplasia that undergo extensive neovascularisation [110]. Importantly, TSP-1 deficiency promotes the initiation and progression of tumor growth, thus establishing a critical inhibitory role for TSP-1 in the early stages of intestinal tumor development [110]. Although it is clear that TSP-1 and TSP-2 have antiangiogenic and anti-tumorigenic properties, it is less clear to what extent the down-regulation of TSP-1 and/or TSP-2 contributes to the stimulation of angiogenesis in tumor cells expressing activated Ras. The chemical induction of skin carcinomas in mice almost invariably involves acquired mutations in H-ras [17,21]. Hawighorst et al. [111] have used this model to study the functions of TSP-1 and TSP-2 during skin carcinogenesis. They found that the levels of TSP-1 declined to undetectable levels early during tumor development, possibly resulting from activation of H-ras. Conversely, the expression of TSP-2 increased dramatically, but TSP-2 expression was observed in the stromal cells rather than in the tumor cells [111]. Thus, chemically induced skin carcinogenesis is accompanied by decreased TSP-1 production and by enhanced stromal cell production of TSP-2, leading to elevated TSP-2 levels in the tumor matrix. These changes in TSP expression and deposition occur during the very early stages of papilloma development concomitantly with increased expression of VEGF in the tumor cells [111] and, possibly, following ras activation. In the same model, overexpression of TSP-1 in the mouse skin greatly impaired initiation of tumor development and angiogenesis of the dysplasias [112]. Conversely, TSP-2 deficiency greatly facilitated tumor development [111]. Further studies should clarify whether early mutational activation of H-ras is indeed causative in the observed

changes in TSP expression during skin tumor development. If so, the repression of TSP-1 expression is presumably one of the critical early events in Ras-stimulated angiogenesis and tumor development.

4. Ras regulation of the inflammatory modulators cyclooxygenase (COX)-1 and COX-2 4.1. COX-2 controls tumor growth and angiogenesis Chronic inflammation has been associated with the development of various common types of cancer and creates a pro-angiogenic environment [113 115]. In line with this notion, the regular intake of nonsteroidal anti-inflammatory drugs (NSAIDs) like aspirin reduces the risk for developing colon cancer by 40-50% [116] and may even cause regression of preexisting adenomas [117]. NSAIDs inhibit cyclooxygenases (COX-1 and COX-2), enzymes that convert arachidonic acid into prostaglandin H2 (PG-H2) which is further converted into inflammatory mediators like PG-F2a, PG-D 2 , PG-E 2 , PG-I 2 and thromboxane A 2 (TX-A 2 ) [118,119]. COX-1 is ubiquitously expressed and controls prostaglandin synthesis under physiological conditions, for instance in the control of renal blood flow and maintenance of the integrity of the gastric mucosa [118]. In contrast, COX-2 expression is inducible by growth factors and oncogenes and is highly expressed in many solid and hematologic tumors, as well as in inflamed tissue. As a result, high PG levels are found under these pathological conditions [118,120]. The importance of elevated COX-2 expression for tumor-associated angiogenesis and tumor progression has been reported in several studies demonstrating inhibitory effects of COX-inhibitors on angiogenesis

Fig. 2. Regulation of COX-2 synthesis by Ras. Activated Ras controls COX-2 production at multiple levels of transcription and translation. Ras-activated signal transduction pathways culminate in the phosphorylation of the transcription factors C/EBP, PEA3 and AP-1, resulting in enhanced transactivation potential. Ras regulation of ERK1/2 and PKB activities may also control mRNA stability by stimulating the binding of proteins to the 3V UTR. Presumably, Ras-regulated pathways also control initiation of COX-2 mRNA translation by stimulating phosphorylation of 4E-BP. Finally, activation of wild-type Ras may contribute to the critical activation of NF-n-B during hypoxia-induced regulation of COX-2 synthesis. See text for references and details.

28

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

and tumor development [115,119,121 123]. In addition, COX-deficient mice are refractory to tumor development in the skin and in the intestines [124 126]. It should be noted, however, that non-COX targets may also be critical for the anti-angiogenic and anti-tumorigenic effects of NSAIDs [127 129]. The following processes that are relevant to tumor outgrowth have been associated with elevated COX-2 levels: (i) increased tumor cell survival; (ii) increased tumor cell invasive capacity; (iii) sustained chronic inflammation; (iv) increased production of toxic and mutagenic molecules through peroxidation of metabolites; and (v) increased tumor-associated angiogenesis [130]. In this section we will only discuss the relevance of elevated COX-2 expression in (Ras-stimulated) angiogenesis. 4.2. Mechanism of COX-2-induced angiogenesis How does increased COX-2 expression, as it is observed in many tumors and pre-malignant lesions, stimulate angiogenesis? At least two distinct COX-2-stimulated processes play an important role. First, it stimulates the production of pro-angiogenic factors like bFGF, PDGF and VEGF [131], and second, it is required for endothelial cell migratory responses through integrin signaling [132]. The product of COX-2 activity, PG-H2, is converted into the biologically active molecules PG-E2, PG-D2, PG-I2, PG-F2a and TX-A2. Although all of these molecules display some form of bioactivity, only TX-A2, PG-I2 and PG-E2 have reported pro-angiogenic effects [133 137]. PGE2 activates a specific class of G protein coupled receptors (EP1-4) and stimulates VEGF and bFGF expression through cAMP signaling [138 141] (Fig. 3). Interestingly, COX-2 synthesis depends on the presence of the PGE2 receptor EP2 [142]. Thus, enhanced PGE2 levels not only cause up-regulation of VEGF and bFGF, but also stimulate COX-2 expression and subsequent PGE2 synthesis in a positive feedback loop. Tumor cell-produced TX-A2, PGI2 and/or PGE2 may stimulate the tumor cells themselves in an autocrine fashion, but they may also activate receptors on nearby endothelial cells, fibroblasts or inflammatory cells in a paracrine fashion. Tumor cells can thus affect the behavior of non-tumor cells in the microenvironment of the tumor. Is COX activity and PGE2 signaling essential for the production of pro-angiogenic factors and the subsequent stimulation of angiogenesis? COX inhibitors indeed prevent the production of VEGF and bFGF by colon cancer cells or fibroblasts [131,143] in in vitro angiogenesis assays, as well as during tumor-associated angiogenesis [131,144]. Furthermore, signaling through EP2 by the COX-generated product PGE2 is essential for VEGF production, angiogenesis and tumor development in the intestine [142,145]. Finally, stromal fibroblasts are a major source of VEGF in the tumor and the expression of both COX-2 and the PGE2 receptor EP3 is essential for VEGF expression by these cells [143,144]. Taken together, COX activity and signaling by its products

are essential for the production of pro-angiogenic growth factors and for the stimulation of angiogenesis in vivo. 4.3. Induction of COX-2 gene expression by Ras COX-2 gene expression is regulated by a variety of signals that are involved in either initiation or promotion of tumor growth: (i) deregulation of the Wnt pathway [146,147]; (ii) loss of p53 expression [148]; (iii) HER-2/neu overexpression [149]; (iv) hypoxia [150]; and (v) signaling by oncogenic Ras and Src [151 154]. Distinct Ras-activated signaling pathways have been implicated in the control of COX-2 expression (Fig. 2). First, the Ras>Rac>MEKK1>JNKK>JNK pathway promotes COX-2 transcription through the cyclic-AMP response element (CRE) via phosphorylation of c-Jun, a component of the transcription factor activator protein 1 (AP1) [155]. In addition, the classical Ras>Raf >MEK>ERK1/2 pathway is required for COX-2 up-regulation by activated H-ras [152] presumably through phosphorylation of the CCAAT/enhancer binding protein h (C/EBP h) [156] and/or the Ets transcription factor PEA3 [149]. H-Rasstimulated phosphorylation of PEA3 is mediated by both MAPK and JNK [157]. PEA-3 activation is also essential for the stimulation of uPA gene expression by oncogenic Ras (see below) [158,159]. The stability of the 3VUTR of COX-2 mRNA is greatly increased by expression of activated K-ras2 in intestinal epithelial cells, a phenomenon that is caused by activation of both the Ras>Raf >MEK>ERK1/2 pathway as well as the Ras >PI(3)K>PDK>PKB pathway [160]. The latter pathway also controls the efficiency of cap-dependent mRNA translation [73]. However, it is not known whether activated Ras stimulates the initiation of COX-2 mRNA translation, as it does with the VEGF mRNA (see above). Deregulated Wnt-signaling occurs early in colon cancer development following inactivation of APC. As a result, the transcription factor TCF is de-repressed and TCF-mediated signaling ensues. A functional TCF-binding promoter element (TBE) in the COX-2 promoter may contribute to enhanced COX-2 expression following APC gene inactivation [147]. In addition, activation of the Wnt pathway leads to up-regulation of PEA3 and to PEA3-mediated COX-2 transcription [161]. Taken together, both Wnt- and K-Rasstimulated pathways cooperate in the stimulation of COX-2 expression and this may be especially relevant during the development of early neoplasias in the colon (Fig. 2). 4.4. The contribution of COX-2 to Ras-stimulated angiogenesis Mutations in Ras are initiating events in tumor development and can be detected in pre-malignant tissues. Similarly, COX-2 overexpression is detected at the very early stages of tumor development in pre-malignant hyperplasias in the pancreas, the colon and the lung (associated with K-ras2

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

29

Fig. 3. Up-regulation of COX-2 and COX-initiated signaling in response to Ras. The up-regulation of COX-2 in tumors harboring activated Ras leads to the enhanced production of prostaglandins. PGE2, PGI2 and thromboxane A2 have reported pro-angiogenic effects. Stimulation of PGE2 receptors EP2 or EP3 either on the tumor cells themselves or on stromal cells leads to increased synthesis of cAMP and to cAMP-stimulated VEGF (and possibly bFGF) gene transcription in both tumor and stromal cells. EP receptor activation also leads to further stimulation of COX-2 synthesis, thereby creating a positive feedback loop. See text for references and details.

mutations) and in the bone marrow (associated with N-ras mutations) [119,162]. Although activated Ras certainly contributes to COX-2 expression (see above), other inputs can also regulate COX-2 expression, such as hypoxia [163], cytokines and growth factors [164]. Interestingly, COX-1 and COX-2 are dispensable for Hras-induced transformation of fibroblasts in vitro [128]. Unfortunately, the ability of COX-1- or COX-2-deficient cells to grow as fibrosarcomas in vivo was not further investigated in this study. During initiation of tumor growth, the up-regulation of these enzymes by activated Ras presumably affects processes other than initial cellular transformation. Obviously, the stimulation of angiogenesis is one such process. The initiation and progression of chemically induced skin carcinomas in the mouse is invariably accompanied by mutations in H-ras [21,165] and is greatly reduced in COX-1- and COX-2-deficient mice [125], as it is in Hras-deficient mice [165]. This suggests that COX-1 and COX-2 play an essential role in H-Ras-initiated skin tumorigenesis. However, the reduced tumorigenesis in both COX1- and COX-2-deficient animals is presumably due to the premature terminal differentiation of keratinocytes [125]. A wealth of data highlight the essential role for COX-2, PG-E2 and the PG-E2 receptors EP2 and EP3 in the stimulation of angiogenesis and tumor development in mice [124,142 145]. However, these studies do not provide proof for the involvement of COX-2 and its products in Ras-induced angiogenesis. With the development of mouse models in which tumorigenesis can be induced by conditionally switching on activated ras alleles [166,167], it should be feasible to study the contribution of COX-2 and PG-E2-EP2/ 3 signaling to Ras-initiated tumor development and angiogenesis. Given the central role for COX-2 in mediating

inflammatory responses and the fact that chronic inflammation is a pro-angiogenic condition [113 115], the stimulation of COX-2 expression by Ras will undoubtedly contribute to Ras-stimulated angiogenesis.

5. Changes in the proteolytic environment induced by Ras Activated Ras stimulates the expression of several proteases that break down the extracellular matrix. These include urokinase plasminogen activator (uPA) [168,169] and matrix metalloproteases (MMP-2 and MMP-9) [170, 171], proteases that play a critical role in the control of tumor angiogenesis [172]. Overexpression of uPA is associated with poor prognosis and enhanced tumor dissemination in several types of cancer [173]. uPA is one of the two proteases (the other being tissue-type plasminogen activator, tPA) that controls conversion of the inactive zymogen plasminogen into the active protease plasmin. Plasmin is a broad specificity serine protease. Its classical function is to degrade fibrin in blood clots (fibrinolysis), but other substrates include components of the extracellular matrix and of basement membranes. Plasmin also activates matrix metalloproteases, resulting in further breakdown of the ECM [174]. The uPA receptor (uPAR) serves to localize plasmin formation to the cell surface [175], thereby facilitating tumor cell migration through extracellular matrices and basement membranes [176]. In addition to directing cellassociated plasmin formation, uPA activates intracellular signaling pathways through the uPAR, thereby stimulating cell proliferation and migration in a manner that is independent of uPA proteolytic activity [177].

30

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

Activated Ras stimulates uPA expression through distinct elements in the uPA gene promoter. First, a combined PEA3/AP1 binding motif at 1967 binds Ets transcription factors (targets for the Ras>Raf >MEK>ERK1/2 pathway) and c-Jun (a target for the Ras>Rac>MEKK1>JNKK>JNK pathway) [158,159]. An additional AP1 motif at 1880 has also been implicated in the Ras-dependent stimulation of uPA expression [178]. In addition to stimulating the classical effector pathways that lead to MAPK and JNK activation, oncogenic Ras also directly activates guanine nucleotide exchange factors for the small GTPase Ral [179]. Ral-dependent signaling is critical for stimulation of uPA expression (but not MMP-2 or MMP-9 expression) by Ras [180], as evidenced by expression of a dominant negative Ral mutant. It is presently unknown how Ral signaling affects uPA promoter activity. Activated Ras also stimulates expression of the matrix metalloproteases MMP-2 and MMP-9. Multiple elements in the MMP-9 promoter, including PEA3- and AP1-binding sites, are essential for promoter stimulation by H-Ras [170]. Thus, optimal stimulation of both uPA and MMP-9 gene expression by Ras requires signaling through MAPK- and JNK-responsive promoter elements. Conversely, stimulation of the MMP-9 promoter by activated H-Ras in kidney epithelial cells depends on the intracellular generation of radical oxygen species and subsequent activation of NF-n-B [181]. In addition to activating transcription factors that act on the MMP-9 promoter to stimulate gene transcription, Ras and/or Myc also stimulates translation of the MMP-9 mRNA in prostate carcinoma cells [182]. As discussed above in Section 2.1, Ras may enhance cap-dependent mRNA translation by disrupting the complex between 4E-BP and eIF-4E through stimulation of 4E-BP phosphorylation. The uPA uPAR complex on endothelial cells may promote the migration of developing sprouts through the provisional matrix that is primarily composed of fibrin [183]. This process is essential during the early phases of angiogenesis. Likewise, enhanced production of uPA (and other proteases) by tumor cells enhances tumor cell migration and invasion. Obviously, enhanced uPA expression in endothelial cells cannot be the direct consequence of Ras activation in tumor cells. This raises the question whether the elevated levels of uPA that are produced by tumor cells contribute to the stimulation of angiogenesis. Two distinct mechanisms should be considered. First, plasmin and MMPs can liberate pro-angiogenic growth factors like VEGF and bFGF from the ECM, thereby making them available for endothelial cell activation [184 187]. In fact, liberation of VEGF by MMP-9 (produced by infiltrating inflammatory cells) controls the onset of angiogenesis in oncogene (SV40-largeT)-induced pancreatic tumors [188]. In addition, the latent form of the pro-angiogenic cytokine TGF-h can be converted into active TGF-h by plasmin [186]. Signaling by activated TGF-h may subsequently synergize with Ras signaling in the induction of VEGF synthesis [84].

Highly vascularised melanomas can be induced in mice by application of specific carcinogens [189]. The majority of chemically induced skin tumors harbor an activated Hras oncogene [21] and activation of Ras (or Ras effector systems) is critical for melanoma development in the mouse [190]. Interestingly, uPA-deficient mice are refractory to melanoma induction by carcinogens [191]. Reduced (plasmin-mediated) liberation of pro-angiogenic growth factors may underlie this phenomenon [191]. Nevertheless, formal proof for the involvement of MMP-9, MMP-2, and/or uPA in the stimulation of angiogenesis by tumor cells expressing activated Ras is lacking. Second, protease-generated degradation products of ECM components themselves may have positive or negative effects on endothelial cell migration, proliferation and angiogenesis [172]. In fact, protease-generated fragments from plasminogen, collagens IV, XV and XVIII possess strong anti-angiogenic and anti-tumor activity in mouse models through mechanisms that are still largely unclear [172]. Many of these biological compounds are now being further tested in clinical trials. It remains to be established to what extent protease-generated ECM fragments positively or negatively control angiogenesis during Ras-induced tumorigenesis. Taken together, oncogenic Ras promotes tumor angiogenesis by at least three independent mechanisms: (i) by stimulating the production of pro-angiogenic growth factors; (ii) by reducing the production of anti-angiogenic factors; and (iii) by ensuring the availability of pro-angiogenic growth factors through local degradation of the ECM (Fig. 4).

Fig. 4. Ras-stimulated pathways that promote angiogenesis. Oncogenic Ras up-regulates VEGF expression in tumor cells, but can also do so in nearby stromal cells through stimulation of COX-2 expression, subsequent prostaglandin synthesis and activation of stromal cell PG receptors (EP). In a parallel pathway, oncogenic Ras stimulates the production of uPA and MMPs that break down the extracellular matrix surrounding the tumor cells. As a result, growth factors are liberated and become available for endothelial cell activation. Moreover, ECM breakdown products themselves directly affect endothelial cell behavior and angiogenesis (positively or negatively). Activated Ras also represses expression of the antiangiogenic matrix glycoprotein TSP-1. See text for references and details.

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337

31

6. Isoprenylation inhibitors as anti-angiogenic compounds Posttranslational prenylation is required for the association of all Ras proteins (H-Ras, N-Ras, K-Ras2-4A, K-Ras24B) with the plasma membrane. The cellular precursor for prenylation is 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA), which is converted into mevalonate by HMG-CoA reductase. Mevalonate is further metabolized into farnesylpyrophosphate and, subsequently, into geranylgeranylpyrophosphate. The latter two molecules can be covalently attached to a large variety of proteins, including Ras, by protein:farnesyl transferase (PFT) and protein:geranylgeranyl transferase (PGGT). The C-terminal CAAX motif is a target for either farnesylation (H-, K-, and NRas) or, when PFT is inhibited, geranylgeranylation (K- and N-Ras). In addition, K-Ras2-4B has a C-terminal lysine-rich region (positively charged) that allows electrostatic interaction with the (negatively charged) plasma membrane. Since farnesylation is critical for proper Ras localization and, hence, for Ras function, FTIs as well as HMG-CoA inhibitors (statins) have been extensively studied for their potential anti-tumor effects. Some of these compounds have shown promising results in in vitro experiments, in tumor models in mice, as well as in clinical trials. Interestingly, growing evidence suggests that both classes of compounds have profound anti-angiogenic effects in vitro as well as in vivo [192 197]. Treatment of tumor cells with these compounds is accompanied by decreased VEGF secretion [195 200], possibly due to reduced Ras signaling. However, the results from phase I clinical trials show that also tumors that do not harbor activated Ras oncogenes can respond to FTI treatment [201,202]. In the absence of activated mutant Ras, tumor growth and VEGF secretion may still require signaling through wild-type Ras. Such tumors may therefore still be FTI-sensitive [95,203]. Nevertheless, it is becoming increasingly clear that even though they were developed as specific Ras inhibitors, inhibition of tumor growth by FTIs and statins may rely on the inhibition of targets other than Ras. Many proteins aside from Ras are isoprenylated, and some of these are involved in cellular signal transduction and cell cycle control themselves [204,205]. In addition to affecting tumor cell behavior, FTIs and statins also inhibit endothelial cell migration and tube formation in vitro [192 194,197]. Again, inhibition of Ras signaling may not always underlie the observed effects [192]. Thus, FTIs and/or statins may interfere with tumor angiogenesis directly through inhibitory effects on endothelial cell activation, and indirectly by lowering VEGF secretion by tumor cells. Taken together, the antitumor activity of prenylationinhibiting compounds is presumably due to inhibition of more than one target and to modulation of multiple biological processes within the tumor. Nevertheless, Ras activity plays a crucial role in the control of tumor angiogenesis and

it seems likely therefore that the inhibition of angiogenesis by these compounds is due, at least in part, to inhibition of Ras function.

Acknowledgements The authors thank Drs. Hans Bos and Pat Crowley for critically reading the manuscript and for their helpful comments.

References
[1] S. Pulciani, E. Santos, A.V. Lauver, L.K. Long, K.C. Robbins, M. Barbacid, Oncogenes in human tumor cell lines: molecular cloning of a transforming gene from human bladder carcinoma cells, Proc. Natl. Acad. Sci. U. S. A. 79 (1982) 2845 2849. [2] L.F. Parada, C.J. Tabin, C. Shih, R.A. Weinberg, Human EJ bladder carcinoma oncogene is homologue of Harvey sarcoma virus ras gene, Nature 297 (1982) 474 478. [3] C.J. Der, T.G. Krontiris, G.M. Cooper, Transforming genes of human bladder and lung carcinoma cell lines are homologous to the ras genes of Harvey and Kirsten sarcoma viruses, Proc. Natl. Acad. Sci. U. S. A. 79 (1982) 3637 3640. [4] K. Shimizu, M. Goldfarb, Y. Suard, M. Perucho, Y. Li, T. Kamata, J. Feramisco, E. Stavnezer, J. Fogh, M.H. Wigler, Three human transforming genes are related to the viral ras oncogenes, Proc. Natl. Acad. Sci. U. S. A. 80 (1983) 2112 2116. [5] K. Shimizu, M. Goldfarb, M. Perucho, M. Wigler, Isolation and preliminary characterization of the transforming gene of a human neuroblastoma cell line, Proc. Natl. Acad. Sci. U. S. A. 80 (1983) 383 387. [6] J.L. Bos, Ras oncogenes in human cancer: a review, Cancer Res. 49 (1989) 4682 4689. [7] G. Carpenter, The EGF receptor: a nexus for trafficking and signaling, BioEssays 22 (2000) 697 707. [8] O. Kranenburg, W.H. Moolenaar, Ras-MAP kinase signaling by lysophosphatidic acid and other G protein-coupled receptor agonists, Oncogene 20 (2001) 1540 1546. [9] K. Motojima, T. Urano, Y. Nagata, H. Shiku, T. Tsunoda, T. Kanematsu, Mutations in the Kirsten-ras oncogene are common but lack correlation with prognosis and tumor stage in human pancreatic carcinoma, Am. J. Gastroenterol. 86 (1991) 1784 1788. [10] M. Sagawa, Y. Saito, S. Fujimura, R.I. Linnoila, K-ras point mutation occurs in the early stage of carcinogenesis in lung cancer, Br. J. Cancer 77 (1998) 720 723. [11] N.R. Lemoine, E.S. Mayall, F.S. Wyllie, E.D. Williams, M. Goyns, B. Stringer, D. Wynford-Thomas, High frequency of ras oncogene activation in all stages of human thyroid tumorigenesis, Oncogene 4 (1989) 159 164. [12] J.J. Yunis, A.J. Boot, M.G. Mayer, J.L. Bos, Mechanisms of ras mutation in myelodysplastic syndrome, Oncogene 4 (1989) 609 614. [13] J.L. Bos, E.R. Fearon, S.R. Hamilton, M. Verlaan-de Vries, J.H. van Boom, A.J. van der Eb, B. Vogelstein, Prevalence of ras gene mutations in human colorectal cancers, Nature 327 (1987) 293 297. [14] R. Bremner, A. Balmain, Genetic changes in skin tumor progression: correlation between presence of a mutant ras gene and loss of heterozygosity on mouse chromosome 7, Cell 61 (1990) 407 417. [15] R. Kumar, S. Sukumar, M. Barbacid, Activation of ras oncogenes preceding the onset of neoplasia, Science 248 (1990) 1101 1104. [16] M. Osaka, S. Matsuo, T. Koh, T. Sugiyama, Specific N-ras mutation in bone marrow within 48 H of 7,12-dimethylbenz[a]anthracene

32

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 treatment in Huggins-Sugiyama rat leukemogenesis, Mol. Carcinog. 16 (1996) 126 131. M. Quintanilla, K. Brown, M. Ramsden, A. Balmain, Carcinogenspecific mutation and amplification of Ha-ras during mouse skin carcinogenesis, Nature 322 (1986) 78 80. S. Sukumar, V. Notario, D. Martin-Zanca, M. Barbacid, Induction of mammary carcinomas in rats by nitroso-methylurea involves malignant activation of H-ras-1 locus by single point mutations, Nature 306 (1983) 658 661. S. Sukumar, B. Armstrong, J.P. Bruyntjes, I. Leav, M.C. Bosland, Frequent activation of the Ki-ras oncogene at codon 12 in N-methylN-nitrosourea-induced rat prostate adenocarcinomas and neurogenic sarcomas, Mol. Carcinog. 4 (1991) 362 368. H. Zarbl, S. Sukumar, A.V. Arthur, D. Martin-Zanca, M. Barbacid, Direct mutagenesis of Ha-ras-1 oncogenes by N-nitroso-N-methylurea during initiation of mammary carcinogenesis in rats, Nature 315 (1985) 382 385. A. Balmain, K. Brown, Oncogene activation in chemical carcinogenesis, Adv. Cancer Res. 51 (1988) 147 182. K. Brown, M. Quintanilla, M. Ramsden, I.B. Kerr, S. Young, A. Balmain, v-ras genes from Harvey and BALB murine sarcoma viruses can act as initiators of two-stage mouse skin carcinogenesis, Cell 46 (1986) 447 456. E.L. Jackson, N. Willis, K. Mercer, R.T. Bronson, D. Crowley, R. Montoya, T. Jacks, D.A. Tuveson, Analysis of lung tumor initiation and progression using conditional expression of oncogenic K-ras, Genes Dev. 15 (2001) 3243 3248. L. Johnson, K. Mercer, D. Greenbaum, R.T. Bronson, D. Crowley, D.A. Tuveson, T. Jacks, Somatic activation of the K-ras oncogene causes early onset lung cancer in mice, Nature 410 (2001) 1111 1116. R. Meuwissen, S.C. Linn, D. Van der Valk, W.J. Mooi, A. Berns, Mouse model for lung tumorigenesis through Cre/lox controlled sporadic activation of the K-Ras oncogene, Oncogene 20 (2001) 6551 6558. C.J. Quaife, C.A. Pinkert, D.M. Ornitz, R.D. Palmiter, R.L. Brinster, Pancreatic neoplasia induced by ras expression in acinar cells of transgenic mice, Cell 48 (1987) 1023 1034. L. Chin, A. Tam, J. Pomerantz, M. Wong, J. Holash, N. Bardeesy, Q. Shen, R. OHagan, J. Pantginis, H. Zhou, J.W. Horner, C. CordonCardo, G.D. Yancopoulos, R.A. DePinho, Essential role for oncogenic Ras in tumour maintenance, Nature 400 (1999) 468 472. J. Rak, J. Filmus, G. Finkenzeller, S. Grugel, D. Marme, R.S. Kerbel, Oncogenes as inducers of tumor angiogenesis, Cancer Metastasis Rev. 14 (1995) 263 277. J. Folkman, What is the evidence that tumors are angiogenesis dependent? J. Natl. Cancer Inst. 82 (1990) 4 6. F.J. Giles, The emerging role of angiogenesis inhibitors in hematologic malignancies, Oncology (Huntingt.) 16 (2002) 23 29. T.C. Thompson, J. Southgate, G. Kitchener, H. Land, Multistage carcinogenesis induced by ras and myc oncogenes in a reconstituted organ, Cell 56 (1989) 917 930. A. Balmain, I.B. Pragnell, Mouse skin carcinomas induced in vivo by chemical carcinogens have a transforming Harvey-ras oncogene, Nature 303 (1983) 72 74. A. Balmain, M. Ramsden, G.T. Bowden, J. Smith, Activation of the mouse cellular Harvey-ras gene in chemically induced benign skin papillomas, Nature 307 (1984) 658 660. P. Bossi, G. Viale, A.K. Lee, R. Alfano, G. Coggi, S. Bosari, Angiogenesis in colorectal tumors: microvessel quantitation in adenomas and carcinomas with clinicopathological correlations, Cancer Res. 55 (1995) 5049 5053. L.F. Brown, A.J. Guidi, K. Tognazzi, H.F. Dvorak, Vascular permeability factor/vascular endothelial growth factor and vascular stroma formation in neoplasia. Insights from in situ hybridization studies, J. Histochem. Cytochem. 46 (1998) 569 575. J. Folkman, K. Watson, D. Ingber, D. Hanahan, Induction of angiogenesis during the transition from hyperplasia to neoplasia, Nature 339 (1989) 58 61. M.P. Wong, N. Cheung, S.T. Yuen, S.Y. Leung, L.P. Chung, Vascular endothelial growth factor is up-regulated in the early pre-malignant stage of colorectal tumour progression, Int. J. Cancer 81 (1999) 845 850. H.F. Dvorak, L.F. Brown, M. Detmar, A.M. Dvorak, Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis, Am. J. Pathol. 146 (1995) 1029 1039. P.W. Manley, G. Martiny-Baron, J.M. Schlaeppi, J.M. Wood, Therapies directed at vascular endothelial growth factor, Expert Opin. Investig. Drugs 11 (2002) 1715 1736. G. Siemeister, G. Martiny-Baron, D. Marme, The pivotal role of VEGF in tumor angiogenesis: molecular facts and therapeutic opportunities, Cancer Metastasis Rev. 17 (1998) 241 248. N. Ikeda, Y. Nakajima, M. Sho, M. Adachi, C.L. Huang, K. Iki, H. Kanehiro, M. Hisanaga, H. Nakano, M. Miyake, The association of K-ras gene mutation and vascular endothelial growth factor gene expression in pancreatic carcinoma, Cancer 92 (2001) 488 499. T. Konishi, C.L. Huang, M. Adachi, T. Taki, H. Inufusa, K. Kodama, N. Kohno, M. Miyake, The K-ras gene regulates vascular endothelial growth factor gene expression in non-small cell lung cancers, Int. J. Oncol. 16 (2000) 501 511. A. Aguayo, H. Kantarjian, T. Manshouri, C. Gidel, E. Estey, D. Thomas, C. Koller, Z. Estrov, S. OBrien, M. Keating, E. Freireich, M. Albitar, Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes, Blood 96 (2000) 2240 2245. A. Aguayo, H.M. Kantarjian, E.H. Estey, F.J. Giles, S. Verstovsek, T. Manshouri, C. Gidel, S. OBrien, M.J. Keating, M. Albitar, Plasma vascular endothelial growth factor levels have prognostic significance in patients with acute myeloid leukemia but not in patients with myelodysplastic syndromes, Cancer 95 (2002) 1923 1930. S. Verstovsek, H. Kantarjian, T. Manshouri, J. Cortes, F.J. Giles, A. Rogers, M. Albitar, Prognostic significance of cellular vascular endothelial growth factor expression in chronic phase chronic myeloid leukemia, Blood 99 (2002) 2265 2267. J.L. Arbiser, M.A. Moses, C.A. Fernandez, N. Ghiso, Y. Cao, N. Klauber, D. Frank, M. Brownlee, E. Flynn, S. Parangi, H.R. Byers, J. Folkman, Oncogenic H-ras stimulates tumor angiogenesis by two distinct pathways, Proc. Natl. Acad. Sci. U. S. A. 94 (1997) 861 866. F. Okada, J.W. Rak, B.S. Croix, B. Lieubeau, M. Kaya, L. Roncari, S. Shirasawa, T. Sasazuki, R.S. Kerbel, Impact of oncogenes in tumor angiogenesis: mutant K-ras up-regulation of vascular endothelial growth factor/vascular permeability factor is necessary, but not sufficient for tumorigenicity of human colorectal carcinoma cells, Proc. Natl. Acad. Sci. U. S. A. 95 (1998) 3609 3614. J. Rak, Y. Mitsuhashi, L. Bayko, J. Filmus, S. Shirasawa, T. Sasazuki, R.S. Kerbel, Mutant ras oncogenes upregulate VEGF/VPF expression: implications for induction and inhibition of tumor angiogenesis, Cancer Res. 55 (1995) 4575 4580. B. Enholm, K. Paavonen, A. Ristimaki, V. Kumar, Y. Gunji, J. Klefstrom, L. Kivinen, M. Laiho, B. Olofsson, V. Joukov, U. Eriksson, K. Alitalo, Comparison of VEGF, VEGF-B, VEGF-C and Ang-1 mRNA regulation by serum, growth factors, oncoproteins and hypoxia, Oncogene 14 (1997) 2475 2483. J. Milanini, F. Vinals, J. Pouyssegur, G. Pages, p42/p44 MAP kinase module plays a key role in the transcriptional regulation of the vascular endothelial growth factor gene in fibroblasts, J. Biol. Chem. 273 (1998) 18165 18172. P.J. Ross, M. George, D. Cunningham, F. DiStefano, H.J. Andreyev, P. Workman, P.A. Clarke, Inhibition of Kirsten-ras expression in human colorectal cancer using rationally selected Kirsten-ras antisense oligonucleotides, Mol. Cancer Ther. 1 (2001) 29 41. T. Tokunaga, T. Tsuchida, H. Kijima, K. Okamoto, Y. Oshika, N. Sawa, Y. Ohnishi, H. Yamazaki, S. Miura, Y. Ueyama, M. Naka-

[17]

[37]

[18]

[38]

[19]

[39]

[20]

[40]

[41]

[21] [22]

[42]

[23]

[43]

[24]

[44]

[25]

[45]

[26]

[46]

[27]

[28]

[47]

[29] [30] [31]

[48]

[32]

[49]

[33]

[50]

[34]

[51]

[35]

[52]

[36]

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 mura, Ribozyme-mediated inactivation of mutant K-ras oncogene in a colon cancer cell line, Br. J. Cancer 83 (2000) 833 839. P.H. Maxwell, P.J. Ratcliffe, Oxygen sensors and angiogenesis, Semin. Cell Dev. Biol. 13 (2002) 29 37. N. Masson, C. Willam, P.H. Maxwell, C.W. Pugh, P.J. Ratcliffe, Independent function of two destruction domains in hypoxia-inducible factor-alpha chains activated by prolyl hydroxylation, EMBO J. 20 (2001) 5197 5206. F. Yu, S.B. White, Q. Zhao, F.S. Lee, HIF-1alpha binding to VHL is regulated by stimulus-sensitive proline hydroxylation, Proc. Natl. Acad. Sci. U. S. A. 98 (2001) 9630 9635. M. Ivan, K. Kondo, H. Yang, W. Kim, J. Valiando, M. Ohh, A. Salic, J.M. Asara, W.S. Lane, W.G. Kaelin Jr., HIFalpha targeted for VHLmediated destruction by proline hydroxylation: implications for O2 sensing, Science 20 (292) (2001) 464 468. E. Hur, K.Y. Chang, E. Lee, S.K. Lee, H. Park, Mitogen-activated protein kinase kinase inhibitor PD98059 blocks the trans-activation but not the stabilization or DNA binding ability of hypoxia-inducible factor-1alpha, Mol. Pharmacol. 59 (2001) 1216 1224. E. Lee, S. Yim, S.K. Lee, H. Park, Two transactivation domains of hypoxia-inducible factor-1alpha regulated by the MEK-1/p42/p44 MAPK pathway, Mol. Cells 14 (2002) 9 15. A. Sodhi, S. Montaner, H. Miyazaki, J.S. Gutkind, MAPK and Akt act cooperatively but independently on hypoxia inducible factor1alpha in rasV12 upregulation of VEGF, Biochem. Biophys. Res. Commun. 287 (2001) 292 300. D.E. Richard, E. Berra, E. Gothie, D. Roux, J. Pouyssegur, p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1alpha (HIF-1alpha) and enhance the transcriptional activity of HIF-1, J. Biol. Chem. 274 (1999) 32631 32637. F. Jung, J. Haendeler, J. Hoffmann, A. Reissner, E. Dernbach, A.M. Zeiher, S. Dimmeler, Hypoxic induction of the hypoxia-inducible factor is mediated via the adaptor protein Shc in endothelial cells, Circ. Res. 91 (2002) 38 45. C. Blancher, J.W. Moore, N. Robertson, A.L. Harris, Effects of ras and von Hippel-Lindau (VHL) gene mutations on hypoxia-inducible factor (HIF)-1alpha, HIF-2alpha, and vascular endothelial growth factor expression and their regulation by the phosphatidylinositol 3V -kinase/Akt signaling pathway, Cancer Res. 61 (2001) 7349 7355. C. Chen, N. Pore, A. Behrooz, F. Ismail-Beigi, A. Maity, Regulation of glut1 mRNA by hypoxia-inducible factor-1. Interaction between H-ras and hypoxia, J. Biol. Chem. 276 (2001) 9519 9525. N.M. Mazure, E.Y. Chen, K.R. Laderoute, A.J. Giaccia, Induction of vascular endothelial growth factor by hypoxia is modulated by a phosphatidylinositol 3-kinase/Akt signaling pathway in Ha-ras-transformed cells through a hypoxia inducible factor-1 transcriptional element, Blood 90 (1997) 3322 3331. E. Berra, G. Pages, J. Pouyssegur, MAP kinases and hypoxia in the control of VEGF expression, Cancer Metastasis Rev. 19 (2000) 139 145. J. Milanini-Mongiat, J. Pouyssegur, G. Pages, Identification of two Sp1 phosphorylation sites for p42/p44 mitogen-activated protein kinases: their implication in vascular endothelial growth factor gene transcription, J. Biol. Chem. 277 (2002) 20631 20639. J.L. Merchant, M. Du, A. Todisco, Sp1 phosphorylation by Erk 2 stimulates DNA binding, Biochem. Biophys. Res. Commun. 19 (254) (1999) 454 461. A.P. Levy, N.S. Levy, M.A. Goldberg, Post-transcriptional regulation of vascular endothelial growth factor by hypoxia, J. Biol. Chem. 271 (1996) 2746 2753. G. Pages, E. Berra, J. Milanini, A.P. Levy, J. Pouyssegur, Stressactivated protein kinases (JNK and p38/HOG) are essential for vascular endothelial growth factor mRNA stability, J. Biol. Chem. 275 (2000) 26484 26491. F.C. White, A. Benehacene, J.S. Scheele, M. Kamps, VEGF mRNA is stabilized by ras and tyrosine kinase oncogenes, as well as by UV

33

[53] [54]

[71]

[72]

[55]

[73]

[56]

[74]

[57]

[75]

[58]

[76]

[59]

[77]

[60]

[78]

[61]

[79]

[62]

[80]

[81]

[63]

[64]

[82]

[65]

[83]

[66]

[84]

[67]

[85]

[68]

[86]

[69]

[87]

[70]

radiation evidence for divergent stabilization pathways, Growth Factors 14 (1997) 199 212. N. Sonenberg, A.C. Gingras, The mRNA 5Vcap-binding protein eIF4E and control of cell growth, Curr. Opin. Cell Biol. 10 (1998) 268 275. A.C. Gingras, B. Raught, N. Sonenberg, eIF4 initiation factors: effectors of mRNA recruitment to ribosomes and regulators of translation, Ann. Rev. Biochem. 68 (1999) 913 963. A.C. Gingras, S.P. Gygi, B. Raught, R.D. Polakiewicz, R.T. Abraham, M.F. Hoekstra, R. Aebersold, N. Sonenberg, Regulation of 4EBP1 phosphorylation: a novel two-step mechanism, Genes Dev. 13 (1999) 1422 1437. T.P. Herbert, A.R. Tee, C.G. Proud, The extracellular signal-regulated kinase pathway regulates the phosphorylation of 4E-BP1 at multiple sites, J. Biol. Chem. 277 (2002) 11591 11596. V.A. Polunovsky, A.C. Gingras, N. Sonenberg, M. Peterson, A. Tan, J.B. Rubins, J.C. Manivel, P.B. Bitterman, Translational control of the antiapoptotic function of Ras, J. Biol. Chem. 275 (2000) 24776 24780. P.A. Scott, K. Smith, R. Poulsom, A. De Benedetti, R. Bicknell, A.L. Harris, Differential expression of vascular endothelial growth factor mRNA vs protein isoform expression in human breast cancer and relationship to eIF-4E, Br. J. Cancer 77 (1998) 2120 2128. C.G. Kevil, A. De Benedetti, D.K. Payne, L.L. Coe, F.S. Laroux, J.S. Alexander, Translational regulation of vascular permeability factor by eukaryotic initiation factor 4E: implications for tumor angiogenesis, Int. J. Cancer 65 (1996) 785 790. A. De Benedetti, A.L. Harris, eIF4E expression in tumors: its possible role in progression of malignancies, Int. J. Biochem. Cell Biol. 31 (1999) 59 72. J.P. Crew, S. Fuggle, R. Bicknell, D.W. Cranston, A. De Benedetti, A.L. Harris, Eukaryotic initiation factor-4E in superficial and muscle invasive bladder cancer and its correlation with vascular endothelial growth factor expression and tumour progression, Br. J. Cancer 82 (2000) 161 166. C.A. Nathan, S. Franklin, F.W. Abreo, R. Nassar, A. De Benedetti, J. Williams, F.J. Stucker, Expression of eIF4E during head and neck tumorigenesis: possible role in angiogenesis, Laryngoscope 109 (1999) 1253 1258. C.G. Kevil, A. De Benedetti, D.K. Payne, L.L. Coe, F.S. Laroux, J.S. Alexander, Translational regulation of vascular permeability factor by eukaryotic initiation factor 4E: implications for tumor angiogenesis, Int. J. Cancer 65 (1996) 785 790. J. Rak, Y. Mitsuhashi, C. Sheehan, A. Tamir, A. Viloria-Petit, J. Filmus, S.J. Mansour, N.G. Ahn, R.S. Kerbel, Oncogenes and tumor angiogenesis: differential modes of vascular endothelial growth factor up-regulation in ras-transformed epithelial cells and fibroblasts, Cancer Res. 60 (2000) 490 498. N.M. Mazure, E.Y. Chen, P. Yeh, K.R. Laderoute, A.J. Giaccia, Oncogenic transformation and hypoxia synergistically act to modulate vascular endothelial growth factor expression, Cancer Res. 56 (1996) 3436 3440. G. Breier, S. Blum, J. Peli, M. Groot, C. Wild, W. Risau, E. Reichmann, Transforming growth factor-beta and Ras regulate the VEGF/ VEGF-receptor system during tumor angiogenesis, Int. J. Cancer 97 (2002) 142 148. J. Grunstein, W.G. Roberts, O. Mathieu-Costello, D. Hanahan, R.S. Johnson, Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function, Cancer Res. 59 (1999) 1592 1598. D. Shweiki, A. Itin, D. Soffer, E. Keshet, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis, Nature 359 (1992) 843 845. Y. Seko, K. Tobe, N. Takahashi, Y. Kaburagi, T. Kadowaki, Y. Yazaki, Hypoxia and hypoxia/reoxygenation activate Src family tyrosine kinases and p21ras in cultured rat cardiac myocytes, Biochem. Biophys. Res. Commun. 226 (1996) 530 535.

34

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 [107] A. Agah, T.R. Kyriakides, J. Lawler, P. Bornstein, The lack of thrombospondin-1 (TSP1) dictates the course of wound healing in double-TSP1/TSP2-null mice, Am. J. Pathol. 161 (2002) 831 839. [108] N. Clarke, N. Arenzana, T. Hai, A. Minden, R. Prywes, Epidermal growth factor induction of the c-jun promoter by a Rac pathway, Mol. Cell. Biol. 18 (1998) 1065 1073. [109] M. Kraemer, R. Tournaire, V. Dejong, N. Montreau, D. Briane, C. Derbin, B. Binetruy, Rat embryo fibroblasts transformed by c-Jun display highly metastatic and angiogenic activities in vivo and deregulate gene expression of both angiogenic and antiangiogenic factors, Cell Growth Differ. 10 (1999) 193 200. [110] L.S. Gutierrez, M. Suckow, J. Lawler, V.A. Ploplis, F.J. Castellino, Thrombospondin 1a regulator of adenoma growth and carcinoma progression in the APC(Min/+) mouse model, Carcinogenesis 24 (2003) 199 207. [111] T. Hawighorst, P. Velasco, M. Streit, Y.K. Hong, T.R. Kyriakides, L.F. Brown, P. Bornstein, M. Detmar, Thrombospondin-2 plays a protective role in multistep carcinogenesis: a novel host anti-tumor defense mechanism, EMBO J. 20 (2001) 2631 2640. [112] T. Hawighorst, H. Oura, M. Streit, L. Janes, L. Nguyen, L.F. Brown, G. Oliver, D.G. Jackson, M. Detmar, Thrombospondin-1 selectively inhibits early-stage carcinogenesis and angiogenesis but not tumor lymphangiogenesis and lymphatic metastasis in transgenic mice, Oncogene 21 (2002) 7945 7956. [113] K.J. OByrne, A.G. Dalgleish, Chronic immune activation and inflammation as the cause of malignancy, Br. J. Cancer 85 (2001) 473 483. [114] A.G. Dalgleish, K.J. OByrne, Chronic immune activation and inflammation in the pathogenesis of AIDS and cancer, Adv. Cancer Res. 84 (2002) 231 276. [115] S.M. Prescott, F.A. Fitzpatrick, Cyclooxygenase-2 and carcinogenesis, Biochim. Biophys. Acta 1470 (2000) M69 M78. [116] W.E. Smalley, R.N. DuBois, Colorectal cancer and nonsteroidal antiinflammatory drugs, Adv. Pharmacol. 39 (1997) 1 20. [117] F.M. Giardiello, G.J. Offerhaus, R.N. DuBois, The role of nonsteroidal anti-inflammatory drugs in colorectal cancer prevention, Eur. J. Cancer 31A (1995) 1071 1076. [118] R.N. DuBois, S.B. Abramson, L. Crofford, R.A. Gupta, L.S. Simon, L.B. Van De Putte, P.E. Lipsky, Cyclooxygenase in biology and disease, FASEB J. 12 (1998) 1063 1073. [119] K. Subbaramaiah, A.J. Dannenberg, Cyclooxygenase 2: a molecular target for cancer prevention and treatment, Trends Pharmacol. Sci. 24 (2003) 96 102. [120] E. Fosslien, Molecular pathology of cyclooxygenase-2 in neoplasia, Ann. Clin. Lab. Sci. 30 (2000) 3 21. [121] M.K. Jones, H. Wang, B.M. Peskar, E. Levin, R.M. Itani, I.J. Sarfeh, A.S. Tarnawski, Inhibition of angiogenesis by nonsteroidal anti-inflammatory drugs: insight into mechanisms and implications for cancer growth and ulcer healing, Nat. Med. 5 (1999) 1418 1423. [122] M.M. Taketo, Cyclooxygenase-2 inhibitors in tumorigenesis (Part II), J. Natl. Cancer Inst. 90 (1998) 1609 1620. [123] J.L. Masferrer, K.M. Leahy, A.T. Koki, B.S. Zweifel, S.L. Settle, B.M. Woerner, D.A. Edwards, A.G. Flickinger, R.J. Moore, K. Seibert, Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors, Cancer Res. 60 (2000) 1306 1311. [124] M. Oshima, J.E. Dinchuk, S.L. Kargman, H. Oshima, B. Hancock, E. Kwong, J.M. Trzaskos, J.F. Evans, M.M. Taketo, Suppression of intestinal polyposis in Apc delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2), Cell 87 (1996) 803 809. [125] H.F. Tiano, C.D. Loftin, J. Akunda, C.A. Lee, J. Spalding, A. Sessoms, D.B. Dunson, E.G. Rogan, S.G. Morham, R.C. Smart, R. Langenbach, Deficiency of either cyclooxygenase (COX)-1 or COX2 alters epidermal differentiation and reduces mouse skin tumorigenesis, Cancer Res. 62 (2002) 3395 3401. [126] P.C. Chulada, M.B. Thompson, J.F. Mahler, C.M. Doyle, B.W. Gaul, C. Lee, H.F. Tiano, S.G. Morham, O. Smithies, R. Langenbach, Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces

[88] C. Li, R.M. Jackson, Reactive species mechanisms of cellular hypoxia-reoxygenation injury, Am. J. Physiol., Cell Physiol. 282 (2002) C227 C241. [89] H.M. Lander, J.S. Ogiste, K.K. Teng, A. Novogrodsky, p21ras as a common signaling target of reactive free radicals and cellular redox stress, J. Biol. Chem. 270 (1995) 21195 21198. [90] H.M. Lander, J.S. Ogiste, S.F. Pearce, R. Levi, A. Novogrodsky, Nitric oxide-stimulated guanine nucleotide exchange on p21ras, J. Biol. Chem. 270 (1995) 7017 7020. [91] G.U. Dachs, G.M. Tozer, Hypoxia modulated gene expression: angiogenesis, metastasis and therapeutic exploitation, Eur. J. Cancer 36 (2000) 1649 1660. [92] N. Ghiso, R.M. Rohan, S. Amano, R. Garland, A.P. Adamis, Suppression of hypoxia-associated vascular endothelial growth factor gene expression by nitric oxide via cGMP, Invest. Ophthalmol. Visual Sci. 40 (1999) 1033 1039. [93] D. Mukhopadhyay, L. Tsiokas, X.M. Zhou, D. Foster, J.S. Brugge, V.P. Sukhatme, Hypoxic induction of human vascular endothelial growth factor expression through c-Src activation, Nature 375 (1995) 577 581. [94] G. Schafer, T. Cramer, G. Suske, W. Kemmner, B. Wiedenmann, M. Hocker, Oxidative stress regulates vascular endothelial growth factor-A gene transcription through Sp1- and Sp3-dependent activation of two proximal GC-rich promoter elements, J. Biol. Chem. 278 (2003) 8190 8198. [95] M.M. Feldkamp, N. Lau, J. Rak, R.S. Kerbel, A. Guha, Normoxic and hypoxic regulation of vascular endothelial growth factor (VEGF) by astrocytoma cells is mediated by Ras, Int. J. Cancer 81 (1999) 118 124. [96] L. Xu, D. Fukumura, R.K. Jain, Acidic extracellular pH induces vascular endothelial growth factor (VEGF) in human glioblastoma cells via ERK1/2 MAPK signaling pathway: mechanism of low pHinduced VEGF, J. Biol. Chem. 277 (2002) 11368 11374. [97] D. Fukumura, L. Xu, Y. Chen, T. Gohongi, B. Seed, R.K. Jain, Hypoxia and acidosis independently up-regulate vascular endothelial growth factor transcription in brain tumors in vivo, Cancer Res. 61 (2001) 6020 6024. [98] A. Mettouchi, F. Cabon, N. Montreau, P. Vernier, G. Mercier, D. Blangy, H. Tricoire, P. Vigier, B. Binetruy, SPARC and thrombospondin genes are repressed by the c-jun oncogene in rat embryo fibroblasts, EMBO J. 13 (1994) 5668 5678. [99] V. Zabrenetzky, C.C. Harris, P.S. Steeg, D.D. Roberts, Expression of the extracellular matrix molecule thrombospondin inversely correlates with malignant progression in melanoma, lung and breast carcinoma cell lines, Int. J. Cancer 59 (1994) 191 195. [100] N. Sheibani, W.A. Frazier, Repression of thrombospondin-1 expression, a natural inhibitor of angiogenesis, in polyoma middle T transformed NIH3T3 cells, Cancer Lett. 107 (1996) 45 52. [101] J. Lawler, Thrombospondin-1 as an endogenous inhibitor of angiogenesis and tumor growth, J. Cell. Mol. Med. 6 (2002) 1 12. [102] O.V. Volpert, S.S. Tolsma, S. Pellerin, J.J. Feige, H. Chen, D.F. Mosher, N. Bouck, Inhibition of angiogenesis by thrombospondin2, Biochem. Biophys. Res. Commun. 217 (1995) 326 332. [103] J.C. Adams, Thrombospondins: multifunctional regulators of cell interactions, Annu. Rev. Cell Dev. Biol. 17 (2001) 25 51. [104] D.J. Good, P.J. Polverini, F. Rastinejad, M.M. Le Beau, R.S. Lemons, W.A. Frazier, N.P. Bouck, A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin, Proc. Natl. Acad. Sci. U. S. A. 87 (1990) 6624 6628. [105] S. Filleur, O.V. Volpert, A. Degeorges, C. Voland, F. Reiher, P. Clezardin, N. Bouck, F. Cabon, In vivo mechanisms by which tumors producing thrombospondin 1 bypass its inhibitory effects, Genes Dev. 15 (2001) 1373 1382. [106] S.E. Crawford, V. Stellmach, J.E. Murphy-Ullrich, S.M. Ribeiro, J. Lawler, R.O. Hynes, G.P. Boivin, N. Bouck, Thrombospondin-1 is a major activator of TGF-beta1 in vivo, Cell 93 (1998) 1159 1170.

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 intestinal tumorigenesis in Min mice, Cancer Res. 60 (2000) 4705 4708. M.K. Jones, I.L. Szabo, H. Kawanaka, S.S. Husain, A.S. Tarnawski, von Hippel Lindau tumor suppressor and HIF-1alpha: new targets of NSAIDs inhibition of hypoxia-induced angiogenesis, FASEB J. 16 (2002) 264 266. X. Zhang, S.G. Morham, R. Langenbach, D.A. Young, Malignant transformation and antineoplastic actions of nonsteroidal antiinflammatory drugs (NSAIDs) on cyclooxygenase-null embryo fibroblasts, J. Exp. Med. 190 (1999) 451 459. D.J. Elder, C. Paraskeva, NSAIDs to prevent colorectal cancer: a question of sensitivity, Gastroenterology 113 (1997) 1999 2003. W. Dempke, C. Rie, A. Grothey, H.J. Schmoll, Cyclooxygenase-2: a novel target for cancer chemotherapy? J. Cancer Res. Clin. Oncol. 127 (2001) 411 417. M. Tsujii, S. Kawano, S. Tsuji, H. Sawaoka, M. Hori, R.N. DuBois, Cyclooxygenase regulates angiogenesis induced by colon cancer cells, Cell 93 (1998) 705 716. O. Dormond, A. Foletti, C. Paroz, C. Ruegg, NSAIDs inhibit alpha V beta 3 integrin-mediated and Cdc42/Rac-dependent endothelialcell spreading, migration and angiogenesis, Nat. Med. 7 (2001) 1041 1047. M.M. Hoper, N.F. Voelkel, T.O. Bates, J.D. Allard, M. Horan, D. Shepherd, R.M. Tuder, Prostaglandins induce vascular endothelial growth factor in a human monocytic cell line and rat lungs via cAMP, Am. J. Respir. Cell Mol. Biol. 17 (1997) 748 756. D.M. Form, R. Auerbach, PGE2 and angiogenesis, Proc. Soc. Exp. Biol. Med. 172 (1983) 214 218. M. Ziche, J. Jones, P.M. Gullino, Role of prostaglandin E1 and copper in angiogenesis, J. Natl. Cancer Inst. 69 (1982) 475 482. E. Spisni, F. Manica, V. Tomasi, Involvement of prostanoids in the regulation of angiogenesis by polypeptide growth factors, Prostaglandins Leukot. Essent. Fat. Acids 47 (1992) 111 115. T.O. Daniel, H. Liu, J.D. Morrow, B.C. Crews, L.J. Marnett, Thromboxane A2 is a mediator of cyclooxygenase-2-dependent endothelial migration and angiogenesis, Cancer Res. 59 (1999) 4574 4577. S. Harada, J.A. Nagy, K.A. Sullivan, K.A. Thomas, N. Endo, G.A. Rodan, B. Rodan, Induction of vascular endothelial growth factor expression by prostaglandin E2 and E1 in osteoblasts, J. Clin. Invest. 93 (1994) 2490 2496. M.M. Hoper, N.F. Voelkel, T.O. Bates, J.D. Allard, M. Horan, D. Shepherd, R.M. Tuder, Prostaglandins induce vascular endothelial growth factor in a human monocytic cell line and rat lungs via cAMP, Am. J. Respir. Cell Mol. Biol. 17 (1997) 748 756. M.E. Pueyo, Y. Chen, G. DAngelo, J.B. Michel, Regulation of vascular endothelial growth factor expression by cAMP in rat aortic smooth muscle cells, Exp. Cell Res. 238 (1998) 354 358. T. Cheng, W. Cao, R. Wen, R.H. Steinberg, M.M. LaVail, Prostaglandin E2 induces vascular endothelial growth factor and basic fibroblast growth factor mRNA expression in cultured rat Muller cells, Invest. Ophthalmol. Visual Sci. 39 (1998) 581 591. M. Sonoshita, K. Takaku, N. Sasaki, Y. Sugimoto, F. Ushikubi, S. Narumiya, M. Oshima, M.M. Taketo, Acceleration of intestinal polyposis through prostaglandin receptor EP2 in Apc(Delta 716) knockout mice, Nat. Med. 7 (2001) 1048 1051. C.S. Williams, M. Tsujii, J. Reese, S.K. Dey, R.N. DuBois, Host cyclooxygenase-2 modulates carcinoma growth, J. Clin. Invest. 105 (2000) 1589 1594. H. Amano, I. Hayashi, H. Endo, H. Kitasato, S. Yamashina, T. Maruyama, M. Kobayashi, K. Satoh, M. Narita, Y. Sugimoto, T. Murata, H. Yoshimura, S. Narumiya, M. Majima, Host prostaglandin E(2)-EP3 signaling regulates tumor-associated angiogenesis and tumor growth, J. Exp. Med. 20 (197) (2003) 221 232. H. Seno, M. Oshima, T.O. Ishikawa, H. Oshima, K. Takaku, T. Chiba, S. Narumiya, M.M. Taketo, Cyclooxygenase 2- and prostaglandin E(2) receptor EP(2)-dependent angiogenesis in Apc(Delta716) mouse intestinal polyps, Cancer Res. 62 (2002) 506 511.

35

[127]

[128]

[129] [130]

[131]

[132]

[133]

[134] [135] [136]

[137]

[138]

[139]

[140]

[141]

[142]

[143]

[144]

[145]

[146] L.R. Howe, H.C. Crawford, K. Subbaramaiah, J.A. Hassell, A.J. Dannenberg, A.M. Brown, PEA3 is up-regulated in response to Wnt1 and activates the expression of cyclooxygenase-2, J. Biol. Chem. 276 (2001) 20108 20115. [147] Y. Araki, S. Okamura, S.P. Hussain, M. Nagashima, P. He, M. Shiseki, K. Miura, C.C. Harris, Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways, Cancer Res. 63 (2003) 728 734. [148] K. Subbaramaiah, N. Altorki, W.J. Chung, J.R. Mestre, A. Sampat, A.J. Dannenberg, Inhibition of cyclooxygenase-2 gene expression by p53, J. Biol. Chem. 274 (1999) 10911 10915. [149] K. Subbaramaiah, L. Norton, W. Gerald, A.J. Dannenberg, Cyclooxygenase-2 is overexpressed in HER-2/neu-positive breast cancer: evidence for involvement of AP-1 and PEA3, J. Biol. Chem. 277 (2002) 18649 18657. [150] Y.S. Ji, Q. Xu, J.F. Schmedtje Jr., Hypoxia induces high-mobilitygroup protein I(Y) and transcription of the cyclooxygenase-2 gene in human vascular endothelium, Circ. Res. 83 (1998) 295 304. [151] K. Subbaramaiah, N. Telang, J.T. Ramonetti, R. Araki, B. DeVito, B.B. Weksler, A.J. Dannenberg, Transcription of cyclooxygenase-2 is enhanced in transformed mammary epithelial cells, Cancer Res. 56 (1996) 4424 4429. [152] H. Sheng, C.S. Williams, J. Shao, P. Liang, R.N. DuBois, R.D. Beauchamp, Induction of cyclooxygenase-2 by activated Ha-ras oncogene in Rat-1 fibroblasts and the role of mitogen-activated protein kinase pathway, J. Biol. Chem. 273 (1998) 22120 22127. [153] W. Xie, B.S. Fletcher, R.D. Andersen, H.R. Herschman, v-src induction of the TIS10/PGS2 prostaglandin synthase gene is mediated by an ATF/CRE transcription response element, Mol. Cell. Biol. 14 (1994) 6531 6539. [154] W. Xie, H.R. Herschman, v-src induces prostaglandin synthase 2 gene expression by activation of the c-Jun N-terminal kinase and the c-Jun transcription factor, J. Biol. Chem. 270 (1995) 27622 27628. [155] W. Xie, H.R. Herschman, Transcriptional regulation of prostaglandin synthase 2 gene expression by platelet-derived growth factor and serum, J. Biol. Chem. 271 (1996) 31742 31748. [156] S.T. Reddy, D.J. Wadleigh, H.R. Herschman, Transcriptional regulation of the cyclooxygenase-2 gene in activated mast cells, J. Biol. Chem. 275 (2000) 3107 3113. [157] R.C. OHagan, R.G. Tozer, M. Symons, F. McCormick, J.A. Hassell, The activity of the Ets transcription factor PEA3 is regulated by two distinct MAPK cascades, Oncogene 19 (13) (1996) 1323 1333. [158] E. Lengyel, E. Stepp, R. Gum, D. Boyd, Involvement of a mitogenactivated protein kinase signaling pathway in the regulation of urokinase promoter activity by c-Ha-ras, J. Biol. Chem. 270 (1995) 23007 23012. [159] S. Silberman, M. Janulis, R.M. Schultz, Characterization of downstream Ras signals that induce alternative protease-dependent invasive phenotypes, J. Biol. Chem. 272 (1997) 5927 5935. [160] H. Sheng, J. Shao, R.N. DuBois, K-Ras-mediated increase in cyclooxygenase 2 mRNA stability involves activation of the protein kinase B1, Cancer Res. 61 (2001) 2670 2675. [161] L.R. Howe, H.C. Crawford, K. Subbaramaiah, J.A. Hassell, A.J. Dannenberg, A.M. Brown, PEA3 is up-regulated in response to Wnt1 and activates the expression of cyclooxygenase-2, J. Biol. Chem. 276 (2001) 20108 20115. [162] F.J. Giles, H.M. Kantarjian, B.N. Bekele, J.E. Cortes, S. Faderl, D.A. Thomas, T. Manshouri, A. Rogers, M.J. Keating, M. Talpaz, S. OBrien, M. Albitar, Bone marrow cyclooxygenase-2 levels are elevated in chronic-phase chronic myeloid leukaemia and are associated with reduced survival, Br. J. Haematol. 119 (2002) 38 45. [163] J.F. Schmedtje Jr., Y.S. Ji, W.L. Liu, R.N. DuBois, M.S. Runge, Hypoxia induces cyclooxygenase-2 via the NF-kappaB p65 transcription factor in human vascular endothelial cells, J. Biol. Chem. 272 (1997) 601 608. [164] C.S. Williams, R.N. DuBois, Prostaglandin endoperoxide synthase: why two isoforms? Am. J. Physiol. 270 (1996) G393 G400.

36

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 [185] D.B. Rifkin, D. Moscatelli, J. Bizik, N. Quarto, F. Blei, P. Dennis, R. Flaumenhaft, P. Mignatti, Growth factor control of extracellular proteolysis, Cell Differ. Dev. 32 (1990) 313 318. [186] D.B. Rifkin, R. Mazzieri, J.S. Munger, I. Noguera, J. Sung, Proteolytic control of growth factor availability, APMIS 107 (1999) 80 85. [187] I. Stamenkovic, Matrix metalloproteinases in tumor invasion and metastasis, Semin. Cancer Biol. 10 (2000) 415 433. [188] G. Bergers, R. Brekken, G. McMahon, T.H. Vu, T. Itoh, K. Tamaki, K. Tanzawa, P. Thorpe, S. Itohara, Z. Werb, D. Hanahan, Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis, Nat. Cell Biol. 2 (2000) 737 744. [189] J. Berkelhammer, R.W. Oxenhandler, R.R. Hook Jr., J.M. Hennessy, Development of a new melanoma model in C57BL/6 mice, Cancer Res. 42 (1982) 3157 3163. [190] G.J. Walker, N.K. Hayward, Pathways to melanoma development: lessons from the mouse, J. Invest. Dermatol. 119 (2002) 783 792. [191] R.L. Shapiro, J.G. Duquette, D.F. Roses, I. Nunes, M.N. Harris, H. Kamino, E.L. Wilson, D.B. Rifkin, Induction of primary cutaneous melanocytic neoplasms in urokinase-type plasminogen activator (uPA)-deficient and wild-type mice: cellular blue nevi invade but do not progress to malignant melanoma in uPA-deficient animals, Cancer Res. 56 (1996) 3597 3604. [192] H.J. Park, D. Kong, L. Iruela-Arispe, U. Begley, D. Tang, J.B. Galper, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors interfere with angiogenesis by inhibiting the geranylgeranylation of RhoA, Circ. Res. 91 (2002) 143 150. [193] L. Vincent, W. Chen, L. Hong, F. Mirshahi, Z. Mishal, T. MirshahiKhorassani, J.P. Vannier, J. Soria, C. Soria, Inhibition of endothelial cell migration by cerivastatin, an HMG-CoA reductase inhibitor: contribution to its anti-angiogenic effect, FEBS Lett. 495 (2001) 159 166. [194] L. Vincent, C. Soria, F. Mirshahi, P. Opolon, Z. Mishal, J.P. Vannier, J. Soria, L. Hong, Cerivastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme a reductase, inhibits endothelial cell proliferation induced by angiogenic factors in vitro and angiogenesis in in vivo models, Arterioscler. Thromb. Vasc. Biol. 22 (2002) 623 629. [195] W. Feleszko, E.Z. Balkowiec, E. Sieberth, M. Marczak, A. Dabrowska, A. Giermasz, A. Czajka, M. Jakobisiak, Lovastatin and tumor necrosis factor-alpha exhibit potentiated antitumor effects against Ha-ras-transformed murine tumor via inhibition of tumor-induced angiogenesis, Int. J. Cancer 81 (1999) 560 567. [196] M.M. Feldkamp, N. Lau, A. Guha, Growth inhibition of astrocytoma cells by farnesyl transferase inhibitors is mediated by a combination of anti-proliferative, pro-apoptotic and anti-angiogenic effects, Oncogene 18 (1999) 7514 7526. [197] W.Z. Gu, S.K. Tahir, Y.C. Wang, H.C. Zhang, S.P. Cherian, S. OConnor, J.A. Leal, S.H. Rosenberg, S.C. Ng, Effect of novel CAAX peptidomimetic farnesyltransferase inhibitor on angiogenesis in vitro and in vivo, Eur. J. Cancer 35 (1999) 1394 1401. [198] S. Charvat, M. Duchesne, P. Parvaz, M.C. Chignol, D. Schmitt, M. Serres, The up-regulation of vascular endothelial growth factor in mutated Ha-ras HaCaT cell lines is reduced by a farnesyl transferase inhibitor, Anticancer Res. 19 (1999) 557 561. [199] S.K. Tahir, W.Z. Gu, H.C. Zhang, J. Leal, J.Y. Lee, P. Kovar, B. Saeed, S.P. Cherian, E. Devine, J. Cohen, R. Warner, Y.C. Wang, D. Stout, D.L. Arendsen, S. Rosenberg, S.C. Ng, Inhibition of farnesyltransferase with A-176120, a novel and potent farnesyl pyrophosphate analogue, Eur. J. Cancer 36 (2000) 1161 1170. [200] B. Zhang, G.C. Prendergast, R.G. Fenton, Farnesyltransferase inhibitors reverse Ras-mediated inhibition of Fas gene expression, Cancer Res. 62 (2002) 450 458. [201] J.E. Karp, J.E. Lancet, S.H. Kaufmann, D.W. End, J.J. Wright, K. Bol, I. Horak, M.L. Tidwell, J. Liesveld, T.J. Kottke, D. Ange, L. Buddharaju, I. Gojo, W.E. Highsmith, R.T. Belly, R.J. Hohl, M.E. Rybak, A. Thibault, J. Rosenblatt, Clinical and biologic activity of

[165] K. Ise, K. Nakamura, K. Nakao, S. Shimizu, H. Harada, T. Ichise, J. Miyoshi, Y. Gondo, T. Ishikawa, A. Aiba, M. Katsuki, Targeted deletion of the H-ras gene decreases tumor formation in mouse skin carcinogenesis, Oncogene 19 (2000) 2951 2956. [166] A. Berns, Turning on tumors to study cancer progression, Nat. Med. 5 (1999) 989 990. [167] A. Berns, Cancer. Improved mouse models, Nature 410 (2001) 1043 1044. [168] G. Brunner, J. Pohl, L.J. Erkell, A. Radler-Pohl, V. Schirrmacher, Induction of urokinase activity and malignant phenotype in bladder carcinoma cells after transfection of the activated Ha-ras oncogene, J. Cancer Res. Clin. Oncol. 115 (1989) 139 144. [169] J.E. Testa, R.L. Medcalf, J.F. Cajot, W.D. Schleuning, B. Sordat, Urokinase-type plasminogen activator biosynthesis is induced by the EJ-Ha-ras oncogene in CL26 mouse colon carcinoma cells, Int. J. Cancer 43 (1989) 816 822. [170] R. Gum, E. Lengyel, J. Juarez, J.H. Chen, H. Sato, M. Seiki, D. Boyd, Stimulation of 92-kDa gelatinase B promoter activity by ras is mitogen-activated protein kinase kinase 1-independent and requires multiple transcription factor binding sites including closely spaced PEA3/ ets and AP-1 sequences, J. Biol. Chem. 271 (1996) 10672 10680. [171] H. Sato, Y. Kida, M. Mai, Y. Endo, T. Sasaki, J. Tanaka, M. Seiki, Expression of genes encoding type IV collagen-degrading metalloproteinases and tissue inhibitors of metalloproteinases in various human tumor cells, Oncogene 7 (1992) 77 83. [172] M.S. Pepper, Role of the matrix metalloproteinase and plasminogen activator-plasmin systems in angiogenesis, Arterioscler. Thromb. Vasc. Biol. 21 (2001) 1104 1117. [173] M.J. Duffy, T.M. Maguire, E.W. McDermott, N. OHiggins, Urokinase plasminogen activator: a prognostic marker in multiple types of cancer, J. Surg. Oncol. 71 (1999) 130 135. [174] H.R. Lijnen, Plasmin and matrix metalloproteinases in vascular remodeling, Thromb. Haemost. 86 (2001) 324 333. [175] V. Ellis, C. Pyke, J. Eriksen, H. Solberg, K. Dano, The urokinase receptor: involvement in cell surface proteolysis and cancer invasion, Ann. N.Y. Acad. Sci. 667 (1992) 13 31. [176] F. Blasi, Proteolysis, cell adhesion, chemotaxis, and invasiveness are regulated by the u-PA-u-PAR-PAI-1 system, Thromb. Haemost. 82 (1999) 298 304. [177] F. Blasi, P. Carmeliet, uPAR: a versatile signalling orchestrator, Nat. Rev., Mol. Cell Biol. 3 (2002) 932 943. [178] S. Ried, C. Jager, M. Jeffers, G.F. Vande Woude, H. Graeff, M. Schmitt, E. Lengyel, Activation mechanisms of the urokinase-type plasminogen activator promoter by hepatocyte growth factor/scatter factor, J. Biol. Chem. 274 (1999) 16377 16386. [179] R.M. Wolthuis, J.L. Bos, Ras caught in another affair: the exchange factors for Ral, Curr. Opin. Genet. Dev. 9 (1999) 112 117. [180] J.A. Aguirre-Ghiso, P. Frankel, E.F. Farias, Z. Lu, H. Jiang, A. Olsen, L.A. Feig, E.B. Kier Joffe, D.A. Foster, RalA requirement for v-Src- and v-Ras-induced tumorigenicity and overproduction of urokinase-type plasminogen activator: involvement of metalloproteases, Oncogene 19 (18) (1999) 4718 4725. [181] J.Q. Yang, W. Zhao, H. Duan, M.E. Robbins, G.R. Buettner, L.W. Oberley, F.E. Domann, v-Ha-RaS oncogene upregulates the 92-kDa type IV collagenase (MMP-9) gene by increasing cellular superoxide production and activating NF-kappaB, Free Radic. Biol. Med. 31 (2001) 520 529. [182] Y. Jiang, R.J. Muschel, Regulation of matrix metalloproteinase-9 (MMP-9) by translational efficiency in murine prostate carcinoma cells, Cancer Res. 62 (2002) 1910 1914. [183] P. Carmeliet, D. Collen, Development and disease in proteinasedeficient mice: role of the plasminogen, matrix metalloproteinase and coagulation system, Thromb. Res. 91 (1998) 255 285. [184] K.A. Houck, D.W. Leung, A.M. Rowland, J. Winer, N. Ferrara, Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms, J. Biol. Chem. 267 (1992) 26031 26037.

O. Kranenburg et al. / Biochimica et Biophysica Acta 1654 (2004) 2337 the farnesyltransferase inhibitor R115777 in adults with refractory and relapsed acute leukemias: a phase 1 clinical-laboratory correlative trial, Blood 97 (2001) 3361 3369. [202] J.E. Karp, S.H. Kaufmann, A.A. Adjei, J.E. Lancet, J.J. Wright, D.W. End, Current status of clinical trials of farnesyltransferase inhibitors, Curr. Opin. Oncol. 13 (2001) 470 476. [203] M.M. Feldkamp, N. Lau, L. Roncari, A. Guha, Isotype-specific Ras.GTP-levels predict the efficacy of farnesyl transferase inhibitors

37

against human astrocytomas regardless of Ras mutational status, Cancer Res. 61 (2001) 4425 4431. [204] P.F. Lebowitz, G.C. Prendergast, Non-Ras targets of farnesyltransferase inhibitors: focus on Rho, Oncogene 17 (1998) 1439 1445. [205] A.D. Cox, C.J. Der, Farnesyltransferase inhibitors and cancer treatment: targeting simply Ras? Biochim. Biophys. Acta 1333 (1997) F51 F71.

Anda mungkin juga menyukai