[CANCER RESEARCH 60, 203–212, January 15, 2000] Review

Regulation of via Vascular Endothelial Receptors1

Tanja Veikkola, Marika Karkkainen, Lena Claesson-Welsh, and Kari Alitalo2 Molecular/Cancer Biology Laboratory, Haartman Institute, University of Helsinki, SF-00014 Helsinki, Finland [T. V., M. K., K. A.], and Department of Medical Biochemistry and Microbiology, Biomedical Center, S-75123 Uppsala, Sweden [L. C-W.] Introduction

Endothelial cell signal transduction mechanisms involved in angio- with several different isoforms generated by alternative splicing, genesis have come into focus in cancer research when it was realized consisting of polypeptides of 121, 145, 165, 189, or 206 amino acid that solid tumors are dependent on neovascularization (1). Unlike residues (15, 24–26). VEGF binds both VEGFR-1 and VEGFR-2 and normal human endothelial cells, which are quiescent except in the is apparently capable of inducing heterodimers between the two reproductive organs of fertile women, endothelial cells in tumors (27–30). VEGF uses symmetrical binding sites at each pole of the express several target features associated with their angiogenic acti- dimer for receptor binding (31, 32). It has been shown that the second vation. On the other hand, unlike the tumor cells, endothelial cells are immunoglobulin homology domain of VEGFR-1 is critical for ligand readily accessible from the blood circulation, and they are not likely binding, but the first three immunoglobulin domains are required to to develop resistant variants to cytostatic therapy (see Ref. 2). Vas- reconstitute full affinity (31, 33–35). Of the other VEGF family cular targeting may involve either the destruction of existing vessels members, PlGF and VEGF-B bind only VEGFR-1, the Orf virus by exploitation of differences between normal and tumor vessels VEGFs bind only VEGFR-2, and VEGF-C and VEGF-D interact with (3–6) or inhibition of the tumor angiogenesis process per se (7–9). In both VEGFR-2 and VEGFR-3 (16–18, 36–42). Recently, NRP-1, a both scenarios, knowledge of the endothelial cell-specific growth cell surface glycoprotein that acts as a receptor for collapsins/sema- factor receptors and their signal transduction and effector mechanisms phorins and controls axon guidance during embryonic development, is essential and will undoubtedly provide additional points of attack to has been identified as an isoform-specific receptor for VEGF165, human cancers. Here we discuss recent results on one important PlGF-2, VEGF-B, and Orf virus VEGFs (39, 43–45). Heterodimers of family of endothelial growth factor receptors implicated in angiogen- PlGF and VEGF are produced by certain cell lines, and they exert esis. Several excellent reviews have appeared on the same topic and mitogenic activity toward endothelial cells (46, 47). VEGF-B/VEGF related topics (10–14). heterodimers have also been obtained in expression vector cotrans- The VEGF3 family currently includes five members in addition to fection experiments (48). In addition to the high-affinity receptors, the prototype VEGF, namely, PlGF, VEGF-B, VEGF-C, VEGF-D, certain splice isoforms of the VEGFs also bind heparan sulfate pro- and Orf virus VEGFs (also called VEGF-E; Refs. 15–18 and Refs. teoglycans on the cell surface and in the pericellular matrix via a below). The VEGFs mediate angiogenic signals to the vascular en- distinct heparin binding domain (16, 49–51). dothelium via high-affinity RTKs. To date, three receptors for the VEGFR-1 is a Mr 180,000 transmembrane glycoprotein, but its VEGFs have been identified. All three are relatively specific for mRNA can also be spliced to produce a shorter soluble endothelial cells and demonstrate structural and functional similarities consisting of only the first six extracellular immunoglobulin homol- to the PDGF receptor family (see Refs. 14 and 19). These receptors ogy domains (20, 27, 52). Such a RNA splice variant, originally are currently designated VEGFR-1, VEGFR-2, and VEGFR-3 and detected in a HUVEC cDNA library, encodes 31 unique amino acid were originally named flt (fms-like ), KDR (kinase residues before a stop codon (52). VEGFR-2 is a Mr 230,000 protein, insert domain-containing receptor)/flk-1 (fetal liver kinase-1), and and no splice variants have been reported for this receptor. In human Ј FLT4, respectively (11, 12, 20–23). All have seven immunoglobulin VEGFR-3, alternative 3 polyadenylation signals result in a 4.5-kb homology domains in their extracellular part and an intracellular transcript and a more prevalent 5.8-kb transcript (53). The latter tyrosine kinase signaling domain split by a kinase insert (Fig. 1). In transcript encodes 65 additional amino acid residues and is the major form detected in tissues. After biosynthesis, the glycosylated M adults, VEGFR-1 and VEGFR-2 are expressed mainly in the blood r vascular endothelium, whereas VEGFR-3 is restricted largely to the 195,000 VEGFR-3 is proteolytically cleaved in the fifth immunoglob- ulin homology domain, but the resulting M 120,000 and M 75,000 lymphatic endothelium. r r chains remain linked by a disulfide bond (41, 54). The VEGF molecule is an antiparallel disulfide-linked homodimer Because hypoxia is a major inducer of VEGF expression in tumors, and the VEGFRs are enhanced in tumor endothelia (see Fig. 2), Received 4/2/99; accepted 11/15/99. The costs of publication of this article were defrayed in part by the payment of page hypoxic regulation of the VEGFR genes has been studied by several charges. This article must therefore be hereby marked advertisement in accordance with groups. In vivo, VEGFR-1 and VEGFR-2 appear to be up-regulated 18 U.S.C. Section 1734 solely to indicate this fact. by hypoxia (55–57). In vitro, VEGFR-1 expression was increased by 1 Supported by the Finnish Academy of Sciences, The University of Helsinki, the Finnish and Swedish Cancer Research Foundation, the Ludwig Institute for Cancer hypoxia, whereas VEGFR-2 was either down-regulated or not af- Research and the Novo Nordisk Foundation. fected (58–60). On the other hand, culture medium from hypoxic 2 To whom requests for reprints should be addressed, at Molecular/Cancer Biology Laboratory, Haartman Institute, University of Helsinki, SF-000014 Helsinki, Finland. cells up-regulated VEGFR-2 protein (59). The latter effect was prob- 3 The abbreviations used are: VEGF, vascular endothelial growth factor; VEGFR, ably mediated by VEGF in the culture medium because VEGF treat- VEGF receptor; PlGF, placenta growth factor; RTK, ; PDGF, ment has been shown to up-regulate VEGFR-2 expression in endo- platelet-derived growth factor; NRP-1, neuropilin-1; HUVEC, human umbilical vein endothelial cell; HIF, hypoxia-inducible factor; MAPK, mitogen-activated ; thelial cells and in cerebral slice cultures (61, 62). The promoter for PKC, protein kinase C; PLC, phospholipase C; PI3-K, phosphatidylinositol 3Ј-kinase; VEGFR-1 contains sequences matching the HIF-1␣ consensus bind- STAT, signal transducer and activator of transcription; KS, Kaposi’s sarcoma; VE- ␣ cadherin, vascular endothelial cadherin; ES, embryonic stem; VHL, von Hippel-Lindau; ing site, whereas the closely related HIF-2 may stimulate VEGFR-2 HHV-8, human herpes virus-8/KS herpes virus; Ang, . promoter activity (60, 63, 64). 203

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

Fig. 1. The currently known VEGFs and their receptors. VEGFR-1 and VEGFR-2 have seven extracellular immunoglobulin homology domains, but in VEGFR-3, the fifth immunoglobulin domain is cleaved on receptor processing into disulfide-linked sub- units. VEGFR-1 and VEGFR-2 mediate angiogenesis, whereas VEGFR-3 is also involved in lymphangiogenesis. NRP-1 binds to specific COOH-terminal sequences present only in certain VEGFs ␣ ␤ that bind to VEGFR-1 and/or VEGFR-2. v 3 and VE- cadherin have been found in complexes with activated VEGFR-2, and the latter also associates with an activated VEGFR-3 complex. sVEGFR-1, soluble VEGFR-1; VEC, VE-Cadherin.

VEGFR Signaling inhibitors of nitric oxide synthase, suggesting that nitric oxide con- tributes to MAPK activation (75). The role of Ras in the VEGFR- Like other RTKs, the VEGFRs are thought to dimerize and undergo MAPK pathway remains to be elucidated. In primary endothelial trans-autophosphorylation on ligand binding. Both VEGFR-2 and cells, Ras was not activated in response to VEGF, and MAPK acti- VEGFR-3 are tyrosine phosphorylated when stimulated with their vation was mediated mainly via a PKC-dependent pathway (74). PKC respective ligands (40, 65), but VEGFR-1 autophosphorylation is less has also been implicated in VEGF-induced MAPK activation in obvious and has been studied mostly in receptor-overexpressing trans- VEGFR-2-transfected fibroblasts (71). Seetharam et al. (66) have fected cells (37, 65–67). Phosphorylated tyrosine residues may serve to control the kinase activity of the receptor and to create docking sites observed activation of the Ras guanine nucleotide exchange factor Sos for cytoplasmic signaling molecules, which provide substrates for the in VEGF-stimulated endothelial cells, but phosphorylation of the kinase. These molecules, either adapters or themselves, link adapter protein Shc was barely detectable. On the other hand, Shc VEGFRs to the signaling pathways that are discussed below. phosphorylation in response to VEGF stimulation has been detected in VEGFR-1 and VEGFR-2. Activation of the MAPK pathway in porcine aortic endothelial cells overexpressing VEGFR-2 (69), and response to VEGF has been observed in many types of endothelial Sck, a Shc homologue, has been shown to interact with the VEGFR-2 cells (66–74). Interestingly, MAPK activation was delayed in cytoplasmic domain in the yeast two-hybrid system (76). Several VEGFR-2-transfected fibroblasts, and the mitogenic response was investigators have reported phosphorylation of the Ras GTPase-acti- weaker than in endothelial cells, suggesting the involvement of cell vating protein in endothelial cells after VEGF stimulation (65, 66, 77). type-specific signaling mechanism(s) (71). In postcapillary venular Ras GTPase-activating protein activation has also been observed in endothelium, VEGF-mediated induction of MAPK was blocked by VEGFR-1-transfected fibroblasts and porcine aortic endothelial cells,

Fig. 2. Immunostaining of VEGFRs in the vascu- lar endothelium of a melanoma metastasis. Adjacent sections were stained with VEGFR-1, VEGFR-2, VEGFR-3, and control antibodies. Arrows point to endothelial cells. Anti-VEGFR-1 and anti-VEGFR-2 were kind gifts from Dr. Herbert Weich.

204

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs but these cells showed no clear MAPK activation or proliferation in VEGF may require the association of VEGFR-2 with cell surface response to VEGF (65, 66). Interestingly, activation of MAPK in adhesive . Activated VEGFR-2 was found in a complex with ␣ ␤ bovine brain capillary endothelial cells by VEGF was selectively integrin v 3, an adhesion molecule specifically expressed on angio- ␣ ␤ inhibited by the Mr 16,000 NH2-terminal fragment of prolactin, an genic endothelium (93). v 3 binds to pericellular matrix proteins antiangiogenic factor that also inhibited proliferation of these cells containing an arginine-glycine-aspartic acid (RGD) peptide motif, ␣ ␤ (68, 78). In addition to MAPK, VEGF has been shown to activate the such as vitronectin and fibronectin. v 3 has been shown to be p38 stress kinase pathway in HUVECs, and this response has been involved in the regulation of the cell cycle and the survival of linked to F-actin reorganization and cell migration (72). endothelial cells (94–96). Various antagonists of this integrin, such as The PLC-␥-PKC pathway has also been implicated in the mitogenic antibodies and cyclic binding peptides, as well as cytokines that action of VEGF. VEGFR-1 interacts with the PLC-␥ SH2 domain in ␣ ␤ interfere with v 3 activity, are capable of inhibiting tumor angio- the yeast two-hybrid assay (79), and several groups have reported that genesis (97). VEGFR-2 tyrosine phosphorylation and VEGF-induced VEGF induces the phosphorylation and activation of PLC-␥ (66–68, mitogenicity were enhanced when endothelial cells were plated on the 70, 71, 73, 77, 80), leading to the hydrolysis of phosphatidylinositol ␣ ␤ v 3 ligand vitronectin, demonstrating that this integrin contributes to 4,5-bisphosphate to diacylglycerols and inositol 1,4,5-trisphosphate. VEGFR-2 signaling (93). Inositol 1,4,5-trisphosphate is likely to be responsible for the increase VE-cadherin, an endothelium-specific cell-cell adhesion protein, 2ϩ in intracellular Ca after VEGF stimulation (27), whereas diacyl- has also been implicated in molecular interactions with the VEGFRs. glycerol, in turn, activates certain PKC isoforms expressed in the Deficiency or truncation of VE-cadherin was lethal in mouse embryos 2ϩ target cells. VEGF selectively activated the Ca -sensitive PKC iso- due to increased endothelial cell apoptosis (98). In normal endothelial ␣ ␤ forms and 2 in bovine aortic endothelial cells, and the mitogenic cells, VEGFR-2 colocalized with VE-cadherin, and VEGF stimulation ␤ effect of VEGF in these cell was inhibited by a PKC- -selective resulted in the formation of a complex between VE-cadherin, ␤-cate- inhibitor (80). nin, PI3-K, and VEGFR-2 and subsequent Akt activation. On the Xia et al. (80) reported activation of PI3-K in response to VEGF other hand, disruption of VE-cadherin function prevented the endo- and demonstrated that PI3-K activity is not required for mitogenesis thelial cells from responding to survival signals induced by VEGF. or activation of PKC (74, 80). However, PI3-K was shown to activate Interestingly, VEGFR-3, but not VEGFR-1, also coimmunoprecipi- Akt, a serine kinase involved in antiapoptotic signaling, and to sub- tated with VE-cadherin. sequently deliver a survival signal in HUVECs treated with a Vascular Permeability May Be Transduced via an Unknown VEGFR-2-selective VEGF mutant (81). VEGFR-2 and the PI3-K/Akt VEGFR. Tumor vessels are leaky in general, and VEGF is one of the pathway may therefore play a role in VEGF-mediated survival of most potent inducers of vascular permeability (99). VEGF-C also has immature vessels (82). Akt has also been reported to directly activate vascular permeability activity, but the response is not transduced via endothelial nitric oxide synthase, suggesting that Akt may regulate the VEGFR-3 (89). On the other hand, PlGF, which only binds to increased production of nitric oxide in response to VEGF stimulation VEGFR-1 (36, 37), is incapable of inducing permeability. These data (83, 84). imply that either VEGFR-2 or some other as yet unidentified receptor STATs are latent cytoplasmic transcription factors. STAT activa- mediates the permeability response. When the third variable domain tion by the VEGFRs has been studied in transient transfection assays in the VEGF polypeptide was substituted with the analogous region of (85). All three receptors were shown to be strong activators of STAT3 PlGF, creating a chimeric molecule with significantly reduced binding and STAT5, whereas STAT1 was not activated by the VEGFRs. to VEGFR-2, the mutant still induced vascular permeability with an However, the role of this pathway in endothelial cell biology is unknown. activity similar to that of wild-type VEGF (100). Therefore, it may be VEGFR-3. Whereas both VEGFR-3 isoforms bind to and phos- that vascular permeability is not mediated by any of the known phorylate adapter protein Shc, phosphorylation is stronger in cells VEGFRs. Interestingly, the c-Src protein seems to be a necessary expressing the long isoform (54, 86, 87). The Shc PTB domain is mediator of the permeability response because adenoviral vectors required for ligand-induced Shc tyrosine phosphorylation by expressing VEGF do not induce vascular permeability in mice defi- VEGFR-3 (88), and mutations in Shc phosphorylation sites increased cient in c-Src (101). VEGFR-3 transforming activity in the soft agar assay, indicating that Importance of the Cellular Background in VEGF Signaling Shc has a negative role in VEGFR-3 signaling. Studies. Because most VEGFR signal transduction studies have been Both VEGFR-3 isoforms bind Grb2 via its SH2 domain in an carried out using endothelial cells that express more than one type of inducible manner (54, 87). Stimulation of VEGFR-3 also activates VEGFR, it has not been possible to attribute the results to a particular MAPK, at least in transfected cells (89). Similarly, both isoforms bind receptor. Attempts to study signal transduction by individual receptors to a glutathione S--SH2 (PLC-␥) fusion protein on stimu- using transfected cells have been compromised by the lack of a proper lation (54, 86). In contrast, no direct interaction has been observed cellular background, which may be an important factor in VEGFR with PI3-K (86). In a human erythroleukemia cell line and in KS cells, signaling, depending on the substrate under study. Use of receptor- VEGF-C induced tyrosine phosphorylation of the cytoskeletal protein specific ligands or mutants of the VEGF family may offer an alter- paxillin by related adhesion focal tyrosine kinase, a recently identified native approach to these questions (89, 102). Another variable in the member of the focal adhesion kinase family (90, 91). signal transduction studies is introduced by the heterogeneity of Although very little is known about the specific signal transduction endothelial cells and their tendency to lose differentiated properties in of VEGFR-3 in the lymphatic endothelium, mutations in VEGFR-3 culture. Keeping these aspects in mind, the signaling mechanisms have recently been linked with hereditary lymphedema, an autosomal discovered thus far are of interest and may offer possible targets for dominant disorder of the lymphatic system that leads to disabling therapeutic intervention (103). For example, global assessment of swelling of the extremities and, in rare cases, to lymphangiosarcomas endothelial cell gene expression by RNA microarray analysis may (see Fig. 1; Ref. 92). This indicates that disturbed VEGFR-3 signaling help to better define the activated state of the endothelial cells on may play key a role in the development of this disease. angiogenesis and drug treatment. However, for candidate drugs, ad- VEGFRs Associate with Cell Adhesion Receptors. According to ditional validation has to be provided by experiments in transgenic new data, endothelial cell proliferation and survival in response to and gene targeted mice. 205

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

The VEGFRs in Vascular Development limb buds, and expressed more widely in the mesenchyme (43, 120). Targeted disruption of the mouse NRP-1 gene caused severe abnor- The various VEGFRs are first expressed in the mouse embryonic malities in the peripheral nervous system (121). Interestingly, VEGF mesoderm between days 7.5–9.5 of development and distinct expres- has recently been reported to act also as a neurotrophic survival factor sion patterns are also found in 17 week human fetuses (104–106). and as a mitogenic agent for Schwann cells (122). Mice overexpress- Disruption of any of the three VEGFR genes leads to embryonic ing NRP-1 formed excess capillaries and abnormal blood vessels and lethality. In embryos with targeted null mutations of VEGFR-2, died in utero (120). It is possible that aberrant angiogenesis in NRP-1 differentiation of both endothelial and hematopoietic cells is blocked, transgenic embryos resulted from inappropriate VEGF activity. and no blood vessels are formed (107). However, at least in vitro, endothelial/hematopoietic precursor cells can be derived from VEGF and VEGFRs in Tumors VEGFR-2-deficient ES cells, demonstrating that VEGFR-2 is not required for the formation of the common hematopoietic/endothelial Expression of VEGF and its receptors correlates with the degree of progenitor cell, the so-called hemangioblast (108, 109). Rather, vascularization of many experimental and clinical tumors as detected VEGFR-2 signaling seems to be necessary for endothelial commit- by in situ hybridization and immunohistochemistry (123–132), and ment because endothelial development is terminated at an early stage both have been used as prognostic indicators of an increased meta- in VEGFR-2-negative ES cell cultures (109). Hematopoietic require- static risk (see Ref. 133). Although the detailed molecular mechanism ment for VEGFR-2, on the other hand, is conditional and depends on of the “angiogenic switch” by which quiescent endothelium becomes the cell culture conditions (108, 109). Although VEGFR-2 is not activated is unknown, VEGF seems to be the main inducer of tumor required for hemangioblast formation, it appears to be essential for the angiogenesis. Tumor hypoxia and oncogenes up-regulate VEGF lev- subsequent VEGF-directed hemangioblast migration to appropriate els in the neoplastic cells, and hypoxia, in combination with the environments in the developing embryo (108–110). In the absence of locally increased VEGF concentrations, up-regulates VEGFR-1 and VEGFR-2 signaling, such cells may rapidly disappear, explaining the VEGFR-2 on tumor endothelial cells (62, 127, 131). VEGF-negative lack of endothelial and hematoipoietic precursors in VEGFR-2 null ES cells or Ras-transformed fibroblasts grow poorly as tumors in embryos. syngeneic mice (134, 135). Interestingly, ES cells lacking HIF-1␣ In human postnatal hematopoietic tissues, VEGFR-2 apparently (deficient in hypoxic induction of VEGF) are tumorigenic. The tumors persists as a specific marker of hematopoietic stem cells, differenti- formed by HIF-1␣-deficient ES cells are poorly vascularized, but they ating them from the lineage-committed hematopoietic precursor cells are apparently more resistant to tumor cell apoptosis under hypoxic (111). Endothelial progenitors or angioblasts have also been isolated conditions (136). A recent report has suggested that in some cases, from peripheral blood and claimed to be incorporated into sites of tumor vessels can also be formed without endothelial cells (137). active angiogenesis (112, 113). Apparently, tissue ischemia or sys- Autocrine VEGF and VEGFR-1 expression has been shown to temic VEGF treatment can mobilize such bone marrow-derived occur in angiosarcomas (138). Of the nonendothelial tumors studied, mononuclear progenitor cells (114, 115). only a few melanomas and leukemia cell lines aberrantly expressed Mouse embryos homozygous for a targeted mutation of the VEGFRs, whereas several tumor cell lines expressed NRP-1 (22, VEGFR-1 locus develop an excess of endothelial cells in both em- 139–141).4 In tumors associated with the VHL disease, VEGF and bryonic and extraembryonic locations, but the endothelial cells fail to VEGFRs are constitutively up-regulated in the absence of hypoxia organize into normal vascular channels (116). The increase in endo- (see Ref. 142). Whereas the normal VHL protein functions as a thelial cell numbers is due to an alteration in cell fate determination component of a ubiquitin-protein complex that targets selected among mesenchymal cells, leading to increased hemangioblast com- proteins such as HIF-1␣ for ubiquitin-mediated proteolysis (143), the mitment (117). However, when only the tyrosine kinase domain of cells containing a defective VHL protein fail to degrade HIF-1␣, VEGFR-1 was deleted, leaving the ligand binding extracellular part leading to constitutive up-regulation of many hypoxia-regulated and the transmembrane domain intact, the gene-targeted mice devel- genes, including VEGF (144). oped normal vessels and survived (118). One possible explanation for VEGFR-3 is up-regulated in tumor angiogenesis in general, for these and other findings is that during embryogenesis, VEGFR-1 acts example, in breast carcinomas (145, 146). In addition, VEGFR-3 has as a VEGF sink, regulating the amount of free VEGF available for been shown to be increased in the endothelium of lymphatic vessels in vascular development. In the absence of VEGFR-1, there would be an metastatic lymph nodes and in lymphangiomas, vascular skin tumors, excess of free VEGF available to its major signal transducing recep- and KS spindle cells (105, 147, 148). tor, VEGFR-2. Therefore, coordinated expression of both VEGFR-1 VEGF and VEGFRs in the Development of AIDS-linked KS. and VEGFR-2 would be essential for controlled early vascular devel- KS, which is the major neoplastic manifestation of HIV-induced opment. AIDS, is an angiogenic tumor composed of endothelial and spindle Disruption of VEGFR-3 led to a defective remodeling of the cells (for a review, see Ref. 149). AIDS-linked KS is always accom- primary vascular plexus and cardiovascular failure after embryonic panied by infection with HHV-8. In KS lesions, HHV-8 is present in day 9.5, but differentiation of endothelial cells, formation of primitive endothelial cells and in spindle cells with endothelial characteristics. vascular networks, and vascular sprouting occurred normally (119). HHV-8 encodes for a G-protein-coupled receptor that has been shown This phenotype suggests that VEGFR-3 is required for maturation of to induce VEGF expression in transfected cells (150), and cell lines the primary vascular plexus into a hierarchy of large and small derived from AIDS-linked KS also express high levels of the vessels. Therefore, VEGFR-3 seems to have a general blood vascular VEGFRs (151). On the other hand, HIV-1 encodes a transcriptional function during early development and only later becomes restricted regulator protein, Tat, which also possesses angiogenic properties. Tat mostly to the lymphatic vascular system. Conditional gene targeting is has been shown to bind to and activate VEGFR-2 (152, 153), to needed to better define the lymphatic functions of VEGFR-3. stimulate KS spindle cell growth (154), and to induce neovascular- Other VEGF-binding proteins, such as NRP-1, also seem to be ization in vivo (155, 156) and in transgenic mice (157). VEGFR-3 is involved in the regulation of angiogenesis during development. In also increased in KS (148), and its ligand, VEGF-C, stimulates the normal mouse embryos, NRP-1 is expressed in a temporally restricted manner in endothelial cells of capillaries and blood vessels and in 4 Michael Klagsbrun, personal communication. 206

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs proliferation of KS cells in vitro (158). It thus seems probable that an autocrine VEGF/VEGFR activation loop plays a part in the develop- ment of AIDS-linked KS. Role of VEGF-induced Vessel Permeability in Tumor Angio- genesis. In contrast to the well-organized normal blood vessels, tu- mor vessels are characteristically poorly functioning, leaky endothe- lial channels with incomplete arteriovenous and perivascular differentiation (159). Tumor blood flow is temporally and spatially heterogenous, and the interstitial pressure in solid tumors is generally much higher than that in normal tissues. Lack of functional lymphatic vessels is another factor leading to interstitial hypertension (159). The microvascular hyperpermeability in tumors causes plasma proteins to leak into the extravascular space, leading to clotting of extravasated fibrinogen and introduction of a provisional plasma-derived matrix. Matrix formation precedes and accompanies the onset of endothelial cell migration and vessel sprouting (160). VEGF derived from hy- poxic areas of tumors is likely to mediate increased permeability. VEGF therefore contributes to tumor angiogenesis by both direct and indirect mechanisms: on one hand, VEGF stimulates endothelial cell proliferation and migration; on the other, it renders vessels hyperper- meable, leading to formation of a matrix that supports blood vessel growth. Interestingly, new studies have shown that both VEGF- Fig. 3. Schematic structures of the Tie and Tie-2 receptors. Both receptors contain one complete and one incomplete immunoglobulin domain (circles) plus three epidermal induced permeability and pathological angiogenesis are attenuated in growth factor homology domains (ovals) and fibronectin type III homology domains 5 PlGF-deficient mice. (yellow), followed by a transmembrane domain and a split tyrosine kinase catalytic domain (blue). The Tie-2 receptor binds four ligands; of these, Ang-3 and Ang-4 may be Angiogenesis Inhibitors and VEGFR Function isogenic in mice and humans, although they behave as an inhibitor and a stimulator of the receptor, respectively. The Tie-2 receptor has been implicated in tumor angiogenesis (166, Studies in animal models have illustrated the dependence of tumor 184, 185). Thus far, ligand(s) for Tie is unknown, on the other hand, Tie might also interact weakly with the Tie-2-ligand complex (question marks), similar to the mode of vascularization and progression on VEGF signaling. A heterodimeric action of some epidermal growth factors and receptors. VEGF variant in which the receptor binding sites at one pole of the ligand were mutated while the other pole was left intact was shown to act as an inhibitor of VEGF function.6 A four-fold excess of this apparently because VEGF-mediated blood vessel invasion is essential mutant VEGF, which bound to its receptors but failed to mediate for coupling the resorption of cartilage with bone formation (172). receptor dimerization and subsequent signaling, was sufficient for Interestingly, VEGF also supports osteoclast recruitment and bone almost complete inhibition of vascular permeability and tissue factor resorption, and this effect has been shown to be mediated via expression induced by wild-type VEGF. A dominant negative mutant VEGFR-1 expressed in osteoclasts (173). Given the role of VEGF in of VEGFR-2 has been expressed from a retroviral construct and was bone development, anti-VEGF therapy could be useful in treating found to prevent glioblastoma vascularization and growth in nude bone-associated pathologies, for example, bone metastases and osteo- mice (161). The study was later extended to other types of tumors, and sarcomas. in most cases, tumor growth was inhibited (162). Antibodies directed against either VEGFR-2 or the VEGFR-2/VEGF complex have also Cross-Talk between Different Receptor Families Required for been effective in inhibiting VEGF-mediated signaling and endothelial Angiogenesis cell proliferation in vitro (163–165). Because the dominant negative VEGFR-2 inhibits angiogenesis in vivo, compounds that block the Recent reports on the association of activated VEGFR-2 with other tyrosine kinase activity of VEGFR-2 should also prevent neovascu- cell surface receptors raise questions on the possible roles these larization. Indeed, specific VEGFR-2 tyrosine kinase inhibitors have coreceptors play during angiogenesis in vivo. Both NRP-1 and inte- ␣ ␤ been shown to abolish VEGF-induced mitogenesis of human endo- grin v 3 have been shown to increase the mitogenic effects of VEGF thelial cells in vitro and to have an antitumor effect in mice (103). (43, 93). Because NRP-1 binds only to certain VEGFs (39, 43–45), it Several such compounds (see Ref. 166) are currently under evaluation could function to specifically potentiate the effects of these family in clinical trials for the treatment of human cancers. members. In addition, NRP-1 may be able to induce biological re- Because soluble extracellular domains of the VEGFRs compete sponses in cells that express NRP-1 but no VEGFRs. VEGFR-2 ␣ ␤ with endothelial cell surface receptors for VEGF binding and inhibit association with VE-cadherin and v 3 may serve to create active VEGF-induced mitogenicity (52, 167, 168), recombinant VEGFRs signaling complexes by clustering proteins involved in VEGF-medi- ␣ ␤ retaining high ligand binding affinity also provide the potential for ated survival signaling (81, 98). v 3 is required for the full activation inhibition of tumor angiogenesis. Tumor growth in mice has been of VEGFR-2, suggesting that the complex might regulate tyrosine shown to be suppressed by the soluble VEGFR-1 and VEGFR-2 phosphatase activity or ensure correct juxtaposition of the receptor receptor “bodies” (168–170). Adult mice seem to tolerate such ther- and putative cytoskeletal substrates. apy well, whereas newborn mice that receive the inhibitor during their In addition to the VEGF/VEGFR system, Tie and Eph families of first two postnatal weeks fail to thrive and die (171). In young endothelial tyrosine kinases have important functions in the formation animals, systemic administration of soluble VEGFR-1 leads to inhi- and maintenance of the vascular system (174–180). Tie-1 and Tie-2 bition of growth plate differentiation and growth of long bones, are restricted to the vascular endothelium and certain hematopoietic progenitor cells. Four known ligands, the Angs, bind to Tie-2, 5 Peter Carmeliet and M. Graziella Persico, personal communication. whereas there are currently no ligands for Tie-1 (Fig. 3; Refs. 181– 6 Robert de Waal, personal communication. 185). Interestingly, recent findings demonstrate that overexpression of 207

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

Ang-1 leads to the formation of vessels resistant to vascular leakage 4. Thorpe, P. E., and Burrows, F. J. Antibody-directed targeting of the vasculature of induced by VEGF (186). Plasma leakage contributes not only to tumor solid tumors. Breast Cancer Res. Treat., 36: 237–251, 1995. 5. Arap, W., Pasqualini, R., and Ruoslahti, E. Chemotherapy targeted to tumor vascu- angiogenesis but also to a variety of other disease processes (160), and lature. Curr. Opin. Oncol., 10: 560–565, 1998. Ang-1 could perhaps be used therapeutically to reverse such adverse 6. Ruegg, C., Yilmaz, A., Bieler, G., Bamat, J., Chaubert, P., and Lejeune, F. J. ␣ ␤ Evidence for the involvement of endothelial cell integrin v 3 in the disruption of effects of VEGF. The Tie-2 pathway has been reported to be essential the tumor vasculature induced by TNF and IFN-␥. Nat. Med., 4: 408–414, 1998. for tumor growth and angiogenesis (168, 187, 188). In the study by 7. Bergers, G., Javaherian, K., Lo, K. M., Folkman, J., and Hanahan, D. Effects of Siemeister et al. (188), the VEGFR pathway was not sufficient to angiogenesis inhibitors on multistage carcinogenesis in mice. Science, 284: 808– 812, 1999. compensate for the lack of Tie-2 signaling, suggesting the existence of 8. Plate, K. H. Control of tumor growth via inhibition of tumor angiogenesis. Adv. Exp. two independent mechanisms, both of which are required for tumor Med. Biol., 451: 57–61, 1998. ␣ angiogenesis (188). Interestingly, the antagonistic Tie-2 ligand Ang-2 9. Eliceiri, B. P., and Cheresh, D. A. The role of v during angiogenesis: insights into potential mechanisms of action and clinical development. J. Clin. has been shown to be up-regulated in the endothelial cells of coopted Investig., 103: 1227–1230, 1999. host vessels of certain tumors, and its increased expression preceded 10. Carmeliet, P., and Collen, D. Role of vascular endothelial growth factor and vascular and coincided with apoptotic regression of these vessels (189). Hy- endothelial growth factor receptors in vascular development. Curr. Top. Microbiol. Immunol., 237: 133–158, 1999. poxia, , and VEGF have been shown to 11. Shibuya, M., Ito, N., and Claesson-Welsh, L. Structure and function of vascular increase Ang-2 mRNA in endothelial cells, suggesting important endothelial -1 and -2. Curr. Top. Microbiol. Immunol., 237: pathways of signal trans-regulation (see Fig. 3; Refs. 190, 191). The 59–83, 1999. 12. Neufeld, G., Cohen, T., Gengrinovitch, S., and Poltorak, Z. Vascular endothelial intense autocrine expression of Ang-2 by endothelial cells may rep- growth factor (VEGF) and its receptors. FASEB J., 13: 9–22, 1999. resent a host defense mechanism against the growing tumor because 13. Ferrara, N. Role of vascular endothelial growth factor in the regulation of angio- Ang-2 expression in the absence of VEGF has been implicated in genesis. Kidney Int., 56: 794–814, 1999. 14. Klagsbrun, M., and D’Amore, P. A. Vascular endothelial growth factor and its destabilizing the interactions of endothelial cells, , and the receptors. Cytokine Growth Factor Rev., 7: 259–270, 1996. surrounding extracellular matrix, which are necessary for endothelial 15. Ferrara, N. Vascular endothelial growth factor: molecular and biological aspects. cell survival (182). Curr. Top. Microbiol. Immunol., 237:1–30, 1999. 16. Persico, M. G., Vincenti, V., and DiPalma, T. Structure, expression and receptor- In contrast to the VEGFs and the Angs, the ligands of the large Eph binding properties of placenta growth factor (PlGF). In: L. Claesson-Welsh (ed.), RTK family, , do not function as soluble molecules but are Vascular Growth Factors and Angiogenesis, pp. 31–40. Berlin: Springer-Verlag, 1999. attached to membrane via either a transmembrane domain or a gly- 17. Ogawa, S., Oku, A., Sawano, A., Yamaguchi, S., Yazaki, Y., and Shibuya, M. A colipid anchor (180). In gene-targeted mice, -B2 was shown to novel type of vascular endothelial growth factor: VEGF-E (NZ-7 VEGF) preferen- specifically mark arterial endothelial cells, whereas its EphB receptors tially utilizes KDR/Flk-1 receptor and carries a potent mitotic activity without heparin-binding domain. J. Biol. Chem., 273: 31273–31282, 1998. specifically and reciprocally marked the venous endothelium (178, 18. Meyer, M., Clauss, M., Lepple-Wienhues, A., Waltenberger, J., Augustin, H. G., 179). Ephrins have also been linked to integrin function because a Ziche, M., Lanz, C., Bo¨ttner, M., Rziha, H-J., and Dehio, C. A novel vascular recent study showed that ephrin-B1 can promote the attachment of endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signaling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine endothelial cells to extracellular matrix components by activating kinases. EMBO J., 18: 363–374, 1999. ␣ ␤ integrins, including v 3 (192). Because inhibition of integrin func- 19. Shibuya, M. Role of VEGF-FLT receptor system in normal and tumor angiogenesis. tion has been shown to disrupt many different aspects of angiogenesis, Adv. Cancer Res., 67: 281–316, 1995. ␣ ␤ 20. Shibuya, M., Yamaguchi, S., Yamane, A., Ikeda, T., Tojo, A., Matsushime, H., and it is intriguing that v 3 has been shown to be associated with both Sato, M. Nucleotide sequence and expression of a novel human receptor type VEGFRs and the Ephs. tyrosine kinase gene (flt) closely related to the fms family. Oncogene, 5: 519–524, Of other receptor-ligand families, PDGF-B and PDGF receptor ␤ 1990. 21. Terman, B. I., Carrion, M. E., Kovacs, E., Rasmussen, B. A., Eddy, R. L., and have been implicated in the establishment of endothelial cell- Shows, T. B. Identification of a new endothelial cell growth factor receptor tyrosine interactions (193, 194). Transforming growth factor ␤ is also an kinase. Oncogene, 6: 1677–1683, 1991. important regulator of vascular structures (195). Endothelial cell- 22. Aprelikova, O., Pajusola, K., Partanen, J., Armstrong, E., Alitalo, R., Bailey, S. K., McMahon, J., Wasmuth, J., Huebner, K., and Alitalo, K. FLT4, a novel class III specific members of the TGF-␤ receptor family include endoglin and receptor tyrosine kinase in chromosome 5q33-qter. Cancer Res., 52: 746–748, 1992. ALK-1. Lack of endoglin led to poor vascular smooth muscle devel- 23. Galland, F., Karamysheva, A., Mattei, M-G., Rosnet, O., Marchetto, S., and Birnbaum, D. Chromosomal localization of FLT4, a novel receptor-type tyrosine opment, arrested endothelial remodeling, and lethality in mouse em- kinase gene. Genomics, 13: 475–478, 1992. bryos, demonstrating that endoglin is essential for developmental 24. Tischer, E., Mitchell, R., Hartman, T., Silva, M., Gospodarowicz, D., Fiddes, J. C., angiogenesis (196). On the other hand, elevated endoglin expression and Abraham, J. A. The human gene for vascular endothelial growth factor. Multiple protein forms are encoded through alternative exon splicing. J. Biol. Chem., 266: has been correlated with the proliferation of tumor endothelial cells 11947–11954, 1991. (197), and expression of ALK-1 coincides with sites of vasculogenesis 25. Houck, K. A., Ferrara, N., Winer, J., Cachianes, G., Li, B., and Leung, D. W. The and angiogenesis in early mouse development (198). Mutations in vascular endothelial growth factor family: identification of a fourth molecular species and characterization of alternative splicing of RNA. Mol. Endocrinol., 5: endoglin or ALK-1 are the cause of hereditary hemorrhagic telangi- 1806–1814, 1991. ectasia (199, 200). Endothelial cell-specific members of the trans- 26. Poltorak, Z., Cohen, T., Sivan, R., Kandelis, Y., Spira, G., Vlodavsky, I., Keshet, E., membrane tyrosyl phosphatase and Notch receptor families have also and Neufeld, G. VEGF145, a secreted vascular endothelial cell isoform, that binds to extracellular matrix. J. Biol. Chem., 272: 7151–7158, 1997. been described (201, 202). The functions of some or many of these 27. De Vries, C., Escobedo, J. A., Ueno, H., Houck, K., Ferrara, N., and Williams, L. T. may at least partially fail in the poorly organized tumor vessels, which The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. in general do not contain pericytes or well-formed basement mem- Science (Washington DC), 255: 989–991, 1992. 28. Terman, B. I., Dougher-Vermazen, M., Carrion, M. E., Dimitrov, D., Armellino, branes. A better understanding of the endothelial signal transduction D. C., Gospodarowicz, D., and Bo¨hlen, P. Identification of the KDR tyrosine kinase pathways should lead to the development of additional molecular as a receptor for vascular endothelial cell growth factor. Biochem. Biophys. Res. Commun., 187: 1579–1586, 1992. tools for the prevention of tumor angiogenesis. 29. Millauer, B., Wizigmann-Voos, S., Schnu¨rch, H., Martinez, R., Moller, N-P. H., Risau, W., and Ullrich, A. High affinity VEGF binding and developmental expres- References sion suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis. Cell, 72: 835–846, 1993. 1. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. 30. Kendall, R. L., Wang, G., and Thomas, K. A. Identification of a natural soluble form Med., 1: 27–31, 1995. of the vascular endothelial growth factor receptor, FLT-1, and its heterodimerization 2. Kerbel, R. S. A cancer therapy resistant to resistance. Nature (Lond.), 390: 335–336, with KDR. Biochem. Biophys. Res. Commun., 226: 324–328, 1996. 1997. 31. Wiesmann, C., Fuh, G., Christinger, H. W., Eigenbrot, C., Wells, J. A., and de Vos, 3. Denekamp, J. The tumour microcirculation as a target in cancer therapy: a clearer A. M. Crystal structure at 1.7 Å resolution of VEGF in complex with domain 2 of perspective. Eur. J. Clin. Invest., 29: 733–736, 1999. the Flt-1 receptor. Cell, 91: 695–704, 1997. 208

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

32. Muller, Y. A., Li, B., Christinger, H. W., Wells, J. A., Cunningham, B. C., and de 55. Tuder, R. M., Flook, B. E., and Voelkel, N. F. Increased gene expression for VEGF Vos, A. M. Vascular endothelial growth factor: crystal structure and functional and the VEGF receptors KDR/Flk and Flt in lungs exposed to acute or to chronic mapping of the kinase domain receptor . Proc. Natl. Acad. Sci. USA, 94: hypoxia. Modulation of gene expression by nitric oxide. J. Clin. Investig., 95: 7192–7197, 1997. 1798–1807, 1995. 33. Davis-Smyth, T., Chen, H., Park, J., Presta, L. G., and Ferrara, N. The second 56. Li, J., Brown, L. F., Hibberd, M. G., Grossman, J. D., Morgan, J. P., and Simons, immunoglobulin-like domain of the VEGF tyrosine kinase receptor Flt-1 determines M. VEGF, flk-1, and flt-1 expression in a rat myocardial infarction model of ligand binding and may initiate a signal transduction cascade. EMBO J., 15: angiogenesis. Am. J. Physiol., 270: H1803–H1811, 1996. 4919–4927, 1996. 57. Marti, H. H., and Risau, W. Systemic hypoxia changes the organ-specific distribu- 34. Barleon, B., Totzke, F., Herzog, C., Blanke, S., Kremmer, E., Siemeister, G., tion of vascular endothelial growth factor and its receptors. Proc. Natl. Acad. Sci. Marme, D., and Martiny-Baron, G. Mapping of the sites for ligand binding and USA, 95: 15809–15814, 1998. receptor dimerization at the extracellular domain of the vascular endothelial growth 58. Takagi, H., King, G. L., Ferrara, N., and Aiello, L. P. Hypoxia regulates vascular factor receptor FLT-1. J. Biol. Chem., 272: 10382–10388, 1997. endothelial growth factor receptor KDR/Flk gene expression through adenosine A2 35. Davis-Smyth, T., Presta, L. G., and Ferrara, N. Mapping the charged residues in the receptors in retinal capillary endothelial cells. Investig. Ophthalmol. Vis. Sci., 37: second immunoglobulin-like domain of the vascular endothelial growth factor/ 1311–1321, 1996. placenta growth factor receptor Flt-1 required for binding and structural stability. 59. Brogi, E., Schatteman, G., Wu, T., Kim, E. A., Varticovski, L., Keyt, B., and Isner, J. Biol. Chem., 273: 3216–3222, 1998. J. M. Hypoxia-induced paracrine regulation of vascular endothelial growth factor 36. Park, J. E., Chen, H. H., Winer, J., Houck, K. A., and Ferrara, N. Placental growth receptor expression. J. Clin. Investig., 97: 469–476, 1996. factor. Potentiation of vascular endothelial growth factor bioactivity in vitro and in 60. Gerber, H. P., Conorelli, F., Park, J., and Ferrara, N. Differential transcriptional vivo and high affinity binding to Flt-1 but not to Flk-1/KDR. J. Biol. Chem., 269: regulation of the two vascular endothelial growth factor receptor genes. Flt-1, but not 25646–25654, 1994. Flk-1/KDR, is up-regulated by hypoxia. J. Biol. Chem., 272: 23659–23667, 1997. 37. Sawano, A., Takahashi, T., Yamaguchi, S., Aonuma, M., and Shibuya, M. Flt-1 but 61. Shen, B. Q., Lee, D. Y., Gerber, H. P., Keyt, B. A., Ferrara, N., and Zioncheck, T. F. not KDR/Flk-1 tyrosine kinase is a receptor for placenta growth factor, which is Homologous upregulation of KDR/Flk-1 receptor expression by vascular endothelial related to vascular endothelial growth factor. Cell Growth Differ., 7: 213–221, 1996. growth factor in vitro. J. Biol. Chem., 273: 29979–29985, 1998. 38. Olofsson, B., Korpelainen, E., Mandriota, S., Pepper, M. S., Aase, K., Kumar, V., 62. Kremer, C., Breier, G., Risau, W., and Plate, K. H. Up-regulation of flk-1/vascular Gunji, Y., Jeltsch, M. M., Shibuya, M., Alitalo, K., and Eriksson, U. Vascular endothelial growth factor receptor 2 by its ligand in a cerebral slice culture system. endothelial growth factor (VEGF) B binds to VEGF receptor-1 and regulates Cancer Res., 57: 3852–3859, 1997. plasminogen activator activity in endothelial cells. Proc. Natl. Acad. Sci. USA, 95: 63. Ikeda, T., Wakiya, K., and Shibuya, M. Characterization of the promoter region for 11709–11714, 1998. the flt-1 tyrosine kinase gene, a receptor for vascular endothelial growth factor. 39. Wise, L. M., Veikkola, T., Mercer, A. A., Savory, L. J., Fleming, S. B., Caesar, C., Growth Factors, 13: 151–162, 1996. Vitali, A., Makinen, T., Alitalo, K., and Stacker, S. The vascular endothelial growth 64. Kappel, A., Volker, R., Damert, A., Flamme, I., Risau, W., and Breier, G. Identi- factor (VEGF)-like protein from orf virus NZ2 binds to VEGFR2 and neuropilin-1. fication of vascular endothelial growth factor (VEGF) receptor-2 (Flk-1) promoter/ Proc. Natl. Acad. Sci. USA, 96: 3071–3076, 1999. enhancer sequences sufficient for angioblast and endothelial cell-specific transcrip- 40. Joukov, V., Pajusola, K., Kaipainen, A., Chilov, D., Lahtinen, I., Kukk, E., Saksela, tion in transgenic mice. Blood, 93: 4284–4292, 1999. O., Kalkkinen, N., and Alitalo, K. A novel vascular endothelial growth factor, 65. Waltenberger, J., Claesson-Welsh, L., Siegbahn, A., Shibuya, M., and Heldin, C-H. VEGF-C, is a ligand for the Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine Different signal transduction properties of KDR and Flt1, two receptors for vascular kinases. EMBO J., 15: 290–298, 1996. endothelial growth factor. J. Biol. Chem., 269: 26988–26995, 1994. 66. Seetharam, L., Gotoh, N., Maru, Y., Neufeld, G., Yamaguchi, S., and Shibuya, M. 41. Lee, J., Gray, A., Yuan, J., Louth, S-M., Avraham, H., and Wood, W. Vascular A unique signal transduction from FLT tyrosine kinase, a receptor for vascular endothelial growth factor-related protein: a ligand and specific activator of the endothelial growth factor VEGF. Oncogene, 10: 135–147, 1995. tyrosine kinase receptor Flt4. Proc. Natl. Acad. Sci. USA, 93: 1988–1992, 1996. 67. Landgren, E., Schiller, P., Cao, Y., and Claesson-Welsh, L. Placenta growth factor 42. Achen, M. G., Jeltsch, M., Kukk, E., Makinen, T., Vitali, A., Wilks, A. F., Alitalo, stimulates MAP kinase and mitogenicity but not phospholipase C-␥ and migration K., and Stacker, S. A. Vascular endothelial growth factor D (VEGF-D) is a ligand of endothelial cells expressing Flt 1. Oncogene, 16: 359–367, 1998. for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc. 68. D’Angelo, G., Struman, I., Martial, J., and Weiner, R. I. Activation of mitogen- Natl. Acad. Sci. USA, 95: 548–553, 1998. activated protein kinases by vascular endothelial growth factor and basic fibroblast 43. Soker, S., Takashima, S., Miao, H. Q., Neufeld, G., and Klagsbrun, M. Neuropilin-1 growth factor in capillary endothelial cells is inhibited by the antiangiogenic factor is expressed by endothelial and tumor cells as an isoform-specific receptor for 16-kDa N-terminal fragment of prolactin. Proc. Natl. Acad. Sci. USA, 92: 6374– vascular endothelial growth factor. Cell, 92: 735–745, 1998. 6378, 1995. 44. Migdal, M., Huppertz, B., Tessler, S., Comforti, A., Shibuya, M., Reich, R., 69. Kroll, J., and Waltenberger, J. The vascular endothelial growth factor receptor KDR Baumann, H., and Neufeld, G. Neuropilin-1 is a placenta growth factor-2 receptor. activates multiple signal transduction pathways in porcine aortic endothelial cells. J. Biol. Chem., 273: 22272–22278, 1998. J. Biol. Chem., 272: 32521–32527, 1997. 45. Makinen, T., Olofsson, B., Karpanen, T., Hellman, U., Soker, S., Klagsbrun, M., 70. Abedi, H., and Zachary, I. Vascular endothelial growth factor stimulates tyrosine Eriksson, U., and Alitalo, K. Differential binding of vascular endothelial growth phosphorylation and recruitment to new focal adhesions of focal adhesion kinase and factor B splice and proteolytic isoforms to neuropilin-1. J. Biol. Chem., 274: paxillin in endothelial cells. J. Biol. Chem., 272: 15442–15451, 1997. 21217–21222, 1999. 71. Takahashi, T., and Shibuya, M. The 230 kDa mature form of KDR/Flk-1 46. DiSalvo, J., Bayne, M. L., Conn, G., Kwok, P. W., Trivedi, P. G., Soderman, D. D., (VEGFR-2) activates the PLC-␥ pathway and partially induces mitotic signals in Palisi, P. M., Sullivan, K. A., and Thomas, K. A. Purification and characterization NIH3T3 fibroblasts. Oncogene, 14: 2079–2089, 1997. of a naturally occurring vascular endothelial growth factor. Placenta growth factor 72. Rousseau, S., Houle, F., Landry, J., and Huot, J. P38 Map kinase activation by heterodimer. J. Biol. Chem., 270: 7717–7723, 1995. vascular endothelial growth factor mediates actin reorganization and cell migration 47. Cao, Y., Chen, H., Zhou, L., Chiang, M-K., Anand-Apte, B., Weatherbee, J. A., in human endothelial cells. Oncogene, 15: 2169–2177, 1997. Wang, Y., Fang, F., Flanagan, J. G., and Tsang, M. L-S. Heterodimers of placenta 73. Mukhopadhyay, D., Nagy, J., Manseau, E., and Dvorak, H. Vascular permeability growth factor/vascular endothelial growth factor. J. Biol. Chem., 271: 3154–3162, factor/vascular endothelial growth factor-mediated signaling in mouse mesentery 1996. vascular endothelium. Cancer Res., 58: 1278–1284, 1998. 48. Olofsson, B., Pajusola, K., Kaipainen, A., Von Euler, G., Joukov, V., Saksela, O., 74. Takahashi, T., Ueno, H., and Shibuya, M. VEGF activates protein kinase C- Orpana, A., Pettersson, R. F., Alitalo, K., and Eriksson, U. Vascular endothelial dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis growth factor B, a novel growth factor for endothelial cells. Proc. Natl. Acad. Sci. in primary endothelial cells. Oncogene, 18: 2221–2230, 1999. USA, 93: 2576–2581, 1996. 75. Parenti, A., Morbidelli, L., Cui, X. L., Douglas, J. G., Hood, J. D., Granger, H. J., 49. Gitay-Goren, H., Soker, S., Vlodavsky, I., and Neufeld, G. The bidning of vascular Ledda, F., and Ziche, M. Nitric oxide is an upstream signal of vascular endothelial endothelial growth factor to its receptors is dependent on cell surface-associated growth factor-induced extracellular signal-regulated kinase 1/2 activation in post- heparin-like molecules. J. Biol. Chem., 267: 6093–6098, 1992. capillary endothelium. J. Biol. Chem., 273: 4220–4226, 1998. 50. Olofsson, B., Pajusola, K., von Euler, G., Chilov, D., Alitalo, K., and Eriksson, U. 76. Igarashi, K., Shigeta, K., Isohara, T., Yamano, T., and Uno, I. Sck interacts with Genomic organization of the mouse and human genes for vascular endothelial KDR and Flt-1 via its SH2 domain. Biochem. Biophys. Res. Commun., 251: 77–82, growth factor B (VEGF-B) and characterization of a second splice isoform. J. Biol. 1998. Chem., 271: 19310–19317, 1996. 77. Guo, D., Jia, Q., Song, H. Y., Warren, R. S., and Donner, D. B. Vascular endothelial 51. Fairbrother, W. J., Champe, M. A., Chistinger, H. W., Keyt, B. A., and Starovasnik, cell growth factor promotes tyrosine phosphorylation of mediators of signal trans- M. A. Solution structure of the heparin-binding domain of vascular endothelial duction that contain SH2 domains. Association with endothelial cell proliferation. growth factor. Structure (Lond.), 6: 637–648, 1998. J. Biol. Chem., 270: 6729–6733, 1995. 52. Kendall, R. L., and Thomas, K. A. Inhibition of vascular endothelial cell growth 78. Struman, I., Bentzien, F., Lee, H., Mainfroid, V., D’Angelo, G., Goffin, V., Weiner, factor activity by an endogenously encoded soluble receptor. Proc. Natl. Acad. Sci. R. I., and Martial, J. A. Opposing actions of intact and N-terminal fragments of the USA, 90: 10705–10709, 1993. human prolactin/growth hormone family members on angiogenesis: an efficient 53. Pajusola, K., Aprelikova, O., Armstrong, E., Morris, S., and Alitalo, K. Two human mechanism for the regulation of angiogenesis. Proc. Natl. Acad. Sci. USA, 96: FLT4 receptor tyrosine kinase isoforms with distinct carboxyterminal tails are 1246–1251, 1999. produced by alternative processing of primary transcripts. Oncogene, 8: 2931–2937, 79. Igarashi, K., Isohara, T., Kato, T., Shigeta, K., Yamano, T., and Uno, I. Tyrosine 1993. 1213 of Flt-1 is a major binding site of Nck and SHP-2. Biochem. Biophys. Res. 54. Pajusola, K., Aprelikova, O., Pelicci, G., Weich, H., Claesson-Welsh, L., and Commun., 246: 95–99, 1998. Alitalo, K. Signalling properties of FLT4, a proteolytically processed receptor 80. Xia, P., Aiello, L. P., Ishii, H., Jiang, Z. Y., Park, D. J., Robinson, G. S., Takagi, H., tyrosine kinase related to two VEGF receptors. Oncogene, 9: 3545–3555, 1994. Newsome, W. P., Jirousek, M. R., and King, G. L. Characterization of vascular 209

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

endothelial growth factor’s effect on the activation of protein kinase C, its isoforms, 105. Kaipainen, A., Korhonen, J., Pajusola, K., Aprelikova, O., Persico, M. G., Terman, and endothelial cell growth. J. Clin. Investig., 98: 2018–2026, 1996. B. I., and Alitalo, K. The related FLT4, FLT1 and KDR receptor tyrosine kinases 81. Gerber, H-P., McMurtrey, A., Kowalski, J., Minhong, Y., Keyt, B. A., Dixit, V., and show distinct expression patterns in human fetal endothelial cells. J. Exp. Med., 178: Ferrara, N. Vascular endothelial growth factor regulates endothelial cell survival 2077–2088, 1993. through the phosphatidylinositol 3Ј-kinase/Akt signal transduction pathway. J. Biol. 106. Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V. M., Fang, G-H., Chem., 273: 30336–30343, 1998. Dumont, D., Breitman, M., and Alitalo, K. Expression of the fms-like tyrosine 82. Alon, T., Hemo, I., Itin, A., Pe’er, J., Stone, J., and Keshet, E. Vascular endothelial kinase FLT4 gene becomes restricted to endothelium of lymphatic vessels during growth factor acts as a survival factor for newly formed retinal vessels and has development. Proc. Natl. Acad. Sci. USA, 92: 3566–3570, 1995. implications for retinopathy of prematurity. Nat. Med., 1: 1024–1028, 1995. 107. Shalaby, F., Rossant, J., Yamaguchi, T. P., Gertsenstein, M., Wu, X. F., Breitman, 83. Fulton, D., Gratton, J. P., McCabe, T. J., Fontana, J., Fujio, Y., Walsh, K., Franke, M. L., and Schuh, A. C. Failure of blood island formation and vasculogenesis in T. F., Papapetropoulos, A., and Sessa, W. C. Regulation of endothelium-derived Flk-1-deficient mice. Nature (Lond.), 376: 62–66, 1995. nitric oxide production by the protein kinase Akt. Nature (Lond.), 399: 597–601, 108. Schuh, A. C., Faloon, P., Hu, Q-L., Bhimani, M., and Kyunghee, C. In vitro 1999. hematopoietic and endothelial potential of flk-1Ϫ/Ϫ embryonic stem cells and 84. Dimmeler, S., Fleming, I., Fisslthaler, B., Hermann, C., Busse, R., and Zeiher, A. M. embryos. Proc. Natl. Acad. Sci. USA, 96: 2159–2164, 1999. Activation of nitric oxide sythase in endothelial cells by Akt-dependent phospho- 109. Hidaka, M., Stanford, W. L., and Bernstein, A. Conditional requirement for the rylation. Nature (Lond.), 399: 601–605, 1999. Flk-1 receptor in the in vitro generation of early hematopoietic cells. Proc. Natl. 85. Korpelainen, E. I., Ka¨rkka¨inen, M., Gunji, Y., Vikkula, M., and Alitalo, K. Endo- Acad. Sci. USA, 96: 7370–7375, 1999. thelial receptor tyrosine kinases activate the STAT signaling pathway: mutant Tie-2 110. Shalaby, F., Ho, J., Stanford, W. L., Fischer, K. D., Schuh, A. C., Schwartz, L., causing venous malformations signals a distinct STAT activation response. Onco- Bernstein, A., and Rossant, J. A requirement for Flk1 in primitive and definitive gene, 18: 1–8, 1999. hematopoiesis and vasculogenesis. Cell, 89: 981–990, 1997. 86. Borg, J-P., deLapeyrie`re, O., Noguchi, T., Rottapel, R., Dubreuil, P., and Birnbaum, 111. Ziegler, B. L., Valtieri, M., Porada, G. A., Maria, R. D., Muller, R., Masella, B., D. Biochemical characterization of two isoforms of FLT4, a VEGF receptor-related Gabbianelli, M., Casella, I., Pelosi, E., Bock, T., Zanjani, E. D., and Peschle, C. tyrosine kinase. Oncogene, 10: 973–984, 1995. KDR receptor: a key marker defining hematopoietic stem cells. Science (Washing- 87. Fournier, E., Dubreuil, P., Birnbaum, D., and Borg, J-P. Mutation at tyrosine residue ton DC), 285: 1553–1558, 1999. 1337 abrogates ligand-dependent transforming capacity of the FLT4 receptor. On- 112. Asahara, T., Murohara, T., Sullivan, A., Silver, M., van der Zee, R., Li, T., cogene, 11: 921–931, 1995. Witzenbichler, B., Schatteman, G., and Isner, J. M. Isolation of putative progenitor 88. Fournier, E., Blaikie, P., Rosnet, O., Margolis, B., Birnbaum, D., and Borg, J-P. Role endothelial cells for angiogenesis. Science (Washington DC), 275: 964–967, 1997. of tyrosine residues and protein interaction domains of SHC adaptor protein in 113. Shi, Q., Rafii, S., Wu, M. H-D., Wijelath, E. S., Yu, C., Ishida, A., Fujita, Y., VEGF receptor 3 signaling. Oncogene, 18: 507–514, 1999. Kothari, S., Mohle, R., Sauvage, L. R., Moore, M. A. S., Storb, R. F., and Hammond, 89. Joukov, V., Kumar, V., Sorsa, T., Arighi, E., Weich, H., Saksela, O., and Alitalo, K. W. P. Evidence for circulationg bone marrow-derived endothelial cells. Blood, 92: A recombinant mutant vascular endothelial growth factor-C that has lost vascular 362–367, 1998. endothelial growth factor receptor-2 binding, activation and vascular permeability 114. Asahara, T., Takahashi, T., Masuda, H., Kalka, C., Chen, D., Iwaguro, H., Inai, Y., activities. J. Biol. Chem., 273: 6599–6602, 1998. Silver, M., and Isner, J. M. VEGF contributes to postnatal neovascularization by 90. Liu, Z. Y., Ganju, R. K., Wang, J. F., Ona, M. A., Hatch, W. C., Zheng, T., mobilizing bone marrow-derived endothelial progenitor cells. EMBO J., 18: 3964– Avraham, S., Gill, P., and Groopman, J. E. Cytokine signaling through the novel 3972, 1999. tyrosine kinase RAFTK in Kaposi’s sarcoma cells. J. Clin. Investig., 99: 1798–1804, 115. Takahashi, T., Kalka, C., Masuda, H., Chen, D., Silver, M., Kearney, M., Magner, 1997. M., Isner, J. M., and Asahara, T. Ischemia- and cytokine-induced mobilization of 91. Wang, J-F., Ganju, R., Liu, Z-Y., Avraham, H., Avraham, S., and Groopman, J. bone marrow-derived endothelial progenitor cells for neovascularization. Nat. Med., Signal transduction in human hematopoietic cells by vascular endothelial growth 5: 434–438, 1999. factor related protein, a novel ligand for the FLT4 receptor. Blood, 90: 3507–3515, 116. Fong, G. H., Rossant, J., Gertsenstein, M., and Breitman, M. L. Role of the Flt-1 1997. receptor tyrosine kinase in regulating the assembly of vascular endothelium. Nature 92. Ferrell, R. E., Levinson, K. L., Esman, J. H., Kimak, M. A., Lawrence, E. C., (Lond.), 376: 66–70, 1995. Barmada, M. M., and Finegold, D. N. Hereditary lymphedema: evidence for linkage 117. Fong, G-H., Zhang, L., Bryce, D-M., and Peng, J. Increased hemangioblast com- and genetic heterogeneity. Hum. Mol. Genet., 7: 2073–2078, 1998. mitment, not vascular disorganization, is the primary defect in flt-1 knock-out mice. 93. Soldi, R., Mitola, S., Strasly, M., Defilippi, P., Tarone, G., and Bussolino, F. Role Development (Camb.), 126: 3015–3025, 1999. of ␣ ␤ in the activation of vascular endothelial growth factor receptor-2. EMBO J., v 3 118. Hiratsuka, S., Minowa, O., Kuno, J., Noda, T., and Shibuya, M. Flt-1 lacking the 18: 882–892, 1999. 94. Brooks, P. C., Montgomery, A. M. P., Rosenfeld, M., Reisfeld, R. A., Hu, T., Klier, tyrosine kinase domain is sufficient for normal development and angiogenesis in ␣ ␤ mice. Proc. Natl. Acad. Sci. USA, 4: 9349–9354, 1998. G., and Cheresh, D. A. Integrin v 3 antagonists promote tumor regression by inducing apoptosis of angiogenic blood vessels. Cell, 79: 1157–1164, 1994. 119. Dumont, D., Jussila, L., Taipale, J., Mustonen, T., Pajusola, K., Breitman, M., and 95. Stro¨mbland, S., Becker, J. C., Yebra, M., Brooks, P. C., and Cheresh, D. A. Alitalo, K. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Suppression of p53 activity and p21WAF/CIP1 expression by vascular cell integrin Science (Washington DC), 282: 946–949, 1998. ␣ ␤ 120. Kitsukawa, T., Shimono, A., Kawakami, A., Kondoh, H., and Hajime, F. Overex- v 3 during angiogenesis. J. Clin. Investig., 98: 426–433, 1996. 96. Scatena, M., Almeida, M., Chaisson, M. L., Fausto, N., Nicosia, R. F., and Giachelli, pression of a membrane protein, neuropilin, in chimeric mice causes abnormalities ␬ ␣ ␤ in the cardiovascular system, nervous system and limbs. Development (Camb.), 121: C. M. NF- B mediates v 3 integrin-induced endothelial cell survival. J. Cell Biol., 141: 1083–1093, 1998. 4309–4318, 1995. 97. Ruoslahti, E. Fibronectin and its integrin receptors in cancer. Adv. Cancer Res., 76: 121. Kitsukawa, T., Shimizu, M., Sanbo, M., Hirata, T., Taniguchi, M., Bekku, Y., Yagi, 1–20, 1999. T., and Fujisawa, H. Neuropilin-semaphorin III/D-mediated chemorepulsive signals 98. Carmeliet, P., Lampugnani, M-G., Moons, L., Breviario, F., Compernolle, V., Bono, play a crucial role in peripheral nerve projection in mice. Neuron, 19: 995–1005, F., Balconi, G., Spagnuolo, R., Oosthuyse, B., Dewerchin, M., Zanetti, A., Angellilo, 1997. A., Mattot, V., Nuyens, D., Lutgens, E., Clotman, F., de Ruiter, M. C., 122. Sondell, M., Lundborg, G., and Kanje, M. Vascular endothelial growth factor has Gittenberger-de Groot, A., Poelmann, R., Lupu, F., Herbert, J-M., Collen, D., and neurotrophic activity and stimulates axonal outgrowth, enhancing cell survival and Dejana, E. Targeted deficiency or cytosolic truncation of the VE-cadherin gene Schwann cell proliferation in the peripheral nervous system. J. Neurosci., 19: in mice impairs VEGF-mediated endothelial survival and angiogenesis. Cell, 98: 5731–5740, 1999. 147–157, 1999. 123. Brown, L. F., Berse, B., Jackman, R. W., Tognazzi, K., Manseau, E. J., Dvorak, 99. Dvorak, H. F., Brown, L. F., Detmar, M., and Dvorak, A. M. Vascular permeability H. F., and Senger, D. R. Increased expression of vascular permeability factor factor/vascular endothelial growth factor, microvascular hyperpermeability, and (vascular endothelial growth factor) and its receptors in kidney and bladder carci- angiogenesis. Am. J. Pathol., 146: 1029–1039, 1995. nomas. Am. J. Pathol., 143: 1255–1262, 1993. 100. Stacker, S. A., Vitali, A., Caesar, C., Domagala, T., Groenen, L. C., Nice, E., Achen, 124. Brown, L. F., Berse, B., Jackman, R. W., Tognazzi, K., Manseau, E. J., Senger, M. G., and Wilks, A. F. A mutant form of vascular endothelial growth factor D. R., and Dvorak, H. F. Expression of vascular permeability factor (vascular (VEGF) that lacks VEGF receptor-2 activation retains the ability to induce vascular endothelial growth factor) and its receptors in adenocarcinomas of the gastrointes- permeability. J. Biol. Chem., 274: 34884–34892, 1999. tinal tract. Cancer Res., 53: 4727–4735, 1993. 101. Eliceiri, B. P., Paul, R., Schwartzberg, P. L., Hood, J. D., Leng, J., and Cheresh, D. 125. Brown, L. F., Berse, B., Jackman, R. W., Tognazzi, K., Guidi, A. J., Dvorak, H. F., A. Selective requirement for Src kinases during VEGF-induced angiogenesis and Senger, D. R., Conolly, J. L., and Schnitt, S. J. Expression of vascular permeability vascular permeability. Mol. Cell, 4: 915–924, 1999. factor (vascular endothelial growth factor) and its receptors in breast cancer. Hum. 102. Keyt, B. A., Nguyen, H. V., Berleau, L. T., Duarte, C. M., Park, J., Chen, H., and Pathol., 26: 86–91, 1995. Ferrara, N. Identification of vascular endothelial growth factor determinants for 126. Hatva, E., Kaipainen, A., Mentula, P., Ja¨a¨skela¨inen, J., Paetau, A., Haltia, M., and binding KDR and FLT-1 receptors. J. Biol. Chem., 271: 5638–5646, 1996. Alitalo, K. Expression of endothelial cell-specific receptor tyrosine kinases and 103. Fong, T. A. T., Shawver, L. K., Sun, L., Tang, C., App, H., Powell, T. J., Kim, Y. H., growth factors in human brain tumors. Am. J. Pathol., 146: 368–378, 1995. Schreck, R., Wang, X., Risau, W., Ullrich, A., Hirth, K. P., and McMahon, G. 127. Plate, K. H., Breier, G., Weich, H. A., and Risau, W. Vascular endothelial growth SU5416 is a potent and selective inhibitor of the vascular endothelial growth factor factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature receptor (Flk-1/KDR) that inhibits tyrosine kinase catalysis, tumor vascularization, (Lond.), 359: 845–848, 1992. and growth of multiple tumor types. Cancer Res., 59: 99–106, 1999. 128. Takahashi, Y., Kitadai, Y., Bucana, C. D., Cleary, K. R., and Ellis, L. M. Expression 104. Dumont, D. J., Fong, G-H., Puri, M., Gradwohl, G., Alitalo, K., and Breitman, M. L. of vascular endothelial growth factor and its receptor, KDR, correlates with vascu- Vascularization of the mouse embryo: a study of flk-1, tek, tie and VEGF expression larity, metastasis, and proliferation of human colon cancer. Cancer Res., 55: 3964– during development. Mech. Dev., 203: 80–92, 1995. 3968, 1995. 210

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

129. Warren, R. S., Yuan, H., Matli, M. R., Gillet, N. A., and Ferrara, N. Regulation by activated protein kinase, and components of focal adhesion in Kaposi’s sarcoma vascular endothelial growth factor of human colon cancer tumorigenesis in a mouse cells. J. Virol., 72: 6131–6137, 1998. model of experimental liver metastasis. J. Clin. Investig., 95: 1789–1797, 1995. 154. Ensoli, B., Barillari, G., Salahuddin, S. Z., Gallo, R. C., and Wong-Staal, F. Tat 130. Hatva, E., Bo¨hling, T., Ja¨a¨skela¨inen, J., Persico, M. G., Haltia, M., and Alitalo, K. protein of HIV-1 stimulates growth of cells derived from Kaposi’s sarcoma lesions Vascular growth factors and receptors in capillary hemangioblastomas and heman- of AIDS patients. Nature (Lond.), 345: 84–86, 1990. giopericytomas. Am. J. Pathol., 148: 763–772, 1996. 155. Albini, A., Benelli, R., Presta, M., Rusnati, M., Ziche, M., Rubartelli, A., 131. Shweiki, D., Itin, A., Soffer, D., and Keshet, E. Vascular endothelial growth factor Paglialunga, G., Bussolino, F., and Noonan, D. HIV-tat protein is a heparin- induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature (Lond.), binding angiogenic growth factor. Oncogene, 12: 289–297, 1996. 359: 843–845, 1992. 156. Ensoli, B., Gendelman, R., Markham, P., Fiorelli, V., Colombini, S., Raffeld, M., 132. Jain, R. K., Safabakhsh, N., Sckell, A., Chen, Y., Jiang, P., Benjamin, L., Yuan, F., Cafaro, A., Chang, H. K., Brady, J. N., and Gallo, R. C. Synergy between basic and Keshet, E. Endothelial cell death, angiogenesis, and microvascular function after fibroblast growth factor and HIV-1 Tat protein in induction of Kaposi’s sarcoma. castration in an androgen-dependent tumor: a role of vascular endothelial growth Nature (Lond.), 371: 674–680, 1994. factor. Proc. Natl. Acad. Sci. USA, 95: 10820–10825, 1998. 157. Vogel, J., Hinrichs, S. H., Reynolds, R. K., Luciw, P. A., and Jay, G. The HIV tat 133. Weidner, N. Intratumor microvessel density as a prognostic factor in cancer. Am. J. gene induces dermal lesions resembling Kaposi’s sarcoma in transgenic mice. Pathol., 147: 9–19, 1995. Nature (Lond.), 335: 606–611, 1988. 134. Ferrara, N., Carver-Moore, K., Chen, H., Dowd, M., Lu, L., O’Shea, K. S., Powell- 158. Marchio`, S., Primo, L., Pagano, M., Palestro, G., Albini, A., Veikkola, T., Cascone, Braxton, L., Hilan, K. J., and Moore, M. W. Heterozygous embryonic lethality I., Alitalo, K., and Bussolino, F. Vascular endothelial growth factor-C stimulates the induced by targeted inactivation of the VEGF gene. Nature (Lond.), 380: 439–442, migration and proliferation of Kaposi’s sarcoma cells. J. Biol. Chem., 274: 27617– 1996. 27622, 1999. 135. Grunstein, J., Roberts, W. G., Mathieu-Costello, O., Hanahan, D., and Johnson, R. S. 159. Jain, R. J. The next frontier of molecular medicine: delivery of therapeutics. Nat. Tumor-derived expression of vascular endothelial growth factor is a critical factor in Med., 4: 655–657, 1998. tumor expansion and vascular function. Cancer Res., 59: 1592–1598, 1999. 160. Dvorak, H. F., Nagy, J. A., Feng, D., Brown, L. F., and Dvorak, A. M. Vascular 136. Carmeliet, P., Dor, Y., Herbert, J-M., Fukumura, D., Brusselmans, K., Dewerchin, permeability factor/vascular endothelial growth factor and the significance of mi- M., Neeman, M., Bono, F., Abramovitch, R., Maxwell, P., Koch, C. J., Ratcliffe, P., crovascular permeability in angiogenesis. Curr. Top. Microbiol., 237: 97–132, 1999. Moons, L., Jain, R. K., Collen, D., and Keshet, E. Role of HIF-1␣ in hypoxia- mediated apoptosis, cell proliferation and tumour angiogenesis. Nature (Lond.), 394: 161. Millauer, B., Shawver, L., Plate, K., Risau, W., and Ullrich, A. Glioblastoma growth 485–490, 1998. inhibited in vivo by a dominant-negative Flk-1 mutant. Nature (Lond.), 367: 576– 137. Maniotis, A. J., Folberg, R., Hess, A., Seftor, E., Gardner, L. M. G., Pe’er, J., Trent, 579, 1994. J. M., Meltzer, P.S., and Hendrix, M. J. C. Vascular channel formation by human 162. Millauer, B., Longhi, M. P., Plate, K. H., Shawver, L. K., Risau, W., Ullrich, A., and melanoma cells in vivo and in vitro: vasculogenic mimicry. Am. J. Pathol. 155: Strawn, L. M. Dominant-negative inhibition of Flk-1 suppresses the growth of many 739–752, 1999. tumor types in vivo. Cancer Res., 56: 1615–1620, 1996. 138. Hashimoto, M., Ohsawa, M., Ohnishi, A., Naka, N., Hirota, S., Kitamura, Y., and 163. Brekken, R. A., Huang, X., King, S. W., and Thorpe, P. E. Vascular endothelial Aozasa, K. Expression of vascular endothelial growth factor and its receptor mRNA growth factor as a marker of tumor endothelium. Cancer Res., 58: 1952–1959, in angiosarcoma. Lab. Invest., 73: 859–863, 1995. 1998. 139. Cohen, T., Gitay-Goren, H., Sharon, R., Shibuya, M., Halaban, R., Levi, B-Z., and 164. Zhu, Z., Rockwell, P., Lu, D., Kotanides, H., Pytowski, B., Hicklin, D. J., Bohlen, Neufeld, G. VEGF121, a vascular endothelial growth factor (VEGF) isoform lacking P., and Witte, L. Inhibition of vascular endothelial growth factor-induced receptor heparin binding ability, requires cell-surface heparan sulfates for efficient binding to activation with anti-kinase insert domain-containing receptor single-chain antibodies the VEGF receptors of human melanoma cells. J. Biol. Chem., 270: 11322–11326, from a phage display library. Cancer Res., 58: 3209–3214, 1998. 1995. 165. Witte, L., Hicklin, D. J., Zhu, Z., Pytowski, B., Kotanides, H., Rockwell, P., and 140. Liu, B., Earl, H. M., Baban, D., Shoaibi, M., Fabra, A., Kerr, D. J., and Seymor, Bohlen, P. Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an L. W. Melanoma cell lines express VEGF receptor KDR and respond to exogenously anti-angiogenic therapeutic strategy. Cancer Metastasis Rev., 17: 155–161, 1998. added VEGF. Biochem. Biophys. Res. Commun., 217: 721–727, 1995. 166. Mohammadi, M., Froum, S., Hamby, J. M., Schroeder, M. C., Panek, R. L., Lu, 141. Fiedler, W., Graeven, U., Su¨leyman, E., Verago, S., Kilic, N., Stockschla¨der, M., G. H., Eliseenkova, A. V., Green, D., Schlessinger, J., and Hubbard, S. R. Crystal and Hossfeld, D. K. Vascular endothelial growth factor, a possible paracrine growth structure of an angiogenesis inhibitor bound to the VEGF receptor tyrosine kinase factor in human acute myeloid leukemia. Blood, 89: 1870–1875, 1997. domain. EMBO J., 17: 5896–5904, 1998. 142. Wizigmann-Voos, S., and Plate, K. H. Pathology, genetics and cell biology of 167. Roeckl, W., Hecht, D., Sztajer, H., Waltenberger, J., Yayon, A., and Weich, H. A. hemangioblastomas. Histol. Histopathol., 11: 1049–1061, 1996. Differential binding characteristics and cellular inhibition by soluble VEGF recep- 143. Lisztwan, J., Imbert, G., Wirbelauer, C., Gstaiger, M., and Krek, W. The von tors 1 and 2. Exp. Cell Res., 241: 161–170, 1998. Hippel-Lindau tumor suppressor protein is a component of an E3 ubiquitin-protein 168. Lin, P., Sankar, S., Shan, S., Dewhirst, M. W., Polverini, P. J., Quinn, T. Q., and ligase activity. Genes Dev., 13: 1822–1833, 1999. Peters, K. G. Inhibition of tumor growth by targeting tumor endothelium using a 144. Maxwell, P. H., Wiesener, M. S., Chang, G-W., Clifford, S. C., Vaux, E. C., soluble vascular endothelial growth factor receptor. Cell Growth Differ., 9: 49–58, Cockman, M. E., Wykoff, C. C., Pugh, C. W., Maher, E. R., and Ratcliffe, P. J. The 1998. tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen- 169. Goldman, C. K., Kendall, R. L., Cabrera, G., Soroceanu, L., Heike, Y., Gillespie, Y., dependent proteolysis. Nature (Lond.), 399: 271–275, 1999. Siegal, G. P., Mao, X., Bett, A. J., Huckle, W. R., Thomas, K. A., and Curiel, D. T. 145. Valtola, R., Salven, P., Heikkila¨, P., Taipale, J., Joensuu, H., Rehn, M., Pihlajaniemi, Paracrine expression of a native soluble vascular endothelial growth factor receptor T., Weich, H., de Waal, R., and Alitalo, K. VEGFR-3 and its ligand VEGF-C are inhibits tumor growth, metastasis, and mortality rate. Proc. Natl. Acad. Sci. USA, associated with angiogenesis in breast cancer. Am. J. Pathol., 154: 1381–1390, 95: 8795–8800, 1998. 1999. 170. Kong, H. L., Hecht, D., Song, W., Kovesdi, I., Hackett, N. R., Yayon, A., and 146. Partanen, T., Alitalo, K., and Miettinen, M. Lack of lymphatic vascular specificity Crystal, R. G. Regional suppression of tumor growth by in vivo transfer of a cDNA of VEGFR-3: a novel marker for human vascular tumors. Cancer (Phila.), 86: encoding a secreted form of the extracellular domain of the flt-1 vascular endothelial 2406–2412, 1999. growth factor receptor. Hum. Gene Ther., 9: 823–833, 1998. 147. Lymboussaki, A., Partanen, T. A., Olofsson, B., Thomas-Crusells, J., Fletcher, 171. Gerber, H-P., Hillan, K. J., Ryan, A. M., Kowalski, J., Keller, G-A., Rangell, L., C. D. M., de Waal, R. M. W., Kaipainen, A., and Alitalo, K. Expression of the Wright, B. D., Radtke, F., Aguet, M., and Ferrara, N. VEGF is required for growth vascular endothelial growth factor C receptor VEGFR-3 in cutaneous lymphatic and survival in neonatal mice. Development (Camb.), 126: 1149–1159, 1999. vascular endothelium and in vascular skin lesions. Am. J. Pathol., 153: 395–403, 172. Gerber, H-P., Vu, T. H., Ryan, A. M., Kowalski, J., Werb, Z., and Ferrara, N. VEGF 1998. couples hypertrophic cartilage remodeling, ossification and angiogenesis during 148. Jussila, L., Valtola, R., Partanen, T. A., Salven, P., Heikkila¨, P., Matikainen, M-T., Renkonen, R., Kaipainen, A., Detmar, M., Tschachler, E., Alitalo, R., and Alitalo, endochondral bone formation. Nat. Med., 5: 623–628, 1999. K. Lymphatic endothelium and Kaposi’s sarcoma spindle cells detected by antibod- 173. Niida, S., Kaku, M., Amano, H., Yoshida, H., Kataoka, H., Nishikawa, S., Tanne, ies against the vascular endothelial growth factor receptor-3. Cancer Res., 58: K., Maeda, N., Nishikawa, S-I., and Kodama, H. Vascular endothelial growth factor 1599–1604, 1998. can substitute for macrophage colony-stimulating factor in the support of osteaclas- 149. Boshoff, C., and Weiss, R. A. Aetiology of Kaposi’s sarcoma: current understanding tic bone resorption. J. Exp. Med., 190: 293–298, 1999. and implications for therapy. Mol. Med. Today, 3: 488–494, 1997. 174. Dumont, D. J., Gradwohl, G., Fong, G-H., Puri, M. C., Gertsenstein, M., Auerbach, 150. Bais, C., Santomasso, B., Coso, O., Arvanitakis, L., Raaka, E. G., Gutkind, J. S., A., and Breitman, M. L. Dominant-negative and targeted null mutations in the Asch, A. S., Cesarman, E., Gerhengorn, M. C., and Mesri, E. A. G-protein-coupled endothelial receptor tyrosine kinase, tek, reveal a critical role in vasculogenesis of receptor of Kaposi’s sarcoma-associated herpesvirus is a viral oncogene and angio- the embryo. Genes Dev., 8: 1897–1909, 1994. genesis activator. Nature (Lond.), 391: 86–89, 1998. 175. Puri, M. C., Rossant, J., Alitalo, K., Bernstein, A., and Partanen, J. The receptor 151. Masood, R., Cai, J., Zheng, T., Smith, D. L., Naidu, Y., and Gill, P. S. Vascular tyrosine kinase TIE is required for integrity and survival of vascular endothelial endothelial growth factor/vascular permeability factor is an autocrine growth factor cells. EMBO J., 14: 5884–5891, 1995. for AIDS-Kaposi sarcoma. Proc. Natl. Acad. Sci. USA, 94: 979–984, 1997. 176. Sato, T. N., Tozawa, Y., Deutsch, U., Wolburg-Buchholz, K., Fujiwara, Y., 152. Albini, A., Soldi, R., Giunciuglio, D., Giraudo, E., Benelli, R., Primo, L., Noonan, Gendron-Maguire, M., Gridley, T., Wolburg, H., Risau, W., and Qin, Y. Distinct D., Salio, M., Camussi, G., Rockl, W., and Bussolino, R. The angiogenesis induced roles of the receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel formation. by HIV-1 Tat protein is mediated by the Flk-1/KDR receptor on vascular endothelial Nature (Lond.), 376: 70–74, 1995. cells. Nat. Med., 2: 1371–1375, 1996. 177. Partanen, J., and Dumont, D. J. Functions of Tie1 and Tie2 receptor tyrosine kinases 153. Ganju, R. K., Munshi, N., Nair, B. C., Liu, Z-Y., Gill, P., and Groopman, J. E. in vascular development. In: L. Claesson-Welsh (ed.), Vascular Growth Factors and Human immunodeficiency virus Tat modulates the Flk-1/KDR receptor, mitogen- Angiogenesis, pp. 159–172. Berlin: Springer-Verlag, 1999. 211

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. REGULATION OF ANGIOGENESIS VIA VEGFRs

178. Wang, H. U., Chen, Z-F., and Anderson, D. J. Molecular distinction and angiogenic 190. Mandriota, S. J., and Pepper, M. S. Regulation of angiopoietin-2 mRNA levels in interaction between embryonic arteries and veins revealed by ephrin-B2 and its bovine microvascular endothelial cells by cytokines and hypoxia. Circ. Res., 83: receptor Eph-B4. Cell, 93: 741–753, 1998. 852–859, 1998. 179. Adams, R. H., Wilkinson, G. A., Weiss, C., Diella, F., Gale, N. W., Deutsch, U., 191. Mandriota, S. J., Pyke, C., Di Sanza, C., Quinodoz, P., Pittet, B., and Pepper, M. S. Risau, W., and Klein, R. Roles of ephrinB ligands and EphB receptors in cardio- Hypoxia-inducible angiopoietin-2 expression is mimicked by diphenyle iodonium vascular development: demarcation of arterial/venous domains, vascular morpho- and occurs in the rat brain and skin in response to systemic hypoxia. Am. J. Pathol., genesis, and sprouting angiogenesis. Genes Dev., 13: 295–306, 1999. in press, 2000. 180. Gale, N. W., and Yancopoulos, G. D. Growth factors acting via endothelial cell- 192. Huynh-Do, U., Stein, E., Lane, A. A., Liu, H., Cerretti, D. P., and Daniel, T. O. specific receptor tyrosine kinases: VEGFs, , and ephrins in vascular Surface densities of ephrin-B1 determine EphB1-coupled activation of cell attach- development. Genes Dev., 13: 1055–1066, 1999. ment through ␣ ␤ and ␣ ␤ . EMBO J., 18: 2165–2173, 1999. 181. Davis, S., Aldrich, T. H., Jones, P. F., Acheson, A., Compton, D. L., Jain, V., Ryan, v 3 5 1 193. Leveen, P., Pekny, M., Gebre-Medhin, S., Swolin, B., Larsson, E., and Betsholtz, C. T. E., Bruno, J., Raziejewski, C., Maisonpierre, P. C., and Yancopoulos, G. D. Mice deficient for PDGF B show renal, cardiovascular, and hematological abnor- Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expres- malities. Genes Dev., 8: 1875–1887, 1994. sion cloning. Cell, 87: 1161–1169, 1997. 182. Maisonpierre, P. C., Suri, C., Jones, P. F., Bartunkova, S., Wiegand, S. J., 194. Lindahl, P., Johansson, B. R., Leveen, P., and Betsholtz, C. Pericyte loss and Radziejewski, C., Compton, D., McClain, J., Aldrich, T. H., Papadopoulos, N., Daly, microaneurysm formation in PDGF-B-deficient mice. Science (Washington DC), T. J., Davis, S., Sato, T. N., and Yancopoulos, G. D. Angiopoietin-2, a natural 277: 242–245, 1997. ␤ antagonist for Tie2 that disrupts in vivo angiogenesis. Science (Washington DC), 195. Pepper, M. S. Transforming growth factor- : vasculogenesis, angiogenesis, and 277: 55–60, 1997. vessel wall integrity. Cytokine Growth Factor Rev., 8: 21–43, 1997. 183. Valenzuela, D. M., Griffiths, J. A., Rojas, J., Aldrich, T. H., Jones, P. F., 196. Li, D. Y., Sorensen, L. K., Brooke, B. S., Urness, L. D., Davis, E. C., Taylor, D. G., Zhou, H., McClain, J., Copeland, N. G., Gilbert, D. J., Jenkins, N. A., Huang, T., Boak, B. B., and Wendel, D. P. Defective angiogenesis in mice lacking endoglin. Papadopoulous, N., Maisonpierre, P. C., Davis, S., and Yancopoulous, G. D. Science (Washington DC), 284: 1534–1537, 1999. Angiopoietins 3 and 4: diverging gene counterparts in mice and humans. Proc. Natl. 197. Miller, D. W., Graulich, W., Karges, B., Stahl, S., Ernst, M., Ramaswamy, A., Acad. Sci. USA, 96: 1904–1909, 1999. Sedlacek, H. H., Muller, R., and Adamkiewicz, J. Elevated expression of endoglin, 184. Nishimura, M., Miki, T., Yashima, R., Yokoi, N., Yano, H., Sato, Y., and Seino, S. a component of the TGF-␤-receptor complex, correlates with proliferation of tumor Angiopoietin-3, a novel member of the angiopoietin family. FEBS Lett., 448: endothelial cells. Int. J. Cancer, 81: 568–572, 1999. 254–256, 1999. 198. Roelen, B. A., van Rooijen, M. A., and Mummery, C. L. Expression of ALK-1, a 185. Kim, I., Kwak, H. J., Ahn, J. E., So, J. N., Liu, M., Koh, K. N., and Koh, G. Y. type 1 serine/threonine kinase receptor, coincides with sites of vasculogenesis and Molecular cloning and characterizatin of a novel angiopoietin family protein, an- angiogenesis in early mouse development. Dev. Dyn., 209: 418–430, 1997. giopoietin-3. FEBS Lett., 443: 353–356, 1999. 199. McAllister, K. A., Grogg, K. M., Johnson, D. W., Gallione, C. J., Baldwin, M. A., 186. Thurston, G., Suri, C., Smith, K., McClain, J., Sato, T. N., Yancopoulos, G. D., and Jackson, C. E., Helmbold, E. A., Markel, D. S., McKinnon, W. C., Murrell, J., McDonald, D. Angiopoietin-1 makes blood vessels resistant to plasma leakage. McCormick, M. K., Pericak-Vance, M. A., Heutink, P., Oostra, B. A., Haitjema, T., Science, 286: 2511–2514, 2000. Westerman, C. J. J., Porteous, M. E., Guttmacher, A. E., Letarte, M., and Marchuk, 187. Peters, K. G., Coogan, A., Berry, D., Marks, J., Iglehart, J. D., Kontos, C. D., Rao, D. A. Endoglin, a TGF-␤ binding protein of endothelial cells, is the gene for P., Sankar, S., and Trogan, E. Expression of Tie2/Tek in breast tumour vasculature hereditary haemorrhagic telangiectasia type 1. Nat. Genet., 8: 345–351, 1994. provides a new marker for evaluation of tumour angiogenesis. Br. J. Cancer, 77: 51–56, 1998. 200. Berg, J. N., Gallione, C. J., Stenzel, T. T., Johnson, D. W., Allen, W. P., Schwartz, 188. Siemeister, G., Schirner, M., Weindel, K., Reusch, P., Menrad, A., Marme, D., and C. E., Jackson, C. E., Porteous, M. E., and Marchuk, D. A. The activin receptor-like Martiny-Baron, G. Two independent mechanisms essential for tumor angiogenesis: kinase 1 gene: genomic structure and mutations in hereditary hemorrhagic telangi- inhibition of human melanoma xenograft growth by interfering with either the ectasia type 2. Am. J. Hum. Genet., 61: 60–67, 1997. vascular endothelial growth factor receptor pathway or the Tie-2 pathway. Cancer 201. Uyttendaele, H., Marazzi, G., Wu, G., Yan, Q., Sassoon, D., and Kitajewski, J. Res., 59: 3185–3191, 1999. Notch4/int-3, a mammary proto-oncogene, is an endothelial cell-specific mamma- 189. Holash, J., Maisonpierre, P. C., Compton, D., Boland, P., Alexander, C. R., Zagzag, lian Notch gene. Development (Camb.), 122: 2251–2259, 1996. D., Yancopoulos, G. D., and Wiegand, S. J. Vessel cooption, regression and growth 202. Fachinger, G., Deutsch, U., and Risau, W. Functional interaction of vascular in tumors mediated by angiopoietins and VEGF. Science (Washington DC), 284: endothelial-protein-tyrosine phosphatase with the Angiopoietin receptor Tie-2. On- 1994–1998, 1999. cogene 18: 5948–5953, 1999.

212

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research. Regulation of Angiogenesis via Vascular Endothelial Growth Factor Receptors

Tanja Veikkola, Marika Karkkainen, Lena Claesson-Welsh, et al.

Cancer Res 2000;60:203-212.

Updated version Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/60/2/203

Cited articles This article cites 195 articles, 91 of which you can access for free at: http://cancerres.aacrjournals.org/content/60/2/203.full#ref-list-1

Citing articles This article has been cited by 82 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/60/2/203.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/60/2/203. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 2, 2021. © 2000 American Association for Cancer Research.