vascular endothelial growth factor as a survival factor in tumor

18
Abstract. This review focuses on v ascular e ndothelial g rowth f actors (VEGF), a group of structurally-related proteins that serve as key growth factors for tumor-associated angiogenesis. Pathways induced by VEGF proteins that regulate biological functions of key cell types involved in tumor angiogenesis, including vascular endothelial cells, pericytes and tumor cells, are discussed. Strategies that are currently being developed and tested based on the emerging definitions of the roles of the multiple cell types involved in tumor vessel development, their selective production of VEGF-related proteins and other pro- angiogenic growth factors, their expression of associated receptors as well as identification of signal transduction pathways involved in VEGF-induced tumor survival and tumor-associated angiogenesis will be reviewed. Neovascularization is the development of blood vessels and is a fundamental and highly regulated process necessary for appropriate embryonic development (1). In adult mammals, this activity occurs only during normal physiological processes such as the female reproductive cycle (2) and wound healing (3). While tumors that are relatively small (< 200 Ìm) use existing vasculature as a source of oxygen and nutrients, in a process termed vessel co-option (4), tumors that are beyond this size have increased nutritional requirements and exist within a hypoxic, metabolically- deficient microenvironment. Studies over the last four decades have demonstrated that actively growing tumors have an absolute requirement for the development of new blood vessels, which support increases in tumor mass, their survival and provide an avenue for tumor invasion and metastasis. Without this process, defined as tumor- associated angiogenesis (5), apoptotic signal transduction pathways are triggered, leading to tumor dormancy and regression. Growth Factors in Tumor-associated Angiogenesis Developing tumors are in a prevascular state and exist in a state of equilibrium between proliferation and apoptosis. As tumor cells proliferate and their layers accumulate, the distance between malignant cells within the expanding tumor mass and the nearest blood vessel increases. When tumors reach a size of 1-2 mm, tumor cells located at a distance greater than 200 Ìm from the nearest blood vessel become hypoxic and are deficient in critical nutrients (6). Without initiation of tumor-associated angiogenesis, tumor growth is restricted (7) and tumors become dormant (8). To allow for tumor expansion, survival and metastasis, tumors undergo what has been defined as the "angiogenic switch", which results from a shift in the balance of anti-angiogenic growth factors that inhibit the formation of new vasculature to favor the production of pro-angiogenic growth factors such as VEGF which induce tumor-associated angiogenesis (9,10) (Figure 1A). Tumor-associated angiogenesis is a complex process that involves the coordinated activity of a number of different cell types within the tumor microenvironment, including tumor cells and stromal cell populations of vascular endothelial cells and pericytes (11). While the literature is replete with studies that define the role of tumor cells during tumor-associated angiogenesis, the role of vascular endothelial cells is currently being elucidated. In contrast, the role of pericytes during this 525 Correspondence to: F.M. Robertson, Department of Molecular Virology, Immunology, and Medical Genetics, 2184 Graves Hall, 333 West 10th Avenue, Columbus, OH 43210, U.S.A. Tel: (614) 499-1833, Fax: (614) 292-0643, e-mail: [email protected] Key Words: Akt/Protein kinase B, anti-angiogenic drugs, inositidylphosphate-3-kinase (IP3-K), pericytes, platelet-derived growth factor beta, (PDGF‚), platelet-derived growth factor receptor beta, (PDGFR-‚), review, splice variant, angiogenesis, vascular endothelial cells, vascular endothelial growth factor (VEGF), vascular targeting. in vivo 18: 525-542 (2004) Review Vascular Endothelial Growth Factor as a Survival Factor in Tumor-associated Angiogenesis NESRINE I. AFFARA and FREDIKA M. ROBERTSON Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210, U.S.A. 0258-851X/2004 $2.00+.40

Upload: lexuyen

Post on 23-Jan-2017

218 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

Abstract. This review focuses on vascular endothelial growthfactors (VEGF), a group of structurally-related proteins thatserve as key growth factors for tumor-associated angiogenesis.Pathways induced by VEGF proteins that regulate biologicalfunctions of key cell types involved in tumor angiogenesis,including vascular endothelial cells, pericytes and tumor cells,are discussed. Strategies that are currently being developed andtested based on the emerging definitions of the roles of themultiple cell types involved in tumor vessel development, theirselective production of VEGF-related proteins and other pro-angiogenic growth factors, their expression of associatedreceptors as well as identification of signal transductionpathways involved in VEGF-induced tumor survival andtumor-associated angiogenesis will be reviewed.

Neovascularization is the development of blood vessels and

is a fundamental and highly regulated process necessary for

appropriate embryonic development (1). In adult mammals,

this activity occurs only during normal physiological

processes such as the female reproductive cycle (2) and

wound healing (3). While tumors that are relatively small

(< 200 Ìm) use existing vasculature as a source of oxygen

and nutrients, in a process termed vessel co-option (4),

tumors that are beyond this size have increased nutritional

requirements and exist within a hypoxic, metabolically-

deficient microenvironment. Studies over the last four

decades have demonstrated that actively growing tumors

have an absolute requirement for the development of new

blood vessels, which support increases in tumor mass, their

survival and provide an avenue for tumor invasion and

metastasis. Without this process, defined as tumor-

associated angiogenesis (5), apoptotic signal transduction

pathways are triggered, leading to tumor dormancy and

regression.

Growth Factors in Tumor-associated Angiogenesis

Developing tumors are in a prevascular state and exist in a

state of equilibrium between proliferation and apoptosis.

As tumor cells proliferate and their layers accumulate, the

distance between malignant cells within the expanding

tumor mass and the nearest blood vessel increases. When

tumors reach a size of 1-2 mm, tumor cells located at a

distance greater than 200 Ìm from the nearest blood vessel

become hypoxic and are deficient in critical nutrients (6).

Without initiation of tumor-associated angiogenesis, tumor

growth is restricted (7) and tumors become dormant (8). To

allow for tumor expansion, survival and metastasis, tumors

undergo what has been defined as the "angiogenic switch",

which results from a shift in the balance of anti-angiogenic

growth factors that inhibit the formation of new vasculature

to favor the production of pro-angiogenic growth factors

such as VEGF which induce tumor-associated angiogenesis

(9,10) (Figure 1A). Tumor-associated angiogenesis is a

complex process that involves the coordinated activity of a

number of different cell types within the tumor

microenvironment, including tumor cells and stromal cell

populations of vascular endothelial cells and pericytes (11).

While the literature is replete with studies that define the

role of tumor cells during tumor-associated angiogenesis,

the role of vascular endothelial cells is currently being

elucidated. In contrast, the role of pericytes during this

525

Correspondence to: F.M. Robertson, Department of Molecular

Virology, Immunology, and Medical Genetics, 2184 Graves Hall,

333 West 10th Avenue, Columbus, OH 43210, U.S.A. Tel: (614)

499-1833, Fax: (614) 292-0643, e-mail: [email protected]

Key Words: Akt/Protein kinase B, anti-angiogenic drugs,

inositidylphosphate-3-kinase (IP3-K), pericytes, platelet-derived

growth factor beta, (PDGF‚), platelet-derived growth factor

receptor beta, (PDGFR-‚), review, splice variant, angiogenesis,

vascular endothelial cells, vascular endothelial growth factor

(VEGF), vascular targeting.

in vivo 18: 525-542 (2004)

Review

Vascular Endothelial Growth Factor as aSurvival Factor in Tumor-associated Angiogenesis

NESRINE I. AFFARA and FREDIKA M. ROBERTSON

Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210, U.S.A.

0258-851X/2004 $2.00+.40

Page 2: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

process has only recently been recognized and remains to

be completely defined. Figure 1B provides a schematic

representation of tumor-associated angiogenesis and the

role of these individual cell types in this process. Following

the angiogenic switch, tumors acquire an angiogenic

phenotype, produce pro-angiogenic growth factors such as

VEGF and express the associated VEGF receptors, which

confers the ability to respond to these proteins in an

autocrine manner.

VEGF production is stimulated following activation or

mutation of oncogenes such as Bcr-Abl, ras and erbB-

2/HER-2/neu (12-14). In addition, the hypoxic tumor

microenvironment and activation of survival pathways work

synergistically with oncogenes to increase VEGF production

(15,16). As a pro-angiogenic growth factor, VEGF activates

vascular endothelial cells within the capillaries adjacent to

expanding tumors to express specific VEGF receptors which

allow these cells to respond to VEGF. Once activated by

VEGF, vascular endothelial cells secrete proteolytic enzymes

that degrade basement membranes and the extracellular

matrix surrounding the pre-existing blood vessel. Digestion

of these elements allows the vascular endothelial cells to

detach from pericytes that reside beneath the vascular

endothelial cells. Following their detachment, vascular

endothelial cells proliferate and migrate toward the source

of pro-angiogenic growth factor production by invasion

in vivo 18: 525-542 (2004)

526

Figure 1. Schematic of tumor-associated angiogenesis. A) "The angiogenic switch" results from production of inducers of angiogenesis which are pro-angiogenic growth factors with a simultaneous down-regulation of inhibitors of angiogenesis. B) Tumor cells and infiltrating inflammatory cells, includingmast cells, neutrophils and macrophages, produce VEGF splice variants that activate vascular endothelial cells lining adjacent blood vessels. Theendothelial cells then detach from the underlying pericytes and extracellular matrix, proliferate, migrate and then organize and differentiate into capillarytubes which provide oxygen and nutrients to expanding tumors.

Page 3: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

through spaces created by enzyme degradation (Figure 1B).

These recruited vascular endothelial cells then secrete

extracellular matrix proteins which provide the building

blocks for the development of new basement membranes

that support the tumor vessels (17,18). In addition, vascular

endothelial cells secrete VEGF and an additional protein,

platelet-derived growth factor beta (PDGF‚), which induce

migration of pericytes into the area of active tumor vessel

formation (19).

The vascular endothelial cells then change from an

actively proliferating population of cells to subsequently

undergo differentiation and organization into hollow tubes

that connect the expanding tumor mass to nearby vascular

beds. Direct interactions between pericytes and vascular

endothelial cells stimulate differentiation of pericytes into

vascular smooth muscle cells that stabilize and maintain the

newly-formed tumor vasculature (20). Blood flow is then

initiated which inhibits the rate of apoptosis within tumors,

providing both a source of oxygen and nutrients and a route

for tumor invasion and metastasis to distant organ sites.

VEGF also acts as a chemotactic factor which results in

recruitment and activation of non-vascular cells, including

inflammatory mast cells, macrophages and neutrophils (Figure

1B) (21-23). Recruited inflammatory cells produce pro-

angiogenic products as well as synthesize matrix metallo-

proteinases (24-29), which has led to these cells being defined

as co-conspirators in tumor-associated angiogenesis (26).

Other cell types have also been identified as playing a role in

tumor-associated angiogenesis such as bone marrow-derived

stromal cells, which respond to pro-angiogenic growth factors,

migrate from the bone marrow to the tumor site and

differentiate into capillary-like structures (30). Additionally,

in a unique process defined as vasculogenic mimicry, tumor

cells within aggressively growing malignancies such as

melanomas trans-differentiate into cells with phenotypic and

functional characteristics of vascular endothelial cells (31).

These cells form a vascular network rich in extracellular

matrix proteins without the need to activate endothelial cells

that are usually required for angiogenesis. It is clear that

further investigation will reveal additional cell types that

contribute to tumor vessel formation as well as elucidate

previously undefined contributions of cells that are known to

play a role in tumor-associated angiogenesis.

Early studies demonstrated the presence of tumor

angiogenesis factors (TAF), which were initially defined as

soluble factors that diffuse into the tumor microenvironment

(32). Although the first pro-angiogenic growth factor

identified was basic fibroblast growth factor (bFGF, FGF-2)

(33), this protein is now believed to be a general angiogenic

growth factor that is not critical for angiogenesis, based on

studies demonstrating that the loss of bFGF does not result

in any significant phenotypic alteration in the vasculature

during embryonic development (34).

The second angiogenic growth factor was identified based

on its ability to induce vascular permeability in guinea pig

skin and was named vascular permeability factor (VPF) (35).

VPF was later found to be similar to proteins purified from

pituitary folliculostellate cells that stimulated proliferation

of cultured endothelial cells and was therefore defined as

vascular endothelial growth factor-A (VEGF-A) (36-38).

Studies using knock-out mice demonstrated that the loss of

only one VEGF-A allele resulted in early embryonic

lethality, indicating that VEGF-A was critical to angiogenesis

during development (39,40). Studies have revealed that

VEGF-A is part of a family of VEGF-related molecules

including VEGF-B (41), VEGF-C (42,43), VEGF-D (44),

VEGF-E (45,46) and placental-derived growth factor (PlGF)

(47). For the purpose of this review, VEGF-A will be

designated as VEGF.

VEGF is recognized as a pivotal growth factor in tumor-

associated angiogenesis (48). Since it can activate and

recruit vascular endothelial cells to undergo all of the

necessary activities required to form new capillary beds,

VEGF has been defined as a complete angiogenic growth

factor (49). VEGF stimulates vasodilation and increases

permeability of pre-existing capillaries, leading to leakage

of plasma proteins (50,51) and deposition of an

extravascular fibrin gel around developing vasculature. This

provides a matrix to support the subsequent proliferation

and migration of vascular endothelial cells (52). As an

additional critical component of its activity as a complete

angiogenic growth factor, VEGF activates signal

transduction pathways that regulate survival of tumor cells

(53-55), vascular endothelial cells (56-58) and pericytes (59),

which exist in the tumor microenvironment that is hypoxic

and deprived of nutrients. The pivotal nature of VEGF to

tumor-associated angiogenesis is demonstrated by the

detection of its presence in all tumors thus far examined and

by the correlation between production of VEGF with tumor

stage and tumor progression (60,61).

In addition to bFGF and VEGF-related proteins, other

pro-angiogenic growth factor families have been identified

which have distinct activities in tumor-associated angiogenesis

and act in concert with VEGF. Angiopoietin-1 (Ang-1)

(62,63), Ang-2 (64), Ang-3 and Ang-4 (65) are members of

the Angiopoietin family of proteins which bind to receptors

within the tyrosine kinase immunoglobulin-like and

epidermal growth factor homology domains (Tie) receptor

family that are expressed by tumor cells, vascular endothelial

cells and pericytes. During embryonic development, ligand

binding of Ang-1 to the Tie-2 receptor tyrosine kinase

increases adherence of vascular endothelial cells to pericytes,

resulting in stabilization of newly-formed blood vessels (66).

In addition, Ang-1 regulates vascular endothelial cell survival,

sprouting and organization into tubules (67). Ang-1 can also

inhibit VEGF-induced permeability and reduce inflammation

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

527

Page 4: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

(68). Despite the fact that each of the ligands bind to the

same receptor, Ang-2 and Ang-3 inhibit the Tie-2 tyrosine

phosphorylation induced by Ang-1, which counteracts the

stabilizing effects of Ang-1 (5). Molecular profiling studies

have demonstrated that VEGF up-regulates Ang-2 during

tumor-associated angiogenesis, resulting in destabilization of

existing vasculature, loss of pericytes and subsequent

enhancement of tumor vessel formation (69,70). The co-

operative activities of VEGF and Ang-2 may, in part, account

for the morphologically distinct tortuous blood vessels

formed during tumor-associated angiogenesis and may also

be responsible for the less stable, leaky vessels that develop

adjacent to expanding tumors (71). Although the members of

the Angiopoietin family and their associated Tie-2 receptor

in vivo 18: 525-542 (2004)

528

Figure 2. Structure and functions of human and murine VEGF exons. A) The human VEGF gene gives rise to 7 distinct VEGF splice variants, includingVEGF121, VEGF145, VEGF165, VEGF165b, VEGF183, VEGF189 and VEGF206. VEGF165b, which has inhibitory activity, lacks exons 6 and 8 andcontains a novel 6 amino acid sequence SLTRKD, designated as exon 9. B) The mouse VEGF gene gives rise to splice variants that are all one aminoacid shorter than the corresponding human splice variants. Mouse splice variants include VEGF120, VEGF144, VEGF164 and VEGF188. The uniquemouse splice variant VEGF205* lacks exons 7 and 8 and contains exon 6, which includes a 61 bp extension of exon 6a, designated as exon 6’, whichencodes a unique 7 amino acid carboxyl-terminal tail, YVGAAAV, that is significantly different from the carboxyl-terminal tail of any VEGF proteinidentified to date.

Page 5: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

have opposing roles in angiogenesis, it is clear that the

balance in production of these growth factors plays a key role

in regulating tumor vessel formation (72). Disruption of this

delicate balance in relative amounts of Ang-1 or Ang-2 has

been observed to result in significant defects in tumor-

associated angiogenesis (73). Recent evidence suggests that

VEGF, Tie-2, Ang-1 and Ang-2 are simultaneously expressed

during tumor growth, yet are differentially-expressed spatially

within the tumor microenvironment as well as temporally-

expressed during tumor vessel development (71). It is clear

that further studies, which continue to define the roles of the

members of this family of proteins, will provide insight into

their use as therapeutics and as targets for therapeutics, not

only in VEGF-induced tumor-associated angiogenesis, but

also in disorders associated with enhanced inflammatory

responses.

In addition to the ability of tumor cells and vascular

endothelial cells to respond to VEGF and the Angiopoietins

during tumor-associated angiogenesis, pericytes also

respond to pro-angiogenic growth factors during this

process (74). Pericytes produce VEGF (75) as well as

respond to it by expression of VEGF receptors (76),

resulting in enhanced recruitment of pericytes to the local

tumor microenvironment and stabilization of newly-

developed tumor vessels. In addition, vascular endothelial

cells produce PDGF‚. Pericytes, as well as their precursors

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

529

Figure 3. VEGF acts as a survival factor through IP3-K/Akt. IP3-K is activated by binding of VEGF to VEGFR-2. IP3-K then catalyzes the productionof phosphatidylinositol 3,4,5-triphosphate (PI-3,4,5,-P3), which binds to the pleckstrin homology domain (PH) of Akt and PDK-1. Akt is then translocatedfrom the cytoplasm to the plasma membrane and undergoes conformational changes. Akt can then be phosphorylated at Thr308 by PDK1 and at Ser473by ILK. Activated Akt then promotes VEGF-induced endothelial cell survival by phosphorylating and inhibiting the pro-apoptotic proteins of the Forkheadtranscription factors and by increasing the expression of the anti-apoptotic FLICE-inhibitory protein (FLIP). VEGF also promotes increased cell massby activation of mTOR/S6K. Activated Akt also induces cell cycle entry by inactivating glycogen synthase kinase-3 beta (GSK-3‚), which results instabilization of cyclin D1 and by inhibiting p21Cip1/WAF1. LY292004 is a specific inhibitor of IP3-K, while Rapamycin is a selective inhibitor of mTOR.

Page 6: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

derived from mesenchymal cells, respond to PDGF‚ viaexpression of the associated receptor tyrosine kinase,

platelet derived growth factor receptor beta (PDGFR-‚).

Further studies are required to more completely define the

interactions of pro-angiogenic growth factors with each of

the cell types involved in tumor-associated angiogenesis.

Structure and Function of Human and MurineVEGF Alternative Splice Variants

VEGF is encoded for by a single gene organized into 8

exons in mice and rats and 9 exons in humans (Figure 2).

Alternative splicing generates distinct peptides named for

the number of amino acids contained within them (77-80).

In humans, the most abundantly produced VEGF splice

variants are VEGF121, VEGF165 and VEGF189 (78) (Figure

2A). The less abundantly produced human VEGF splice

variants identified to date are VEGF145 (80), VEGF183 (81),

and VEGF206 (79). Although the majority of human VEGF

splice variants are pro-angiogenic, an anti-angiogenic splice

variant VEGF165b was found to be down-regulated in renal

tumors, and the loss of VEGF165b coincided with induction

of the angiogenic switch in this tissue (82). VEGF165b lacks

exon 8, which is replaced by exon 9, resulting in the

production of a protein with the same length as VEGF165

but with a carboxyl-terminal tail composed of 6 unique

amino acids, SLTRKD (Figure 2A). A very recent study

reported that VEGF165b is present in differentiated renal

podocytes but was not present in this cell type undergoing

active proliferation, suggesting that alternative splicing of

VEGF pre-mRNA is an integral part of regulating

neovascularization (83). Further studies may reveal the

existence of other human VEGF splice variants with

functional activities that vary from those described to date.

Elucidation of the activities of the distinct VEGF splice

variants may provide insight into their utility as both

therapeutics and therapeutic targets.

In contrast to human VEGF splice variants, murine

VEGF variants are one amino acid shorter than the

analogous human splice variants, but in most cases the

mouse VEGF proteins possess biological activities similar

to human VEGF splice variants (Figure 2B). The murine

VEGF splice variants identified to date include VEGF120,

VEGF144, VEGF164 and VEGF188 (84). In addition to these

murine proteins, our laboratory identified, sequenced and

characterized the activities of a novel murine splice variant

designated as VEGF205* (Genbank Accession AY120866)

(Figure 2B). This VEGF splice variant was first identified

by detection of its mRNA and protein only in mouse skin

papillomas and squamous cell carcinomas, which was

completely undetectable in normal skin (85). VEGF205*

contains exons 1-6 and lacks exons 7 and 8. In place of these

exons is a 61 bp extension to exon 6a, defined as exon 6’

(Figure 2B). Due to the presence of a stop codon in exon

6’, translation of murine VEGF205* mRNA results in the

production of a truncated 145 amino acid protein containing

a carboxyl-terminal tail consisting of 7 unique amino acids,

YVGAAAV. The exon structure of murine VEGF205* and

the resultant VEGF splice variant protein are significantly

different from any other human or mouse VEGF splice

in vivo 18: 525-542 (2004)

530

Figure 4. VEGF205* stimulates phospho-Akt-1-Ser473 which is inhibited by LY294002. A) Western blots of anti-phospho-Akt-1-Ser-473, Akt-1 and ‚-actin. B) Densitometric analysis of Western blots shown in A. Data are shown as the ratio of phospho-Akt-1-Ser473 to Akt-1 and are expressed as mean± S.E., N=3. *, p<0.02.

Page 7: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

531

Figure 5. Strategies to target cells involved in tumor-associated angiogenesis. A) Targeting tumor cells and tumor-associated endothelial cells. Avastinìis a humanized anti-VEGF monoclonal antibody (MAb VEGF) that binds and neutralizes VEGF. VEGFR-2 chimeric receptors are composed of theextracellular VEGF-binding domain of VEGFR-2 and the intracellular domain of the apoptotic receptor Fas. Following binding of VEGF, apoptosis ofboth tumor cells and vascular endothelial cells is induced. B) Targeting vascular endothelial cells and pericytes. Blockade of Tie-2 and PDGFR-‚ receptortyrosine kinases using Gleevecì/Imatinib, SU6668, or SU11248 targets pericytes and tumor endothelial cells. SU5416 and Avastinì block VEGFR-2. C)Targeting tumor cells, vascular endothelial cells and pericytes. LY294002 can inhibit survival of tumor cells, vascular endothelial cells and pericytes.

Page 8: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

in vivo 18: 525-542 (2004)

532

variant described to date, including human VEGF206

(Figure 2 A and B). The size, amino acid sequence of the

carboxyl-terminal tail and biological activities of VEGF205*

are unique.

The biological activity of VEGF splice variants occurs

following receptor-ligand binding and subsequent

transduction of specific signals. Identification of VEGF

receptor families has been crucial to understanding the

functions of the individual VEGF splice variants. Thus far,

there have been two distinct VEGF receptor families

identified, which include the VEGF receptor tyrosine kinase

family and the Neuropilin family. The VEGF receptor

tyrosine kinase family includes VEGF Receptor-1

(VEGFR-1), which was first isolated and characterized as

Fms-like tyrosine kinase-1 (Flt-1) (86-88), VEGFR-2, also

defined as Fetal Liver Kinase 1/Kinase-insert- Domain-

containing Receptor (Flk-1/KDR) (89,90) and VEGFR-3,

first defined as Flt-4 (91).

Despite their structural homology and the ability of

VEGFR-1 and VEGFR-2 to bind VEGF and induce

receptor autophosphorylation at specific tyrosine residues,

these receptors have distinct expression patterns within

different organs, which may overlap, and they also activate

specific signal transduction pathways (92). The differential

ability of VEGF receptor-ligand binding to induce selective

biological activities, including increased vascular permeability

as well as proliferation, migration, differentiation and

organization of vascular endothelial cells into vascular tubules

during tumor-associated angiogenesis, is due to phosphorylation

of specific VEGFR-associated tyrosine residues (93). For

example, following binding of VEGF to VEGFR-2, vascular

endothelial cells undergo proliferation, which is due to

phosphorylation of tyrosine residue 1059, while migration of

these cells is stimulated following phosphorylation of tyrosine

residue 951 (94). Conversely, binding of VEGF to VEGFR-1

inhibits VEGFR-2-mediated endothelial cell proliferation, but

not migration (95). VEGFR-1 can also act as a decoy

receptor that sequesters VEGF, preventing it from binding

to VEGFR-2 (96,97). In contrast to the ability of VEGF

splice variants to differentially bind VEGFR-1 and VEGFR-

2, VEGFR-3 binds only to VEGF-C and VEGF-D, and this

VEGF receptor is primarily involved in, and is critical to,

appropriate development of lymphatic vessels (98). In

addition, VEGFR-3 is expressed on a variety of tumor cells

of different origins and may be useful as a marker of early

stages of tumor progression (42,99). Although further studies

are required to completely define the roles of these VEGF

receptors in tumor-associated angiogenesis, there are a

number of therapeutic approaches that are based on the

interactions of VEGF and VEGFR-2 currently being

developed that target tumor vessels (100-102).

The Neuropilins are a second family of VEGF receptors

whose members identified thus far include neuropilin-1 (NP-1)

(103) and neuropilin-2 (NP-2) (104). NP-1 and NP-2 belong to

a family of collapsins/semaphorins receptors first described as

neuronal axon guidance factors present in developing

embryos (105,106). These receptors are now known to be

expressed by tumor cells, vascular endothelial cells and

pericytes (103,104). Both NP-1 and NP-2 are alternatively

spliced (107) and contain short intracellular domains which,

at least in part, confer the biological activities of these

receptors (108). NP-1 and NP-2 bind VEGF165 as well as

the heparin-binding form of PlGF, PlGF-2 (109) and

VEGF-B (110). NP-1 acts as a co-receptor for VEGFR-2

(103), resulting in enhanced binding affinity of VEGF165 to

VEGFR-2 and effectively increasing the local concentration

of VEGFR-2 at the cell surface (111,112). Only NP-2 can

bind VEGF145 (104). In contrast, neither NP-1 nor NP-2

binds VEGF121, a VEGF splice variant which lacks the

ability to bind heparin and heparan sulfate proteoglycans

(103,104). These observations have prompted speculation

that one of the primary functions of NP-1 and NP-2 is to act

as receptors for specific VEGF splice variants. Taken

together with a recent report that blocking NP-1 effectively

inhibits angiogenesis in murine retinal neovascularization

(113), these studies suggest that the Neuropilin receptors

may serve as targets for the development of VEGF splice

variant-specific anti-angiogenic therapies. Further studies are

necessary to determine whether there are other members of

this receptor family, to fully elucidate the role of these

receptors in tumor-associated angiogenesis and to determine

if these receptors can be used to target this process.

Production of multiple VEGF splice variants allows this

one group of proteins to regulate specific functions of tumor

cells, vascular endothelial cells and pericytes, resulting in

precise regulation of tumor-associated angiogenesis

(49,55,58,59). In part, the distinct biological activities of

VEGF splice variants are determined by the exons present

within the splice variant pre-mRNA (Figure 2 A and B).

Exon 1 encodes for the signal sequence common to all

VEGF splice variants. The presence of Exon 1 indicates that

each of the splice variant proteins can be secreted. Exon 2

encodes for a specific N-terminal sequence which is also

common to all known human and rodent VEGF splice

variants. Exon 3 is also contained within all known VEGF

splice variants and encodes for VEGF dimerization sites

and for binding to VEGFR-1 (86-88). Exon 4 encodes for

binding of VEGF alternative splice variants to VEGFR-2

(89,90). Exon 5 encodes for a plasmin cleavage site, which

allows production of smaller VEGF peptide fragments

through proteolytic cleavage (114,115). Exon 6 encodes for

binding of VEGF splice variants to heparin and heparan

sulfate proteoglycans on the surface of cells (116). Exon 6

is either not present, as occurs with murine VEGF120 and

VEGF164 (Figure 2B), and human VEGF165b (Figure 2A),

or is significantly altered by loss of part of exon 6 designated

Page 9: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

as exon 6b, which occurs in murine VEGF188 and VEGF144

(Figure 2B). Exon 6 is also altered by extension of 6a,

designated as exon 6’, which occurs in VEGF205* (Figure

2B). Exon 7 encodes for an additional heparin- binding

domain (116) and it is also lost in a number of VEGF splice

variants including murine VEGF120, VEGF144 and

VEGF205*. The presence or absence of exons 6 and 7

determines the ability of the specific VEGF splice variants

to either freely diffuse within the tumor microenvironment

or to bind heparin and heparan sulfate proteoglycans,

therefore remaining sequestered and acting as slow release

molecules. For example, VEGF121 lacks the charged basic

amino acids encoded by exons 6 and 7 and, therefore, is a

soluble growth factor that does not bind heparin (114)

(Figure 2A). In contrast, VEGF189 contains exons 6 and 7,

and has a high affinity for binding heparin, resulting in

sequestration of this splice variant by the extracellular

matrix and heparan sulfate proteoglycans on the cell surface

(115). Other VEGF splice variants that contain either exon

6 or exon 7 but not both possess only one of the two

heparin-binding domains (Figure 2B). VEGF splice variants

with this exon configuration, such as VEGF164, can both

freely diffuse in the area surrounding the tumor as well as

remain bound to the cell surface and extracellular matrix.

The number of VEGF splice variants which have deletions,

insertions, or alterations in either exon 6 and/or exon 7

suggests that production of VEGF splice variants that

differentially bind heparin or freely diffuse is tightly

regulated during angiogenesis. For example, the exon

structure of the unique murine VEGF205* splice variant

predicts that it is actively secreted based on the presence of

exon 1, that it can dimerize and bind both VEGFR-1 and

VEGFR-2 based on the presence of exons 3 and 4.

Additionally, VEGF205* is predicted to bind heparin and

heparan sulfate proteoglycans based on the presence of

exon 6, resulting in its retention within the local tumor

microenvironment. Since VEGF205* contains an extension

of exon 6a, designated as 6’, that encodes for a protein with

a novel carboxyl-terminus tail, this VEGF splice variant is

expected to have activities that differ from other VEGF

splice variants. We have defined one such activity of

VEGF205*, which is stimulation of survival pathways of

vascular endothelial cells.

VEGF-induced Signal Transduction Pathways

VEGF splice variants act as survival factors through the

inositidylphosphate-3-kinase (IP3-K)/Akt signal transduction

pathway (57,58,117). Akt (also known as Protein kinase B) is

a serine/threonine kinase with homology to both protein

kinase C (PKC) and cyclic-AMP-dependent protein kinase A

(PKA) (118-120). As shown in Figure 3, Akt is a core

component of the IP3-K signaling pathway (121,122) and is

composed of an amino-terminal pleckstrin homology (PH)

domain that binds phospholipids (123), a kinase domain, and

a proline-rich carboxyl-terminal regulatory domain. In

response to growth factors including VEGF splice variants,

IP3-K is recruited to the inner surface of the plasma

membrane resulting in the generation of the membrane-

bound lipid phosphatidylinositol 3,4,5-triphosphate (PIP3)

(57,124,125) (Figure 3). Following binding of PIP3 to the PH

domain, Akt is translocated from the cytoplasm to the inner

surface of the plasma membrane (126). This results in

conformational changes rendering Akt accessible to

phosphorylation at two amino acid residues, threonine-308

(Thr-308), which lies within the Akt kinase domain and

serine-473 (Ser-473), which is located within the Akt

regulatory domain (127). Simultaneous phosphorylation of

Akt at Thr-308 and Ser-473 (128) is carried out by two

distinct kinases: 3-phosphoinositide-dependent protein

kinase-1 (PDK-1), which phosphorylates Thr-308 (129) and

integrin-linked kinase-1 (ILK-1), which phosphorylates Ser-

473 (130-132) (Figure 3).

The majority of studies that have examined VEGF as a

survival factor have used human VEGF165 splice variant

(57,117). Activation of IP3-K/Akt by VEGF165 occurs

through its selective association with VEGFR-2 (133,134).

VEGF activates Akt, resulting in protection of cells against

apoptosis by inducing the expression of anti-apoptotic

proteins, including FLICE-inhibitory protein (FLIP), an

inhibitor of FLICE/caspase 8. Inhibition of FLICE protects

vascular endothelial cells from apoptosis induced by Fas-

mediated caspase 8 activation (135). VEGF also stimulates

phosphorylation and subsequent inactivation of pro-

apoptotic proteins within the family of Forkhead

transcription factors, which occurs through an IP3-K/Akt-

dependent pathway (Figure 3) (136).

Although the majority of studies to date have focused on

the role of Akt as a survival factor, Akt can also act as a

multi-functional protein kinase which regulates both cell

proliferation and increases in cell mass (Figure 3) (137). Akt

phosphorylates and consequently blocks the activity of

glycogen synthase kinase-3 beta (GSK-3‚), thereby

preventing degradation of cyclin D1, which is a protein

required for progression of cells from the G1-phase of the

cell cycle into the S-phase (Figure 3) (138). In addition, Akt

regulates cell proliferation by phosphorylating the cell cycle

inhibitory protein, p21Cip1/WAF1, which inhibits interactions

between p21Cip1/WAF1 and proliferating cell nuclear antigen

(PCNA), thereby reversing repression of DNA replication

(139). In addition to its ability to regulate cell proliferation,

Akt can control cell mass by directly activating the

mammalian target of rapamycin (mTOR), also known as

FK506-binding protein-rapamycin-associated protein,

FRAP1 (137,140,141). mTOR was identified through

characterization of the activity of Rapamycin (Sirolimus;

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

533

Page 10: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

Rapamune), a macrolide antibiotic isolated from a strain of

Streptomyces hygroscopicus contained in a soil sample

collected on Easter Island, Rapa Nui. mTOR phosphorylates

and activates ribosomal protein S6 kinase (S6K), which

stimulates translation of mRNAs that regulate cell mass and

regulate the cell-cycle such as cyclin D1 (142,143). In

addition, activated Akt can directly phosphorylate S6 kinase.

Taken together, these studies demonstrate that VEGF can

induce signal transduction pathways associated with survival,

proliferation and expansion in cell mass (144) (Figure 3).

Our laboratory has found that the IP3-K/Akt pathway is

activated by specific VEGF splice variants, suggesting that

these variants can selectively block apoptosis, allowing

proliferation of cells involved in tumor-associated

angiogenesis, which leads to increased tumor mass. As

shown in Figure 4, the novel murine VEGF splice variant

VEGF205* (50 ng/ml), but not VEGF120, led to a 3.5-fold

increase (p<0.02) in Akt activation in human vascular

endothelial cells. This response was measured by the

quantitation of phosphorylation levels of Akt at Serine-473

residue by means of a phospho-specific antibody, which was

inhibited by the specific IP3-K inhibitor, LY294002 (145).

These results demonstrate that recombinant VEGF205*

selectively stimulates the IP3-K signal transduction pathway

in vascular endothelial cells. Activation of the IP3-K/Akt

pathway by distinct VEGF splice variants may lead to

multiple biological effects which are dependent upon the

cell types that respond to VEGF through expression of

specific receptors, as well as on the stage of tumor-

associated angiogenesis during which specific VEGF splice

variants activate Akt.

Targeting Tumor-associated Angiogenesis UsingVEGF-directed Strategies

The recognition that tumors undergoing active expansion

are dependent upon tumor-associated angiogenesis has

brought significant advances in anti-tumor therapy. Instead

of using conventional chemotherapeutics, which target all

cells undergoing active proliferation, including bone marrow

and the gastrointestinal tract, approaches that selectively

target tumor vessels are now being developed. Tumors are

genetically unstable and accumulate mutations as they

progress, invade and metastasize. In contrast, vascular

endothelial cells and pericytes within the stromal

compartment are genetically stable. Since agents that block

tumor-associated angiogenesis are primarily targeted at cell

populations within the stromal compartment, they bypass

the development of drug resistance by tumor cells that

commonly occurs with administration of conventional

chemotherapeutic drugs (146,147). Furthermore,

endothelial cells lining the inner surface of tumor vessels are

readily accessible to systemically delivered anti-angiogenic

drugs, which is in contrast to standard anti-tumor

chemotherapies given intravenously that do not reach all of

the tumor cells within a large tumor mass (148). The

approaches that are now being explored to block tumor-

associated angiogenesis are relatively new and, although

some show promise, approval for widespread common

clinical use has not yet been obtained.

Since both tumor cells and vascular endothelial cells

respond to VEGF, the first approach to block tumor growth

would be to simultaneously target both of these cell types

(Figure 5A). One such approach under clinical evaluation,

which shows promise as an adjunct to conventional

chemotherapeutic regimens, is recombinant humanized

monoclonal anti-VEGF165 antibody (rhuMAb VEGF),

Avastinì (Bevacizumab; Genentech, San Francisco, CA,

USA) (102). Clinical trials are currently ongoing using

Avastinì in patients with metastatic colorectal carcinoma

in combination with a conventional chemotherapy regimen

of 5-Fluorouracil/Leucovorin (149,150). There are also

other clinical trials now ongoing including phase III clinical

trials using Avastinì in combination with cytotoxic

chemotherapies for treatment of metastatic breast cancer,

lung cancer and renal cell cancer, and phase II clinical trials

using Avastinì as an adjunct to conventional treatment

regimens in a variety of other malignancies (149,151,152).

Widespread use of Avastinì awaits approval by the U.S.

Food and Drug Administration, which is predicted to occur

within 2004 (http://www.gene.com/gene/pipeline/status/

oncology/avastin.

While Avastinì was initially developed to target tumor-

associated vascular endothelial cells, the successful results

of clinical trials using this treatment approach as well as our

continued understanding of the role of pericytes in tumor-

associated angiogenesis suggest that Avastinì may inhibit

pericyte functions during this process (Figure 5B), and may

also inhibit VEGF-induced activation of survival pathways

in tumor cells (Figure 5A). Additional targeting strategies

currently being developed include the use of small

interfering RNA (SiRNA) (153,154) and anti-sense therapy

(155,156) to knock-down VEGF gene expression, which

suppresses VEGF synthesis and reduces tumor growth.

Since both tumor cells and vascular endothelial cells

express VEGFR-2, development of chimeric receptors

consisting of the extracellular VEGF-binding domain of

VEGFR-2 combined with the intracellular domain of a

death-triggering molecule would reverse VEGF-induced

survival of both of these cell populations (Figure 5A). For

example, VEGF-induced apoptosis of vascular endothelial

cells was triggered following transfection of these cells with

a chimeric receptor composed of the extracellular VEGF-

binding domain of VEGFR-2, and the intracellular domain

of the apoptotic receptor Fas (157). Another therapeutic

strategy is the use of nuclease-resistant ribozymes directed

in vivo 18: 525-542 (2004)

534

Page 11: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

against VEGFR-2, which degrades mRNA encoding for that

receptor. One such anti-VEGFR-2 ribozyme therapeutic is

Angiozyme (Ribozyme, Boulder, CO, USA), which is

currently being tested in phase II clinical trials (158).

An alternative anti-angiogenic strategy, that has been

defined as antiangiogenic scheduling or metrononomic

scheduling of chemotherapy, is based on administration of

cytotoxic drugs at lower doses and for longer times than is

conventionally used (159). This approach is based on the

observation that apoptosis of vascular endothelial cells

preceded apoptosis of tumor cells following standard

cytotoxic chemotherapy (160). However, during the

treatment-free interval that is required for recovery of bone

marrow when administering standard chemotherapy at the

maximum tolerated dose, vascular endothelial cells involved

in tumor-associated angiogenesis resumed rapid proliferation,

thus supporting tumor regrowth. In addition tumor cells can

acquire drug resistance during this treatment-free interval. To

overcome tumor recurrence, cytotoxic drugs such as

cyclophosphamide combined with the anti-angiogenic agent

TNP-470 were administered at lower doses and at regular

intervals. This anti-angiogenic scheduling provided maximum

and continuous exposure of tumor-associated vascular

endothelial cells to an anti-angiogenic drug and low doses of

a cytotoxic anti-tumor drug (159). This scheduling strategy

resulted in sustained tumor regression due to loss of tumor

vessels, which occurred prior to acquisition of resistance to

cytotoxic chemotherapeutic drugs by tumor cells (159-161).

Tumor vessels development occurs due to the coordinated

activity of vascular endothelial cells and pericytes. One

current strategy in anti-angiogenic therapy is to target both

of these cell types that comprise capillary sprouts formed

during tumor-associated angiogenesis (162-164) (Figure 5B).

In contrast to vascular endothelial cells in quiescent and

mature vasculature, during active tumor-associated

angiogenesis these cells secrete PDGF‚, whose receptor,

PDGFR-‚ is selectively expressed on the surface of

pericytes. In contrast, neither PDGF‚ nor PDGFR-‚ are

produced or expressed by tumor cells. Therefore, targeting

pericytes by inhibiting the PDGFR-‚ receptor tyrosine

kinase with simultaneous inhibition of the VEGFR-2

receptor tyrosine kinase expressed by both vascular

endothelial cells and pericytes would block activation of

VEGF-induced survival pathways resulting in apoptosis of

these cells. Ultimately, this strategy would result in

diminished tumor-associated angiogenesis leading to tumor

regression (Figure 5B). Use of inhibitors of PDGFR-‚ and

VEGFR-2 is a newly-described therapeutic approach that

was recently defined as a multiplex receptor-targeting

strategy (162,165). Regression of tumors was observed

following treatment with either SU6668 (Sugen/Pharmacia

& Upjohn, Bridgewater, NJ, USA), or Gleevecì (STI571/

Imatinib; Novartis Pharmaceuticals, East Hanover, NJ,

USA) (166), which inhibit the PDGFR-‚ receptor tyrosine

kinase expressed by pericytes, in combination with SU5416

(Sugen), which inhibits VEGFR-2 receptor tyrosine kinase

that is expressed by both vascular endothelial cells and

pericytes, as well as inhibits production of matrix- degrading

enzymes produced during tumor vessel formation (162,165).

This combination of inhibitors disrupted pericyte-endothelial

associations within tumors to a greater extent than either of

the single agents alone. Using a similar strategy, inhibition

of the Tie-2 receptor tyrosine kinase was observed to

significantly decrease tumor-associated angiogenesis and

diminish tumor growth (167,168). Since Tie-2 and VEGFR-

2 share homology in their ATP binding kinase domains

(169), compounds that simultaneously target homologous

regions contained within both of these receptors may hold

promise as effective anti-angiogenic therapies. For example,

nakijiquinone C-derived analogues are being developed that

simultaneously target the ATP binding kinase domains of

these two receptors (169). Taken together, these studies

clearly indicate that combinatorial strategies that take

advantage of our understanding of the complex nature of

tumor-associated angiogenesis and target the multiple cell

populations, receptors, growth factors and signal

transduction pathways involved in this process will be

significantly more effective than the use of single agents.

The last strategy to be discussed for targeting VEGF-

induced tumor-associated angiogenesis involves targeting

pathways that are shared by all three cell types involved in

tumor vessel formation (Figure 5C). Until tumor angio-

genesis is completed and the activated vascular endothelial

cells organize into functional new blood vessels, all of these

cells including tumor cells, vascular endothelial cells and

pericytes are within a hypoxic and nutritionally-deprived

milieu and are at risk of undergoing apoptosis. Since

VEGF-induced IP3-K/Akt signaling is a survival pathway

common to all of these cell types, targeting this pathway

may represent a successful therapeutic strategy to

simultaneously induce apoptosis of each of these cell types

(Figure 5C). Recent pre-clinical data support the anti-

angiogenic properties of LY294002 (145), which was

observed to inhibit survival of vascular endothelial cells by

inducing the p53 tumor suppressor gene and stimulating

expression of p21Cip1/WAF1 (170). Alternatives to LY294002

that also target the IP3-K/Akt signaling pathway include

agents that inhibit the serine/threonine kinase mTOR

(Figure 3), such as Rapamycin and its ester derivative, cell

cycle inhibitor-779 (CCI-779; Wyeth, Madison, NJ, USA).

These agents have been shown to have both anti-tumor and

anti-angiogenic activities (171,172).

The ability of tumor cells to trans-differentiate into cells

that have capabilities of vascular endothelial cells increases

the complexity of development of therapeutic strategies to

target the tumor vasculature. Recent studies demonstrated

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

535

Page 12: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

that the anti-angiogenic inhibitor Endostatin, which is a

20 kD proteolytic fragment of collagen type XVIII, failed

to inhibit tumor-derived vascular networks that arose from

vasculogenic mimicry (31). However, the identification of

IP3-K/Akt as an essential regulatory survival pathway

common to all of the cell types now recognized to be

involved in tumor-associated angiogenesis holds promise

as a means of targeting this process, regardless of the

specific cell population involved in formation of tumor

vessels (Figure 5C) (173).

While the use of agents that target Akt-associated

survival and proliferation pathways hold promise, systemic

treatment with Akt inhibitors can have severe side-effects

including cardiotoxicity (174) and toxicity to the nervous

system (175). To overcome such limitations, new drug-

delivery strategies include conjugating the IP3-

K/Akt/mTOR pathway inhibitors, LY294002 and

Rapamycin, to peptides that home to tumor vessels (Figure

5C). This drug-delivery strategy takes advantage of the fact

that tumor-associated vascular endothelial cells express

specific surface markers, defined as molecular addresses,

that are absent in mature, quiescent blood vessels (176-178).

For example, tumor-associated vascular endothelial cells

express integrins such as ·V‚3 and ·V‚5, which enable their

attachment to extracellular matrix proteins (177,179). A

technique based on in vivo phage display technology can be

used to deliver therapeutic agents to tumor-associated blood

vessels (Figure 5C). This uses small synthetic peptides

displayed within the protein coat of M13 filamentous

bacteriophage, which act as homing ligands that direct anti-

angiogenic agents, cytotoxic drugs and/or pro-apoptotic

molecules to the integrins expressed only on tumor-

associated vascular endothelial cells (180). In contrast to the

multiple studies that have defined proteins present on

vascular endothelial cells during tumor vessel formation,

peptides that are differentially-expressed by tumor-

associated pericytes are only now being identified (181).

Following elucidation of the molecular addresses present on

cells involved in tumor vessel development and stabilization,

other therapeutic opportunities will be available to

differentially target tumor-associated pericytes by combining

this directed drug-delivery strategy with agents such as

inhibitors of the VEGFR-2 and PDGFR-‚ receptor tyrosine

kinases, which include SU5416, SU6668, SU11248 (182) and

Gleevecì/Imatinib (Figure 5B).

Development of improved strategies for treatment of

tumors is dependent upon our continued understanding of

the complexity of tumor-associated angiogenesis. Successful

therapeutic approaches will probably target pathways that

are common to the cell populations known to be involved

in this process, which will be used in combination with

enhanced drug-delivery methods. This has the potential to

significantly improve the efficacy of anti-tumor therapy by

targeting tumor-associated vasculature without affecting the

angiogenesis that occurs under normal conditions such as

wound healing. These approaches will also limit the toxicity

and drug resistance that are commonly observed using

conventional treatment protocols.

Conclusion

Development of new blood vessels is required for tumor

survival and expansion of the tumor mass. It is clear that we

are on the verge of defining the cell types involved in tumor-

associated angiogenesis. In addition, the growth factors and

their associated receptors expressed by specific cell types

involved in this process, that work in concert with VEGF

splice variants to precisely co-regulate the temporal

sequence of events critical to tumor-associated angiogenesis,

are only now being defined and their function elucidated.

As this field emerges, our improved understanding of the

role of VEGF proteins in tumor-associated angiogenesis will

continue to provide opportunities to enhance the selectivity

and specificity of anti-angiogenic and anti-tumor

therapeutics.

Acknowledgements

The authors acknowledge Dr. Robert M. Snapka (The Ohio State

University, Columbus, Ohio, USA) for critical reading of the

manuscript and helpful discussion.

References:

1 Risau W: Embryonic angiogenesis factors. Pharmacol Ther 51:

371-6, 1991.

2 Torry RJ and Rongish BJ: Angiogenesis in the uterus:

potential regulation and relation to tumor angiogenesis. Am J

Reprod Immunol 27: 171-9, 1992.

3 Arnold F and West DC: Angiogenesis in wound healing.

Pharmacol Ther 52: 407-22, 1991.

4 Pezzella F, Pastorino U, Tagliabue E, Andreola S, Sozzi G,

Gasparini G, Menard S, Gatter KC, Harris AL, Fox S, Buyse

M, Pilotti S, Pierotti M and Rilke F: Non-small-cell lung

carcinoma tumor growth without morphological evidence of

neo-angiogenesis. Am J Pathol 151: 1417-23, 1997.

5 Holash J, Maisonpierre PC, Compton D, Boland P, Alexander

CR, Zagzag D, Yancopoulos GD and Wiegand SJ: Vessel

cooption, regression and growth in tumors mediated by

angiopoietins and VEGF. Science 284: 1994-8, 1999.

6 Hlatky L, Hahnfeldt P and Folkman J: Clinical application of

antiangiogenic therapy: microvessel density, what it does and

doesn't tell us. J Natl Cancer Inst 94: 883-93, 2002.

7 Folkman J: Anti-angiogenesis: new concept for therapy of

solid tumors. Ann Surg 175: 409-16, 1972.

8 Holmgren L, O'Reilly MS and Folkman J: Dormancy of

micrometastases: balanced proliferation and apoptosis in the

presence of angiogenesis suppression. Nat Med 1: 149-53, 1995.

9 Bergers G and Benjamin LE: Tumorigenesis and the angiogenic

switch. Nat Rev Cancer 3: 401-10, 2003.

in vivo 18: 525-542 (2004)

536

Page 13: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

10 Hanahan D and Folkman J: Patterns and emerging

mechanisms of the angiogenic switch during tumorigenesis.

Cell 86: 353-64, 1996.

11 Jung YD, Ahmad SA, Liu W, Reinmuth N, Parikh A, Stoeltzing

O, Fan F and Ellis LM: The role of the microenvironment and

intercellular cross-talk in tumor angiogenesis. Semin Cancer

Biol 12: 105-12, 2002.

12 Petit AM, Rak J, Hung MC, Rockwell P, Goldstein N, Fendly

B and Kerbel RS: Neutralizing antibodies against epidermal

growth factor and ErbB-2/neu receptor tyrosine kinases down-

regulate vascular endothelial growth factor production by

tumor cells in vitro and in vivo: angiogenic implications for

signal transduction therapy of solid tumors. Am J Pathol 151:

1523-30, 1997.

13 Rak J, Mitsuhashi Y, Bayko L, Filmus J, Shirasawa S, Sasazuki T

and Kerbel RS: Mutant ras oncogenes upregulate VEGF/VPF

expression: implications for induction and inhibition of tumor

angiogenesis. Cancer Res 55: 4575-80, 1995.

14 Ebos JM, Tran J, Master Z, Dumont D, Melo JV, Buchdunger

E and Kerbel RS: Imatinib mesylate (STI-571) reduces Bcr-

Abl-mediated vascular endothelial growth factor secretion in

chronic myelogenous leukemia. Mol Cancer Res 1: 89-95, 2002.

15 Arbiser JL, Moses MA, Fernandez CA, Ghiso N, Cao Y,

Klauber N, Frank D, Brownlee M, Flynn E, Parangi S, Byers

H R and Folkman J: Oncogenic H-ras stimulates tumor

angiogenesis by two distinct pathways. Proc Natl Acad Sci

USA 94: 861-6, 1997.

16 Mazure NM, Chen EY, Yeh P, Laderoute KR and Giaccia AJ:

Oncogenic transformation and hypoxia synergistically act to

modulate vascular endothelial growth factor expression.

Cancer Res 56: 3436-40, 1996.

17 Folkman J: Angiogenesis and its inhibitors. In: Important

Advances in Oncology (DeVita VT, Hellman S and Rosenberg

SA, eds.). Philadelphia, J B Lippincott, 1985, pp 42-62.

18 Distler JH, Hirth A, Kurowska-Stolarska M, Gay RE, Gay S and

Distler O: Angiogenic and angiostatic factors in the molecular

control of angiogenesis. Q J Nucl Med 47: 149-61, 2003.

19 Jain RK and Booth, MF: What brings pericytes to tumor

vessels? J Clin Invest 112: 1134-6, 2003.

20 Benjamin LE, Hemo I and Keshet E: A plasticity window for

blood vessel remodelling is defined by pericyte coverage of the

preformed endothelial network and is regulated by PDGF-B

and VEGF. Development 125: 1591-8, 1998.

21 Leek RD, Hunt NC, Landers RJ, Lewis CE, Royds A and Harris

AL: Macrophage infiltration is associated with VEGF and

EGFR expression in breast cancer. J Pathol 190: 430-6, 2000.

22 Zhang H and Issekutz AC: Growth factor regulation of

neutrophil-endothelial cell interactions. J Leukoc Biol 70: 225-

32, 2001.

23 Gruber BL, Marchese MJ and Kew R: Angiogenic factors

stimulate mast-cell migration. Blood 86: 2488-93, 1995.

24 Crowther, M, Brown, N. J, Bishop, ET and Lewis CE:

Microenvironmental influence on macrophage regulation of

angiogenesis in wounds and malignant tumors. J Leukoc Biol

70: 478-90, 2001.

25 Barbera-Guillem E, Nyhus JK, Wolford CC, Friece CR and

Sampsel JW: Vascular endothelial growth factor secretion by

tumor-infiltrating macrophages essentially supports tumor

angiogenesis and IgG immune complexes potentiate the

process. Cancer Res 62: 7042-9, 2002.

26 Coussens LM, Raymond WW, Bergers G, Laig-Webster M,

Behrendtsen O, Werb Z, Caughey GH and Hanahan D:

Inflammatory mast cells up-regulate angiogenesis during squamous

epithelial carcinogenesis. Genes Dev 13: 1382-97, 1999.

27 Pollard JW: Opinion: Tumour-educated macrophages

promote tumour progression and metastasis. Nat Rev Cancer

4: 71-8, 2004.

28 Taichman NS, Young S, Cruchley AT, Taylor P and Paleolog

E: Human neutrophils secrete vascular endothelial growth

factor. J Leukoc Biol 62: 397-400, 1997.

29 Norrby K: Mast cells and angiogenesis. Apmis 110: 355-71,

2002.

30 Annabi B, Lee YT, Turcotte S, Naud E, Desrosiers RR,

Champagne M, Eliopoulos N, Galipeau J and Beliveau R:

Hypoxia promotes murine bone-marrow-derived stromal cell

migration and tube formation. Stem Cells 21: 337-47, 2003.

31 Hendrix MJ Seftor EA, Hess AR and Seftor RE: Vasculogenic

mimicry and tumour-cell plasticity: lessons from melanoma.

Nat Rev Cancer 3: 411-21, 2003.

32 Folkman J: Proceedings: Tumor angiogenesis factor. Cancer

Res 34: 2109-13, 1974.

33 Shing Y, Folkman J, Sullivan R, Butterfield C, Murray J and

Klagsbrun M: Heparin affinity: purification of a tumor-

derived capillary endothelial cell growth factor. Science 223:

1296-9, 1984.

34 Dono R, Texido G, Dussel R, Ehmke H and Zeller R:

Impaired cerebral cortex development and blood pressure

regulation in FGF-2-deficient mice. Embo J 17: 4213-25, 1998.

35 Senger DR, Perruzzi CA, Feder J and Dvorak HF: A highly

conserved vascular permeability factor secreted by a variety of

human and rodent tumor cell lines. Cancer Res 46: 5629-32, 1986.

36 Ferrara N and Henzel WJ: Pituitary follicular cells secrete a novel

heparin-binding growth factor specific for vascular endothelial

cells. Biochem Biophys Res Commun 161: 851-8, 1989.

37 Plouet J, Schilling J and Gospodarowicz D: Isolation and

characterization of a newly identified endothelial cell mitogen

produced by AtT-20 cells. EMBO J 8: 3801-6, 1989.

38 Gospodarowicz D, Abraham JA and Schilling J: Isolation and

characterization of a vascular endothelial cell mitogen

produced by pituitary-derived folliculo stellate cells. Proc Natl

Acad Sci USA 86: 7311-5, 1989.

39 Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea

KS, Powell-Braxton L, Hillan KJ and Moore MW:

Heterozygous embryonic lethality induced by targeted

inactivation of the VEGF gene. Nature 380: 439-42, 1996.

40 Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L,

Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K,

Eberhardt C, Declercq C, Pawling J, Moons L, Collen D,

Risau W and Nagy A: Abnormal blood vessel development

and lethality in embryos lacking a single VEGF allele. Nature

380: 435-9, 1996.

41 Olofsson B, Pajusola K, Kaipainen A, von Euler G, Joukov V,

Saksela O, Orpana A, Pettersson RF, Alitalo K and Eriksson

U: Vascular endothelial growth factor B, a novel growth factor

for endothelial cells. Proc Natl Acad Sci USA 93: 2576-81, 1996.

42 Joukov V, Pajusola K, Kaipainen A, Chilov D, Lahtinen I,

Kukk E, Saksela O, Kalkkinen N and Alitalo K: A novel

vascular endothelial growth factor, VEGF-C, is a ligand for the

Flt4 (VEGFR-3) and KDR (VEGFR-2) receptor tyrosine

kinases. EMBO J 15: 1751, 1996.

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

537

Page 14: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

43 Lee J, Gray A, Yuan J, Luoh SM, Avraham H and Wood WI:

Vascular endothelial growth factor-related protein: a ligand

and specific activator of the tyrosine kinase receptor Flt4. Proc

Natl Acad Sci USA 93: 1988-92, 1996.

44 Orlandini M, Marconcini L, Ferruzzi R and Oliviero S:

Identification of a c-fos-induced gene that is related to the

platelet-derived growth factor/vascular endothelial growth

factor family. Proc Natl Acad Sci USA 93: 11675-80, 1996.

45 Ogawa S, Oku A, Sawano A, Yamaguchi S, Yazaki Y and

Shibuya M: A novel type of vascular endothelial growth factor,

VEGF-E (NZ-7 VEGF), preferentially utilizes KDR/Flk-1

receptor and carries a potent mitotic activity without heparin-

binding domain. J Biol Chem 273: 31273-82, 1998.

46 Shibuya M: Vascular endothelial growth factor receptor-2: Its

unique signaling and specific ligand, VEGF-E. Cancer Sci 94:

751-6, 2003.

47 Maglione D, Guerriero V, Viglietto G, Delli-Bovi P and

Persico MG: Isolation of a human placenta cDNA coding for

a protein related to the vascular permeability factor. Proc Natl

Acad Sci USA 88: 9267-71, 1991.

48 Ferrara N and Davis-Smyth T: The biology of vascular

endothelial growth factor. Endocr Rev 18: 4-25, 1997.

49 Ferrara N and Gerber HP: The role of vascular

endothelial growth factor in angiogenesis. Acta Haematol

106: 148-56, 2001.

50 Dvorak HF: Tumors: wounds that do not heal. Similarities

between tumor stroma generation and wound healing. N Engl

J Med 315: 1650-9, 1986.

51 Roberts WG and Palade GE: Increased microvascular

permeability and endothelial fenestration induced by vascular

endothelial growth factor. J Cell Sci 108: 2369-79, 1995.

52 Dvorak HF, Harvey VS, Estrella P, Brown LF, McDonagh J

and Dvorak AM: Fibrin containing gels induce angiogenesis.

Implications for tumor stroma generation and wound healing.

Lab Invest 57: 673-86, 1987.

53 Chung J, Bachelder RE, Lipscomb EA, Shaw LM and

Mercurio AM: Integrin (alpha 6 beta 4) regulation of eIF-4E

activity and VEGF translation: a survival mechanism for

carcinoma cells. J Cell Biol 158: 165-74, 2002.

54 Bachelder RE, Crago A, Chung J, Wendt MA, Shaw LM,

Robinson G and Mercurio AM: Vascular endothelial growth

factor is an autocrine survival factor for neuropilin-expressing

breast carcinoma cells. Cancer Res 61: 5736-40, 2001.

55 Harmey JH and Bouchier-Hayes D: Vascular endothelial

growth factor (VEGF), a survival factor for tumour cells:

implications for anti-angiogenic therapy. Bioessays 24: 280-

3, 2002.

56 Bussolati B, Deambrosis I, Russo S, Deregibus MC and

Camussi G: Altered angiogenesis and survival in human

tumor-derived endothelial cells. Faseb J 17: 1159-61, 2003.

57 Gerber HP, McMurtrey A, Kowalski J, Yan M, Keyt BA, Dixit

V and Ferrara N: Vascular endothelial growth factor regulates

endothelial cell survival through the phosphatidylinositol 3'-

kinase/Akt signal transduction pathway. Requirement for Flk-

1/KDR activation. J Biol Chem 273: 30336-43, 1998.

58 Castilla MA, Neria F, Renedo G, Pereira DS, Gonzalez-

Pacheco FR, Jimenez S, Tramon P, Deudero JJ, Alvarez

Arroyo MV, Yague S and Caramelo C: Tumor-induced

endothelial cell activation: Role of vascular endothelial growth

factor (VEGF). Am J Physiol Cell Physiol 7: 7, 2004.

59 Suzuma K, Naruse K, Suzuma I, Takahara N, Ueki K, Aiello

LP and King GL: Vascular endothelial growth factor induces

expression of connective tissue growth factor via KDR, Flt1

and phosphatidylinositol 3-kinase-akt-dependent pathways in

retinal vascular cells. J Biol Chem 275: 40725-31, 2000.

60 Matsuyama W, Hashiguchi T, Mizoguchi A, Iwami F,

Kawabata M, Arimura K and Osame M: Serum levels of

vascular endothelial growth factor dependent on the stage

progression of lung cancer. Chest 118: 948-51, 2000.

61 Ryden L, Linderholm B, Nielsen NH, Emdin S, Jonsson PE

and Landberg G: Tumor specific VEGF-A and

VEGFR2/KDR protein are co-expressed in breast cancer.

Breast Cancer Res Treat 82: 147-54, 2003.

62 Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC,

Davis S, Sato TN and Yancopoulos GD: Requisite role of

angiopoietin-1, a ligand for the TIE2 receptor, during

embryonic angiogenesis. Cell 87: 1171-80, 1996.

63 Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain

V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC and

Yancopoulos GD: Isolation of angiopoietin-1, a ligand for the

TIE2 receptor, by secretion-trap expression cloning. Cell 87:

1161-9, 1996.

64 Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand

SJ, Radziejewski C, Compton D, McClain J, Aldrich TH,

Papadopoulos N, Daly TJ, Davis S, Sato TN and Yancopoulos

GD: Angiopoietin-2, a natural antagonist for Tie2 that disrupts

in vivo angiogenesis. Science 277: 55-60, 1997.

65 Tsigkos S, Koutsilieris M and Papapetropoulos A:

Angiopoietins in angiogenesis and beyond. Expert Opin

Investig Drugs 12: 933-41, 2003.

66 Ellis LM, Ahmad S, Fan F, Liu W, Jung YD, Stoeltzing O,

Reinmuth N and Parikh AA: Angiopoietins and their role in

colon cancer angiogenesis. Oncology (Huntingt) 16: 31-5,

2002.

67 Metheny-Barlow LJ and Li LY: The enigmatic role of

angiopoietin-1 in tumor angiogenesis. Cell Res 13: 309-17, 2003.

68 Gamble JR, Drew J, Trezise L, Underwood A, Parsons M,

Kasminkas L, Rudge J, Yancopoulos G and Vadas MA:

Angiopoietin-1 is an antipermeability and anti-inflammatory

agent in vitro and targets cell junctions. Circ Res 87: 603-7, 2000.

69 Zhang L, Yang N, Park JW, Katsaros D, Fracchioli S, Cao G,

O'Brien-Jenkins A, Randall TC, Rubin SC and Coukos G:

Tumor-derived vascular endothelial growth factor up-regulates

angiopoietin-2 in host endothelium and destabilizes host

vasculature, supporting angiogenesis in ovarian cancer. Cancer

Res 63: 3403-12, 2003.

70 Shih SC, Robinson GS, Perruzzi CA, Calvo A, Desai K, Green

JE, Ali IU, Smith LE and Senger DR: Molecular profiling of

angiogenesis markers. Am J Pathol 161: 35-41, 2002.

71 Tse V, Xu L, Yung YC, Santarelli JG, Juan D, Fabel K,

Silverberg G and Harsh GT: The temporal-spatial expression

of VEGF, angiopoietins-1 and 2 and Tie-2 during tumor

angiogenesis and their functional correlation with tumor

neovascular architecture. Neurol Res 25: 729-38, 2003.

72 Larcher F, Franco M, Bolontrade M, Rodriguez-Puebla M,

Casanova L, Navarro M, Yancopoulos G, Jorcano JL and

Conti CJ: Modulation of the angiogenesis response through

Ha-ras control, placenta growth factor and angiopoietin

expression in mouse skin carcinogenesis. Mol Carcinog 37: 83-

90, 2003.

in vivo 18: 525-542 (2004)

538

Page 15: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

73 Yu Q and Stamenkovic I: Angiopoietin-2 is implicated in the

regulation of tumor angiogenesis. Am J Pathol 158: 563-70, 2001.

74 Gerhardt H and Betsholtz C: Endothelial-pericyte interactions

in angiogenesis. Cell Tissue Res 314: 15-23, 2003.

75 Darland DC, Massingham LJ, Smith SR, Piek E, Saint-Geniez

M and D'Amore PA: Pericyte production of cell-associated

VEGF is differentiation-dependent and is associated with

endothelial survival. Dev Biol 264: 275-88, 2003.

76 Witmer AN, van Blijswijk BC, van Noorden CJ, Vrensen GF

and Schlingemann RO: In vivo angiogenic phenotype of

endothelial cells and pericytes induced by vascular endothelial

growth factor-A. J Histochem Cytochem 52: 39-52, 2004.

77 Tischer E, Mitchell R, Hartman T, Silva M, Gospodarowicz

D, Fiddes JC and Abraham JA: The human gene for vascular

endothelial growth factor. Multiple protein forms are

encoded through alternative exon splicing. J Biol Chem 266:

11947-54, 1991.

78 Leung DW, Cachianes G, Kuang WJ, Goeddel DV and

Ferrara N: Vascular endothelial growth factor is a secreted

angiogenic mitogen. Science 246: 1306-9, 1989.

79 Houck KA, Ferrara N, Winer J, Cachianes G, Li B and Leung

DW: The vascular endothelial growth factor family:

identification of a fourth molecular species and characterization

of alternative splicing of RNA. Mol Endocrinol 5: 1806-14,

1991.

80 Poltorak Z, Cohen T, Sivan R, Kandelis Y, Spira G, Vlodavsky

I, Keshet E and Neufeld G: VEGF145, a secreted vascular

endothelial growth factor isoform that binds to extracellular

matrix. J Biol Chem 272: 7151-8, 1997.

81 Lei J, Jiang A and Pei D: Identification and characterization of

a new splicing variant of vascular endothelial growth factor:

VEGF183. Biochim Biophys Acta 1443: 400-6, 1998.

82 Bates DO, Cui TG, Doughty JM, Winkler M, Sugiono M,

Shields JD, Peat D, Gillatt D and Harper SJ: VEGF165b, an

inhibitory splice variant of vascular endothelial growth factor,

is down-regulated in renal cell carcinoma. Cancer Res 62:

4123-31, 2002.

83 Cui TG, Foster RR, Saleem MA, Mathieson PW, Gillatt DA,

Bates DO and Harper SJ: Differentiated human podocytes

endogenously express an inhibitory isoform of vascular

endothelial growth factor (VEGF165b) mRNA and protein.

Am J Physiol Renal Physiol 25: 25, 2003.

84 Shima DT, Kuroki M, Deutsch U, Ng YS, Adamis AP and

D'Amore PA: The mouse gene for vascular endothelial

growth factor. Genomic structure, definition of the

transcriptional unit and characterization of transcriptional

and post-transcriptional regulatory sequences. J Biol Chem

271: 3877-83, 1996.

85 Tober KL, Cannon RE, Spalding JW, Oberyszyn TM, Parrett

ML, Rackoff AI, Oberyszyn AS, Tennant RW and Robertson

FM: Comparative expression of novel vascular endothelial

growth factor/vascular permeability factor transcripts in skin,

papillomas and carcinomas of v-Ha-ras Tg.AC transgenic

mice and FVB/N mice. Biochem Biophys Res Commun 247:

644-53, 1998.

86 Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland NG

and Lemischka IR: A receptor tyrosine kinase cDNA isolated

from a population of enriched primitive hematopoietic cells

and exhibiting close genetic linkage to c-kit. Proc Natl Acad

Sci USA 88: 9026-30, 1991.

87 Quinn TP, Peters KG, De Vries C, Ferrara N and Williams

LT: Fetal liver kinase 1 is a receptor for vascular endothelial

growth factor and is selectively expressed in vascular

endothelium. Proc Natl Acad Sci USA 90: 7533-7, 1993.

88 de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N

and Williams LT: The fms-like tyrosine kinase, a receptor

for vascular endothelial growth factor. Science 255: 989-

91, 1992.

89 Terman BI, Carrion ME, Kovacs E, Rasmussen BA, Eddy

RL and Shows TB: Identification of a new endothelial cell

growth factor receptor tyrosine kinase. Oncogene 6: 1677-

83, 1991.

90 Terman BI, Dougher-Vermazen M, Carrion ME, Dimitrov D,

Armellino DC, Gospodarowicz D and Bohlen P: Identification

of the KDR tyrosine kinase as a receptor for vascular

endothelial cell growth factor. Biochem Biophys Res Commun

187: 1579-86, 1992.

91 Galland F, Karamysheva A, Pebusque MJ, Borg JP, Rottapel

R, Dubreuil P, Rosnet O and Birnbaum D: The FLT4 gene

encodes a transmembrane tyrosine kinase related to the

vascular endothelial growth factor receptor. Oncogene 8: 1233-

40, 1993.

92 Gille H, Kowalski J, Li B, LeCouter J, Moffat B, Zioncheck

TF, Pelletier N and Ferrara N: Analysis of biological effects

and signaling properties of Flt-1 (VEGFR-1) and KDR

(VEGFR-2). A reassessment using novel receptor-specific

vascular endothelial growth factor mutants. J Biol Chem 276:

3222-30, 2001.

93 Zachary I: VEGF signalling: integration and multi-tasking

in endothelial cell biology. Biochem Soc Trans 31: 1171-7,

2003.

94 Zeng H, Sanyal S and Mukhopadhyay D: Tyrosine residues

951 and 1059 of vascular endothelial growth factor receptor-2

(KDR) are essential for vascular permeability factor/vascular

endothelial growth factor-induced endothelium migration and

proliferation, respectively. J Biol Chem 276: 32714-9, 2001.

95 Zeng H, Dvorak HF and Mukhopadhyay D: Vascular

permeability factor (VPF)/vascular endothelial growth factor

(VEGF) peceptor-1 down-modulates VPF/VEGF receptor-2-

mediated endothelial cell proliferation, but not migration,

through phosphatidylinositol 3-kinase-dependent pathways. J

Biol Chem 276: 26969-79, 2001.

96 Hiratsuka S, Minowa O, Kuno J, Noda T and Shibuya M: Flt-

1 lacking the tyrosine kinase domain is sufficient for normal

development and angiogenesis in mice. Proc Natl Acad Sci

USA 95: 9349-54, 1998.

97 Yang S, Toy K, Ingle G, Zlot C, Williams PM, Fuh G, Li B,

de Vos A and Gerritsen ME: Vascular endothelial growth

factor-induced genes in human umbilical vein endothelial cells:

relative roles of KDR and Flt-1 receptors. Arterioscler

Thromb Vasc Biol 22: 1797-803, 2002.

98 Lohela M, Saaristo A, Veikkola T and Alitalo K:

Lymphangiogenic growth factors, receptors and therapies.

Thromb Haemost 90: 167-84, 2003.

99 Van Trappen PO, Steele D, Lowe DG, Baithun S, Beasley N,

Thiele W, Weich H, Krishnan J, Shepherd JH, Pepper MS,

Jackson DG, Sleeman JP and Jacobs IJ: Expression of vascular

endothelial growth factor (VEGF)-C and VEGF-D and their

receptor VEGFR-3, during different stages of cervical

carcinogenesis. J Pathol 201: 544-54, 2003.

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

539

Page 16: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

100 Buchler P, Reber HA, Ullrich A, Shiroiki M, Roth M, Buchler

MW, Lavey RS, Friess H and Hines OJ: Pancreatic cancer

growth is inhibited by blockade of VEGF-RII. Surgery 134:

772-82, 2003.

101 Becker CM, Farnebo FA, Iordanescu I, Behonick DJ, Shih

MC, Dunning P, Christofferson R, Mulligan RC, Taylor GA,

Kuo CJ and Zetter BR: Gene therapy of prostate cancer with

the soluble vascular endothelial growth factor receptor Flk1.

Cancer Biol Ther 1: 548-53, 2002.

102 Kim KJ, Li B, Houck K, Winer J and Ferrara N: The vascular

endothelial growth factor proteins: identification of

biologically relevant regions by neutralizing monoclonal

antibodies. Growth Factors 7: 53-64, 1992.

103 Soker S, Takashima S, Miao HQ, Neufeld G and Klagsbrun

M: Neuropilin-1 is expressed by endothelial and tumor cells as

an isoform-specific receptor for vascular endothelial growth

factor. Cell 92: 735-45, 1998.

104 Gluzman-Poltorak Z, Cohen T, Herzog Y and Neufeld G:

Neuropilin-2 is a receptor for the vascular endothelial growth

factor (VEGF) forms VEGF-145 and VEGF-165 [corrected]. J

Biol Chem 275: 18040-5, 2000.

105 He Z and Tessier-Lavigne M: Neuropilin is a receptor for the

axonal chemorepellent Semaphorin III. Cell 90: 739-51, 1997.

106 Chen H, Chedotal A, He Z, Goodman CS and Tessier-Lavigne

M: Neuropilin-2, a novel member of the neuropilin family, is a

high affinity receptor for the semaphorins Sema E and Sema

IV but not Sema III. Neuron 19: 547-59, 1997.

107 Rossignol M, Gagnon ML and Klagsbrun M: Genomic

organization of human neuropilin-1 and neuropilin-2 genes:

identification and distribution of splice variants and soluble

isoforms. Genomics 70: 211-22, 2000.

108 Wang L, Zeng H, Wang P, Soker S and Mukhopadhyay D:

Neuropilin-1-mediated vascular permeability factor/vascular

endothelial growth factor-dependent endothelial cell

migration. J Biol Chem 278: 48848-60, 2003.

109 Migdal M, Huppertz B, Tessler S, Comforti A, Shibuya M, Reich

R, Baumann H and Neufeld G: Neuropilin-1 is a placenta growth

factor-2 receptor. J Biol Chem 273: 22272-8, 1998.

110 Makinen T, Olofsson B, Karpanen T, Hellman U, Soker S,

Klagsbrun M, Eriksson U and Alitalo K: Differential

binding of vascular endothelial growth factor B splice and

proteolytic isoforms to neuropilin-1. J Biol Chem 274:

21217-22, 1999.

111 Soker S, Miao HQ, Nomi M, Takashima S and Klagsbrun M:

VEGF165 mediates formation of complexes containing

VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor

binding. J Cell Biochem 85: 357-68, 2002.

112 Whitaker GB, Limberg BJ and Rosenbaum JS: Vascular

endothelial growth factor receptor-2 and neuropilin-1 form a

receptor complex that is responsible for the differential

signaling potency of VEGF(165) and VEGF(121). J Biol

Chem 276: 25520-31, 2001.

113 Oh H: A novel molecular mechanism involving neuropilin-

1 for vascular endothelial growth factor-induced retinal

angiogenesis. Nippon Ganka Gakkai Zasshi 107: 651-6,

2003.

114 Houck KA, Leung DW, Rowland AM, Winer J and Ferrara N:

Dual regulation of vascular endothelial growth factor

bioavailability by genetic and proteolytic mechanisms. J Biol

Chem 267: 26031-7, 1992.

115 Park JE, Keller GA and Ferrara N: The vascular endothelial

growth factor (VEGF) isoforms: differential deposition into the

subepithelial extracellular matrix and bioactivity of extracellular

matrix-bound VEGF. Mol Biol Cell 4: 1317-26, 1993.

116 Neufeld G, Cohen T, Gitay-Goren H, Poltorak Z, Tessler S,

Sharon R, Gengrinovitch S and Levi BZ: Similarities and

differences between the vascular endothelial growth factor

(VEGF) splice variants. Cancer Metastasis Rev 15: 153-8, 1996.

117 Fujio Y and Walsh K: Akt mediates cytoprotection of

endothelial cells by vascular endothelial growth factor in an

anchorage-dependent manner. J Biol Chem 274: 16349-54, 1999.

118 Coffer PJ and Woodgett JR: Molecular cloning and

characterisation of a novel putative protein-serine kinase

related to the cAMP-dependent and protein kinase C families.

Eur J Biochem 201: 475-81, 1991.

119 Jones PF, Jakubowicz T, Pitossi FJ, Maurer F and Hemmings

BA: Molecular cloning and identification of a serine/threonine

protein kinase of the second-messenger subfamily. Proc Natl

Acad Sci USA 88: 4171-5, 1991.

120 Bellacosa A, Testa JR, Staal SP and Tsichlis PN: A retroviral

oncogene, akt, encoding a serine-threonine kinase containing

an SH2-like region. Science 254: 274-7, 1991.

121 Franke TF, Yang SI, Chan TO, Datta K, Kazlauskas A,

Morrison DK, Kaplan DR and Tsichlis PN: The protein

kinase encoded by the Akt proto-oncogene is a target of the

PDGF-activated phosphatidylinositol 3-kinase. Cell 81: 727-

36, 1995.

122 Burgering BM and Coffer PJ: Protein kinase B (c-Akt) in

phosphatidylinositol-3-OH kinase signal transduction. Nature

376: 599-602, 1995.

123 James SR, Downes CP, Gigg R, Grove SJ, Holmes AB and

Alessi DR: Specific binding of the Akt-1 protein kinase to

phosphatidylinositol 3,4,5-trisphosphate without subsequent

activation. Biochem J 315: 709-13, 1996.

124 Guo D, Jia Q, Song HY, Warren RS and Donner DB:

Vascular endothelial cell growth factor promotes tyrosine

phosphorylation of mediators of signal transduction that

contain SH2 domains. Association with endothelial cell

proliferation. J Biol Chem 270: 6729-33, 1995.

125 Xia P, Aiello LP, Ishii H, Jiang ZY, Park DJ, Robinson GS,

Takagi H, Newsome WP, Jirousek MR and King GL:

Characterization of vascular endothelial growth factor's effect

on the activation of protein kinase C, its isoforms and

endothelial cell growth. J Clin Invest 98: 2018-26, 1996.

126 Bellacosa A, Chan TO, Ahmed NN, Datta K, Malstrom S,

Stokoe D, McCormick F, Feng J and Tsichlis P: Akt activation

by growth factors is a multiple-step process: the role of the PH

domain. Oncogene 17: 313-25, 1998.

127 Milburn CC, Deak M, Kelly SM, Price NC, Alessi DR and Van

Aalten DM: Binding of phosphatidylinositol 3,4,5-trisphosphate

to the pleckstrin homology domain of protein kinase B induces a

conformational change. Biochem J 375: 531-8, 2003.

128 Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N,

Cohen P and Hemmings BA: Mechanism of activation of protein

kinase B by insulin and IGF-1. EMBO J 15: 6541-51, 1996.

129 Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR,

Reese CB and Cohen P: Characterization of a 3-

phosphoinositide-dependent protein kinase which

phosphorylates and activates protein kinase Balpha. Curr Biol

7: 261-9, 1997.

in vivo 18: 525-542 (2004)

540

Page 17: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

130 Delcommenne M, Tan C, Gray V, Rue L, Woodgett J and

Dedhar S: Phosphoinositide-3-OH kinase-dependent regulation

of glycogen synthase kinase 3 and protein kinase B/AKT by the

integrin-linked kinase. Proc Natl Acad Sci USA 95: 11211-6,

1998.

131 Persad S, Attwell S, Gray V, Mawji N, Deng JT, Leung D,

Yan J, Sanghera J, Walsh MP and Dedhar S: Regulation of

protein kinase B/Akt-serine 473 phosphorylation by integrin-

linked kinase: critical roles for kinase activity and amino

acids arginine 211 and serine 343. J Biol Chem 276: 27462-9,

2001.

132 Troussard AA, Mawji NM, Ong C, Mui A, St -Arnaud R and

Dedhar S: Conditional knock-out of integrin-linked kinase

demonstrates an essential role in protein kinase B/Akt

activation. J Biol Chem 278: 22374-8, 2003.

133 Thakker GD, Hajjar DP, Muller WA and Rosengart TK: The

role of phosphatidylinositol 3-kinase in vascular endothelial

growth factor signaling. J Biol Chem 274: 10002-7, 1999.

134 Dayanir V, Meyer RD, Lashkari K and Rahimi N:

Identification of tyrosine residues in vascular endothelial

growth factor receptor-2/FLK-1 involved in activation of

phosphatidylinositol 3-kinase and cell proliferation. J Biol

Chem 276: 17686-92, 2001.

135 Suhara T, Mano T, Oliveira BE and Walsh K:

Phosphatidylinositol 3-kinase/Akt signaling controls endothelial

cell sensitivity to Fas-mediated apoptosis via regulation of

FLICE-inhibitory protein (FLIP). Circ Res 89: 13-9, 2001.

136 Abid MR, Guo S, Minami T, Spokes KC, Ueki K, Skurk C,

Walsh K and Aird WC: Vascular endothelial growth factor

activates PI3K/Akt/forkhead signaling in endothelial cells.

Arterioscler Thromb Vasc Biol 1: 1, 2003.

137 Alvarez B, Garrido E, Garcia-Sanz JA and Carrera AC:

Phosphoinositide 3-kinase activation regulates cell division

time by coordinated control of cell mass and cell cycle

progression rate. J Biol Chem 278: 26466-73, 2003.

138 Diehl JA, Cheng M, Roussel MF and Sherr CJ: Glycogen

synthase kinase-3beta regulates cyclin D1 proteolysis and

subcellular localization. Genes Dev 12: 3499-511, 1998.

139 Rossig L, Jadidi AS, Urbich C, Badorff C, Zeiher AM and

Dimmeler S: Akt-dependent phosphorylation of p21(Cip1)

regulates PCNA binding and proliferation of endothelial cells.

Mol Cell Biol 21: 5644-57, 2001.

140 Nave BT, Ouwens M, Withers DJ, Alessi DR and Shepherd

PR: Mammalian target of rapamycin is a direct target for

protein kinase B: identification of a convergence point for

opposing effects of insulin and amino-acid deficiency on

protein translation. Biochem J 344 Pt 2: 427-31, 1999.

141 Aoki M, Blazek E and Vogt PK: A role of the kinase mTOR

in cellular transformation induced by the oncoproteins P3k

and Akt. Proc Natl Acad Sci USA 98: 136-41, 2001.

142 Sawyers CL: Will mTOR inhibitors make it as cancer drugs?

Cancer Cell 4: 343-8, 2003.

143 Grewe M, Gansauge F, Schmid RM, Adler G and Seufferlein

T: Regulation of cell growth and cyclin D1 expression by the

constitutively active FRAP-p70s6K pathway in human

pancreatic cancer cells. Cancer Res 59: 3581-7, 1999.

144 Yu Y and Sato JD: MAP kinases, phosphatidylinositol 3-

kinase and p70 S6 kinase mediate the mitogenic response of

human endothelial cells to vascular endothelial growth factor.

J Cell Physiol 178: 235-46, 1999.

145 Vlahos CJ, Matter WF, Hui KY and Brown RF: A specific

inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-

8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem

269: 5241-8, 1994.

146 Kerbel RS: Inhibition of tumor angiogenesis as a strategy to

circumvent acquired resistance to anti-cancer therapeutic

agents. Bioessays 13: 31-6, 1991.

147 Boehm T, Folkman J, Browder T and O'Reilly MS: Anti-

angiogenic therapy of experimental cancer does not induce

acquired drug resistance. Nature 390: 404-7, 1997.

148 Jain RK: Barriers to drug delivery in solid tumors. Sci Am 271:

58-65, 1994.

149 Fernando NH and Hurwitz HI: Inhibition of vascular

endothelial growth factor in the treatment of colorectal cancer.

Semin Oncol 30: 39-50, 2003.

150 McCarthy M: Antiangiogenesis drug promising for metastatic

colorectal cancer. Lancet 361: 1959, 2003.

151 Yang JC, Haworth L, Sherry RM, Hwu P, Schwartzentruber DJ,

Topalian SL, Steinberg SM, Chen HX and Rosenberg SA: A

randomized trial of bevacizumab, an anti-vascular endothelial

growth factor antibody, for metastatic renal cancer. N Engl J Med

349: 427-34, 2003.

152 Cobleigh MA, Langmuir VK, Sledge GW, Miller KD, Haney L,

Novotny WF, Reimann JD and Vassel A: A phase I/II dose-

escalation trial of bevacizumab in previously treated metastatic

breast cancer. Semin Oncol 30: 117-24, 2003.

153 Filleur S, Courtin A, Ait-Si-Ali S, Guglielmi J, Merle C,

Harel-Bellan A, Clezardin P and Cabon F: SiRNA-mediated

inhibition of vascular endothelial growth factor severely limits

tumor resistance to antiangiogenic thrombospondin-1 and

slows tumor vascularization and growth. Cancer Res 63: 3919-

22, 2003.

154 Yin JQ, Gao J, Shao R, Tian WN, Wang J and Wan Y:

SiRNA agents inhibit oncogene expression and attenuate

human tumor cell growth. J Exp Ther Oncol 3: 194-204, 2003.

155 Riedel F, Gotte K, Li M, Hormann K and Grandis JR:

Abrogation of VEGF expression in human head and neck

squamous cell carcinoma decreases angiogenic activity in vitroand in vivo. Int J Oncol 23: 577-83, 2003.

156 Im SA, Kim JS, Gomez-Manzano C, Fueyo J, Liu TJ, Cho MS,

Seong CM, Lee SN, Hong YK and Yung WK: Inhibition of

breast cancer growth in vivo by antiangiogenesis gene therapy

with adenovirus-mediated antisense-VEGF. Br J Cancer 84:

1252-7, 2001.

157 Quinn TP, Soifer SJ, Ramer K, Williams LT and Nakamura

MC: A receptor for vascular endothelial growth factor that

stimulates endothelial apoptosis. Cancer Res 61: 8629-37,

2001.

158 Weng DE and Usman N: Angiozyme: a novel angiogenesis

inhibitor. Curr Oncol Rep 3: 141-6, 2001.

159 Browder T, Butterfield CE, Kraling BM, Shi B, Marshall B,

O'Reilly MS and Folkman J: Antiangiogenic scheduling of

chemotherapy improves efficacy against experimental drug-

resistant cancer. Cancer Res 60: 1878-86, 2000.

160 Folkman J: Angiogenesis and apoptosis. Semin Cancer Biol

13: 159-67, 2003.

161 Bergers G and Hanahan D: Combining antiangiogenic agents

with metronomic chemotherapy enhances efficacy against late-

stage pancreatic islet carcinomas in mice. Cold Spring Harb

Symp Quant Biol 67: 293-300, 2002.

Affara and Robertson: VEGF-induced Survival in Tumor Angiogenesis

541

Page 18: Vascular Endothelial Growth Factor as a Survival Factor in Tumor

162 Bergers G, Song S, Meyer-Morse N, Bergsland E and

Hanahan D: Benefits of targeting both pericytes and

endothelial cells in the tumor vasculature with kinase

inhibitors. J Clin Invest 111: 1287-95, 2003.

163 Saharinen P and Alitalo K: Double target for tumor mass

destruction. J Clin Invest 111: 1277-80, 2003.

164 Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH

and Augustin, H. G: Heterogeneity of angiogenesis and blood

vessel maturation in human tumors: implications for

antiangiogenic tumor therapies. Cancer Res 60: 1388-93, 2000.

165 Erber R, Thurnher A, Katsen AD, Groth G, Kerger H,

Hammes HP, Menger MD, Ullrich A and Vajkoczy P:

Combined inhibition of VEGF- and PDGF-signaling enforces

tumor vessel regression by interfering with pericyte-mediated

endothelial cell survival mechanisms. FASEB J 4: 4, 2003.

166 Buchdunger E, Cioffi, CL, Law N, Stover D, Ohno-Jones S,

Druker BJ and Lydon NB: Abl protein-tyrosine kinase

inhibitor STI571 inhibits in vitro signal transduction mediated

by c-kit and platelet-derived growth factor receptors. J

Pharmacol Exp Ther 295: 139-45, 2000.

167 Siemeister G, Schirner M, Weindel K, Reusch P, Menrad A,

Marme D and Martiny-Baron G: Two independent

mechanisms essential for tumor angiogenesis: inhibition of

human melanoma xenograft growth by interfering with either

the vascular endothelial growth factor receptor pathway or the

Tie-2 pathway. Cancer Res 59: 3185-91, 1999.

168 Lin P, Buxton JA, Acheson A, Radziejewski C, Maisonpierre

PC, Yancopoulos GD, Channon KM, Hale LP, Dewhirst MW,

George SE and Peters KG: Antiangiogenic gene therapy

targeting the endothelium-specific receptor tyrosine kinase Tie2.

Proc Natl Acad Sci USA 95: 8829-34, 1998.

169 Kissau L, Stahl P, Mazitschek R, Giannis A and Waldmann H:

Development of natural product-derived receptor tyrosine

kinase inhibitors based on conservation of protein domain

fold. J Med Chem 46: 2917-31, 2003.

170 Su JD, Mayo LD, Donner DB and Durden DL: PTEN and

phosphatidylinositol 3'-kinase inhibitors up-regulate p53 and

block tumor-induced angiogenesis: evidence for an effect on

the tumor and endothelial compartment. Cancer Res 63:

3585-92, 2003.

171 Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel

S, Hornung M, Bruns CJ, Zuelke C, Farkas S, Anthuber M,

Jauch KW and Geissler EK: Rapamycin inhibits primary and

metastatic tumor growth by antiangiogenesis: involvement of

vascular endothelial growth factor. Nat Med 8: 128-35, 2002.

172 Sternberg CN and Vogelzang NJ: Gemcitabine, paclitaxel,

pemetrexed and other newer agents in urothelial and kidney

cancers. Crit Rev Oncol Hematol 46 Suppl: S105-15, 2003.

173 Hess AR, Seftor EA, Seftor RE and Hendrix MJ:

Phosphoinositide 3-kinase regulates membrane Type 1-matrix

metalloproteinase (MMP) and MMP-2 activity during melanoma

cell vasculogenic mimicry. Cancer Res 63: 4757-62, 2003.

174 Matsui T, Nagoshi T and Rosenzweig A: Akt and PI 3-kinase

signaling in cardiomyocyte hypertrophy and survival. Cell

Cycle 2: 220-3, 2003.

175 Yuan J and Yankner BA: Apoptosis in the nervous system.

Nature 407: 802-9, 2000.

176 Hoffman JA, Giraudo E, Singh M, Zhang L, Inoue M, Porkka

K, Hanahan D and Ruoslahti E: Progressive vascular changes

in a transgenic mouse model of squamous cell carcinoma.

Cancer Cell 4: 383-91, 2003.

177 Arap W, Pasqualini R and Ruoslahti E: Cancer treatment by

targeted drug delivery to tumor vasculature in a mouse model.

Science 279: 377-80, 1998.

178 Burg MA, Pasqualini R, Arap W, Ruoslahti E and Stallcup

WB: NG2 proteoglycan-binding peptides target tumor

neovasculature. Cancer Res 59: 2869-74, 1999.

179 Koivunen E, Wang B and Ruoslahti E: Isolation of a highly

specific ligand for the alpha 5 beta 1 integrin from a phage

display library. J Cell Biol 124: 373-80, 1994.

180 Kolonin M, Pasqualini R and Arap W: Molecular addresses in

blood vessels as targets for therapy. Curr Opin Chem Biol 5:

308-13, 2001.

181 Joyce JA, Laakkonen P, Bernasconi M, Bergers G, Ruoslahti

E and Hanahan D: Stage-specific vascular markers revealed by

phage display in a mouse model of pancreatic islet

tumorigenesis. Cancer Cell 4: 393-403, 2003.

182 Sun L, Liang C, Shirazian S, Zhou Y, Miller T, Cui J, Fukuda

JY, Chu JY, Nematalla A, Wang X, Chen H, Sistla A, Luu TC,

Tang F, Wei J and Tang C: Discovery of 5-[5-fluoro-2-oxo-1,2-

dihydroindol-(3Z)-ylidenemethyl]-2,4- dimethyl-1H-pyrrole-3-

carboxylic acid (2-diethylaminoethyl) amide, a novel tyrosine

kinase inhibitor targeting vascular endothelial and platelet-

derived growth factor receptor tyrosine kinase. J Med Chem

46: 1116-9, 2003.

Received February 2, 2004Accepted May 14, 2004

in vivo 18: 525-542 (2004)

542