Top Banner
Available online at www.sciencedirect.com Vessel abnormalization: another hallmark of cancer? Molecular mechanisms and therapeutic implications Katrien De Bock 1,2 , Sandra Cauwenberghs 1,2 and Peter Carmeliet 1,2 As a result of excessive production of angiogenic molecules, tumor vessels become abnormal in structure and function. By impairing oxygen delivery, abnormal vessels fuel a vicious cycle of non-productive angiogenesis, which creates a hostile microenvironment from where tumor cells escape through leaky vessels and which renders tumors less responsive to chemoradiation. While anti-angiogenic strategies focused on inhibiting new vessel growth and destroying pre-existing vessels, clinical studies showed modest anti-tumor effects. For many solid tumors, anti-VEGF treatment offers greater clinical benefit when combined with chemotherapy. This is partly due to a normalization of the tumor vasculature, which improves cytotoxic drug delivery and efficacy and offers unprecedented opportunities for anti-cancer treatment. Here, we overview key novel molecular players that induce vessel normalization. Addresses 1 Vesalius Research Center, K.U.Leuven, Leuven, Belgium 2 Vesalius Research Center, VIB, Leuven, Belgium Corresponding author: Carmeliet, Peter ([email protected]) Current Opinion in Genetics & Development 2011, 21:73–79 This review comes from a themed issue on Genetic and cellular mechanisms of oncogenesis Edited by Chris Marshall and Karen Vousden Available online 22nd November 2010 0959-437X/$ – see front matter # 2010 Elsevier Ltd. All rights reserved. DOI 10.1016/j.gde.2010.10.008 Introduction Angiogenesis promotes tumor growth and malignancy. In contrast to the healthy vasculature, tumor vessels are, however, highly abnormal structurally and functionally [13]. This is the result of an uncontrolled, relentless production of angiogenic stimulators, in excess of inhibi- tors, which tips the balance in favor of hyperactive vessel growth (Figure 1). These abnormal tumor vessels are characterized by a mal-shaped, irregular, disorganized and tortuous architecture with a highly dysfunctional and leaky endothelial cell (EC) layer [1,3]. The abnormal tumor vasculature exhibits remarkable spatiotemporal heterogeneity. In certain regions, ECs with irregular shape are stacked upon each other and obstruct blood flow by extending multiple protrusions, while in other sites, ECs move away or die and leave behind gaps. Also, they are often loosely connected, have wider junctions and are covered by fewer and abnormal mural pericytes (PCs) [1,2,4 ]. These changes not only impair drug delivery and perfusion, but also convert the tumor into a hostile hypoxic and acidic milieu, from where cancer cells escape through leaky vessels [5,6]. Such an unnatural milieu also promotes a vicious cycle of non-productive angiogenesis and stimulates pro-malignant reprogram- ming of tumor cell metabolism (Figure 1). In addition, it hampers the anti-tumor immune defense and highjacks inflammatory cells for angiogenesis, enhances tumor tis- sue swelling (potentially life-threatening in brain tumors) and makes chemoradiotherapy less efficient [1]. Paradoxi- cally thus, even though tumors crave for oxygen, they stimulate a non-productive process of angiogenesis extre- mely, so that abnormal tumor vessels deliver less — rather than more — oxygen to the hypoxic cancer cells, which in turn continues to fuel the cycle. Thus, tumor vessel abnormalization promotes tumor invasiveness, dissemi- nation and overall malignancy. Current anti-angiogenic therapies are based on the con- cept of starving and depriving the tumor from its nutrient supply by destroying existing vessels and preventing new vessel growth (anti-angiogenic ‘vessel pruning’) [7,8]. However, despite successes, clinical trials with VEGF- targeted monotherapy have shown a more modest pro- longation of progression-free or overall survival of cancer patients than anticipated [8,9]. Aside from the benefit of anti-VEGF monotherapy in glioblastoma and renal cell carcinoma, other solid cancers (breast, lung and color- ectal) showed a greater therapeutic effect when anti- VEGF was combined with conventional chemotherapy. Recent findings have resolved the paradox of how an anti- angiogenic vessel pruning strategy (which would be expected to impede vascular supply of cytotoxic drugs) can, in fact, improve chemotherapy by partially normal- izing the tumor vasculature and thereby enhancing drug delivery [1]. This has not only fostered the novel concept that judicious use of vessel pruning agents may induce vessel normalization by restoring the angiogenic balance, but also raised the question whether more selective anti- angiogenic ‘vessel normalization’ strategies could be developed. In this review, we will briefly illustrate some examples of both strategies. Targeting the VEGF-family for vessel stabilization VEGF stimulates EC migration, proliferation, survival, permeability and lumen formation [10] and is indispensa- ble for physiological angiogenesis [11]. Given its import- ance in cancer and numerous other angiogenic disorders, www.sciencedirect.com Current Opinion in Genetics & Development 2011, 21:7379
7

Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

May 16, 2023

Download

Documents

Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

Available online at www.sciencedirect.com

Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implicationsKatrien De Bock1,2, Sandra Cauwenberghs1,2 and Peter Carmeliet1,2

As a result of excessive production of angiogenic molecules,

tumor vessels become abnormal in structure and function. By

impairing oxygen delivery, abnormal vessels fuel a vicious

cycle of non-productive angiogenesis, which creates a hostile

microenvironment from where tumor cells escape through

leaky vessels and which renders tumors less responsive to

chemoradiation. While anti-angiogenic strategies focused on

inhibiting new vessel growth and destroying pre-existing

vessels, clinical studies showed modest anti-tumor effects. For

many solid tumors, anti-VEGF treatment offers greater clinical

benefit when combined with chemotherapy. This is partly due

to a normalization of the tumor vasculature, which improves

cytotoxic drug delivery and efficacy and offers unprecedented

opportunities for anti-cancer treatment. Here, we overview key

novel molecular players that induce vessel normalization.

Addresses1 Vesalius Research Center, K.U.Leuven, Leuven, Belgium2 Vesalius Research Center, VIB, Leuven, Belgium

Corresponding author: Carmeliet, Peter

([email protected])

Current Opinion in Genetics & Development 2011, 21:73–79

This review comes from a themed issue on

Genetic and cellular mechanisms of oncogenesis

Edited by Chris Marshall and Karen Vousden

Available online 22nd November 2010

0959-437X/$ – see front matter

# 2010 Elsevier Ltd. All rights reserved.

DOI 10.1016/j.gde.2010.10.008

IntroductionAngiogenesis promotes tumor growth and malignancy. In

contrast to the healthy vasculature, tumor vessels are,

however, highly abnormal structurally and functionally

[1–3]. This is the result of an uncontrolled, relentless

production of angiogenic stimulators, in excess of inhibi-

tors, which tips the balance in favor of hyperactive vessel

growth (Figure 1). These abnormal tumor vessels are

characterized by a mal-shaped, irregular, disorganized

and tortuous architecture with a highly dysfunctional

and leaky endothelial cell (EC) layer [1,3]. The abnormal

tumor vasculature exhibits remarkable spatiotemporal

heterogeneity. In certain regions, ECs with irregular

shape are stacked upon each other and obstruct blood

flow by extending multiple protrusions, while in other

sites, ECs move away or die and leave behind gaps. Also,

they are often loosely connected, have wider junctions

www.sciencedirect.com

and are covered by fewer and abnormal mural pericytes

(PCs) [1,2,4��]. These changes not only impair drug

delivery and perfusion, but also convert the tumor into

a hostile hypoxic and acidic milieu, from where cancer

cells escape through leaky vessels [5,6]. Such an unnatural

milieu also promotes a vicious cycle of non-productive

angiogenesis and stimulates pro-malignant reprogram-

ming of tumor cell metabolism (Figure 1). In addition,

it hampers the anti-tumor immune defense and highjacks

inflammatory cells for angiogenesis, enhances tumor tis-

sue swelling (potentially life-threatening in brain tumors)

and makes chemoradiotherapy less efficient [1]. Paradoxi-

cally thus, even though tumors crave for oxygen, they

stimulate a non-productive process of angiogenesis extre-

mely, so that abnormal tumor vessels deliver less — rather

than more — oxygen to the hypoxic cancer cells, which in

turn continues to fuel the cycle. Thus, tumor vessel

abnormalization promotes tumor invasiveness, dissemi-

nation and overall malignancy.

Current anti-angiogenic therapies are based on the con-

cept of starving and depriving the tumor from its nutrient

supply by destroying existing vessels and preventing new

vessel growth (anti-angiogenic ‘vessel pruning’) [7,8].

However, despite successes, clinical trials with VEGF-

targeted monotherapy have shown a more modest pro-

longation of progression-free or overall survival of cancer

patients than anticipated [8,9]. Aside from the benefit of

anti-VEGF monotherapy in glioblastoma and renal cell

carcinoma, other solid cancers (breast, lung and color-

ectal) showed a greater therapeutic effect when anti-

VEGF was combined with conventional chemotherapy.

Recent findings have resolved the paradox of how an anti-

angiogenic vessel pruning strategy (which would be

expected to impede vascular supply of cytotoxic drugs)

can, in fact, improve chemotherapy by partially normal-

izing the tumor vasculature and thereby enhancing drug

delivery [1]. This has not only fostered the novel concept

that judicious use of vessel pruning agents may induce

vessel normalization by restoring the angiogenic balance,

but also raised the question whether more selective anti-

angiogenic ‘vessel normalization’ strategies could be

developed. In this review, we will briefly illustrate some

examples of both strategies.

Targeting the VEGF-family for vesselstabilizationVEGF stimulates EC migration, proliferation, survival,

permeability and lumen formation [10] and is indispensa-

ble for physiological angiogenesis [11]. Given its import-

ance in cancer and numerous other angiogenic disorders,

Current Opinion in Genetics & Development 2011, 21:73–79

Page 2: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

74 Genetic and cellular mechanisms of oncogenesis

Figure 1

Oncogenes - inflammation

Angiogenic stimulators

Angiogenic inhibitors

Vessel abnormalization

InvasionMetastasis

Tumor tissueswelling

Hypoxia

Acidosis

chemoradiationtherapy

+++

immuneresponse

serumdeprivation

Current Opinion in Genetics & Development

Mechanism for tumor vessel abnormalization. During tumor

development, oncogenic mutations and inflammation foster the

relentless production of angiogenic growth factors, in excess of

angiogenic inhibitors. The angiogenic dysbalance favors hyperactive

vessel growth and vessel abnormalization, which impairs tumor

perfusion, and renders it more hypoxic and acidic. This creates a

continuous self-perpetuating cycle of non-productive angiogenesis that

hampers the anti-tumor immune defense, enhances tumor tissue

swelling, and makes chemoradiation therapy less efficient. Moreover,

the leaky, abnormal vasculature promotes tumor invasiveness and

dissemination.

VEGF has become a prime target for anti-angiogenic

therapy [9,12]. Apart from its effects on pruning pre-exist-

ing vessels and inhibiting growth of new vessels, blockage

of VEGF or its signaling pathways also induces vessel

normalization [1] (Figure 2). For instance, switching off

VEGF expression in experimental tumors prunes imma-

ture vessels, while increasing PC coverage and vessel

maturation [13]. Aside from inducing similar effects,

pharmacological neutralization of VEGF also decreases

tumor vessel permeability and interstitial fluid pressure,

improves tumor perfusion and restores the hydrostatic

pressure gradient across the vascular wall, which leads to

a deeper penetration of drugs and chemotherapy [14,15].

The resulting ‘normalization window’ increases tumor

oxygenation and improves radiation therapy in most but

not all tumor studies [16,17]. This normalization window is

transient, since prolonged VEGF-inhibitor therapy ulti-

mately prunes the large majority of tumor vessels, including

the normalized ones.

Molecularly, VEGF inhibition causes vessel normaliza-

tion, in part by the upregulation of angiopoietin-1 (see

below) [17], matrix metalloproteinases (which remodel

the vascular basement membrane and surrounding

matrix) [17], and activating PDGFRb signaling (see

below) [18]. Interestingly, myeloid production of VEGF

is important, as deletion of VEGF from these inflamma-

tory cells promotes vessel normalization and maturation

Current Opinion in Genetics & Development 2011, 21:73–79

[19]. Though ablation of myeloid-cell VEGF accelerates

initial tumor progression, tumors are more sensitive to

chemotherapy [19]. Of all preclinical anti-angiogenic

vessel normalization factors identified, clinical evidence

for efficacy in cancer patients has been only provided for

anti-VEGF agents so far. For instance, VEGF blockage

prolongs the survival of glioblastoma patients and mice by

alleviating intracerebral edema through restoration of the

blood–brain barrier as part of the vessel normalization

process, even despite continuous tumor growth [20,21��].Anti-VEGF treatment also enhances tumor vessel PC

coverage while reducing interstitial fluid pressure and

induces other signs of vessel normalization in patients

with rectal cancer [22].

Placental growth factor (PlGF), another member of the

VEGF family, is redundant for vascular growth in de-

velopment and health, but contributes to the angiogenic

switch in numerous diseases by acting as a multi-tasking

pro-angiogenic cytokine [23]. Although not all agents

show efficacy [24], PlGF blockage by other antibodies

reduces tumor growth and metastasis partly via vessel

pruning [25,26�]. Since anti-PlGF is a weaker vessel-

pruning agent than anti-VEGF and improves vessel

maturation, it does not cause severe tumor hypoxia, yet

improves chemotherapy, indicating that it induces vessel

normalization [25,26�]. More recent data confirm that

PlGF blockage partially normalizes tumor vessels in

hepatocellular carcinoma (HCC) models by counteracting

tumor-induced capillarization of sinusoidal vessels and

coverage with vaso-constrictive PC-like hepatic stellate

cells, and by reducing vessel tortuosity and the formation

of hypo-perfused string vessels; overall, these changes

decrease tumor hypoxia and malignancy [25,26�].Additional proof for structural and functional tumor vessel

normalization (including improved tumor perfusion and

oxygenation) was observed in a more recent study upon

lowering PlGF in tumors [27]. Since PlGF is upregulated

in tumor-bearing mice and patients and in ocular models

of neovascularization receiving VEGF-targeted therapy

[23], and because PlGF-blockage enhances VEGF-tar-

geted therapy without causing adverse effects [25,26�], a

combination of both strategies warrants further study.

Role of PDGFs and angiopoietins in vesselnormalizationPDGFB, secreted by sprouting ECs, is a well character-

ized recruitment signal for PCs, expressing PDGF re-

ceptor-b (PDGFRb) [28]. Ablation of PDGFB or its

receptor, or removal of its retention signal (required for

PDGFB to bind to PCs) leads to abnormal vessel leakage,

dilatation and tortuosity in the embryonic and tumor

vasculature [28], while overexpression increases PC cov-

erage and maturation [29,30] (Figure 3). Blocking this

PC-recruitment signal causes, however, variable effects

on tumor progression and metastasis. Since cancer-associ-

ated fibroblasts contract in response to PDGFB, this

www.sciencedirect.com

Page 3: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

Mechanisms of vessel abnormalization in cancer De Bock, Cauwenberghs and Carmeliet 75

Figure 2

Anti-VEGF treatment normalizes the tumor vasculature. VEGF inhibition prunes the immature vasculature and stabilizes the remaining vessels by

enhancing their pericyte coverage, a process that is mediated via induction of Ang-1 and PDGFRb signaling. Also, VEGF neutralization decreases

tumor vessel permeability and improves tumor perfusion and oxygenation, which leads to a deeper penetration of drugs and enhanced

chemoradiotherapeutic efficacy.

growth factor increases the interstitial fluid pressure,

which impairs drug delivery, explaining in part why

PDGFB blockage improves chemotherapy [31]. Also,

some but not all studies show that blockage of PDGFB

renders tumor vessels more sensitive to VEGF inhibitors

[32,33]. However, these beneficial effects may be offset

by the risk of increased metastasis of tumor cells, which

intravasate more readily through PC-deficient vessels

[34]. By inducing the formation of a complex between

VEGFR2 and PDGFRb, which inhibits downstream

signaling of the latter, VEGF promotes vessel abnorma-

lization [18].

Various angiopoietin family members, which bind to their

Tie2 receptor, also regulate vessel normalization [35]

(Figure 3). Ang-1 promotes vessel normalization and

counteracts the VEGF-induced vascular permeability

by establishing more inter-endothelial junctions

[36,37], and favors tumor vessel maturation through PC

recruitment [35,38]. Part of the normalization activity of

anti-VEGF relies on upregulating Ang-1 expression [15].

www.sciencedirect.com

Ang-2, by contrast, induces opposite effects and renders

tumor vessels abnormal. Hence, loss or inhibition of Ang-

2 promotes tumor vessel PC coverage and vessel quies-

cence [35,39�,40], though its effects on tumor vasculature

are often contextual. In line with findings that Ang-2

plasma levels correlate with metastatic cancer and are

upregulated in cancer patients receiving VEGF blockers

[41], combined targeting of VEGF and Ang-2 exhibits

superior anti-cancer efficacy [42].

Role of PHD2 oxygen sensor in tumor vesselnormalizationOne of the essential functions of blood vessels is to

supply oxygen. In order to adequately sense and respond

to changing oxygen tensions, cellular oxygen sensing is

performed by the HIF-prolyl hydroxylases (PHD),

which control the stability of the hypoxia-inducible

factors (HIFs), that orchestrate the hypoxic response

[43,44]. Activation of HIF-1a in tumor cells by

oncogenes, hypoxia and inflammatory signals upregu-

lates VEGF production and thereby promotes vessel

Current Opinion in Genetics & Development 2011, 21:73–79

Page 4: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

76 Genetic and cellular mechanisms of oncogenesis

Figure 3

Pericyte Ang-1 PDGFRβ

HIF1

Ang-2 Tie2 PDGFβ

Endothelial cell

Angiopoietins in cancer

*Ang-1 decreases vessel leakage *Ang-1 promotes tightening endothelial junctions *Ang-1 enhances pericyte recruitment

*Ang-2 inhibits Ang-1 *Ang-2 reduces pericyte recruitment and vessel maturation *Ang-2 levels correlate with poor patient prognosis *Loss of Ang-2 reduces early stage tumor growth

PDGF signaling in cancer

*Ablation of PDGF(R) abnormalizes the tumor vasculature *Overexpression of PDGFB enhances tumor vessel maturation *PDGFB promotes vessel pericyte coverage *VEGF reduces PDGFR signaling

Current Opinion in Genetics & Development

Role of PDGFs and angiopoietins in cancer. By binding to its Tie2 receptor, Ang-1 promotes pericyte coverage and the establishment of tight

intercellular junctions. Ang-2, whose expression is controlled by HIF, inhibits Ang-1 and thus renders the tumor vessels abnormal. PDGFB is secreted

by endothelial cells and signals through PDGFRb to stimulate pericyte recruitment. The role of both Ang as well as PDGF(R) signaling in vessel

(ab)normalization is overviewed in the inserted boxes.

abnormalization. Endothelial PHD2, via regulation of

HIF-2a, does not affect physiological angiogenesis and

vessel branching, but improves tumor oxygenation by

sensing and readapting oxygen supply in conditions of

oxygen deprivation [4��]. Haplodeficiency of PHD2 does

not affect tumor vessel density and area, turtuosity or

lumen size, but induces normalization of the EC lining.

Tumor vessels of heterozygous PHD2 deficient mice are

lined by a single monolayer of regular, orderly formed,

polarized, cobblestone ECs with few fenestrations.

These changes do not affect primary tumor growth

but improve tumor perfusion and oxygenation, thereby

preventing a metastatic switch and overall prolonging

survival [4��].

Mechanistically, PHD2 haplodeficient ECs exhibit a

‘phalanx’ phenotype: they are more quiescent, and exhi-

bit lower proliferation and migration responses to VEGF

[4��]. Their molecular signature includes the upregula-

tion of soluble VEGFR1, a molecular VEGF trap, which

reduces VEGF signaling, induces vessel quiescence and

shapes vessel sprouts [45,46]. Moreover, the normalized

EC layer in heterozygous PHD2 deficient mice forms a

Current Opinion in Genetics & Development 2011, 21:73–79

tighter barrier, not only because ECs express more junc-

tional proteins (such as ZO-1 and VE-cadherin) but also

because they are surrounded by more PCs and a stable

basement membrane [4��]. VE-cadherin, a HIF-2a de-

pendent gene, induces a ‘normalized and quiescent’

endothelial layer by redirecting VEGF signaling towards

EC survival [47,48]. Consistent with the identified role of

HIF-2a downstream of PHD2, reduced HIF-2a levels

cause improper remodeling of nascent vessels into larger

conduits in the embryo [49], and the formation of an

aberrant vascular network in tumors due to impaired

remodeling [50]. In addition, EC specific deletion of

HIF-2a increases vascular leakage while reducing tumor

neovascularization [51�]. Interestingly, the Ang-1 induced

normalization of immature vessels is associated with

reduced PHD2 expression [52]. Intriguingly, the effect

of PHD2 is dose-dependent, as homozygous loss of

PHD2 increases vessel branching [53].

Does Notch contribute to the ‘phalanx cell’phenotype?Another candidate that recently emerged as a mediator of

vessel quiescence is Notch, an EC receptor for Dll4.

www.sciencedirect.com

Page 5: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

Mechanisms of vessel abnormalization in cancer De Bock, Cauwenberghs and Carmeliet 77

Since Dll4/Notch signaling controls vessel branching by

suppressing the formation of endothelial tip cells (so-

called, because they lead the branch at the forefront)

[54], inhibition of this pathway induces the formation of

more but hypoperfused vessels [55,56]. Conversely, over-

expression of Dll4 by tumor cells reduces vessel density,

while enhancing lumen size and perfusion, overall

improving tumor oxygenation [57]. However, chronic

blockage of its ligand Dll4 in healthy animals resulted

in a dose-dependent formation of vascular neoplasms

characterized by irregularly shaped blood vessels [58��].These vascular abnormalities are likely due to reduced

EC quiescence and suggest a role for Dll4/Notch in the

maintenance of the phalanx cell phenotype. Indeed,

inactivation of RBP-J, a transcription factor implicated

in Notch signaling, in ECs leads to widespread spon-

taneous angiogenesis [59], while stimulation of ECs with

Dll4 induces a quiescent phenotype that is reminiscent of

the phalanx cell signature, including a reduction of

VEGF-responses because of the downregulation of

VEGFR2 [54]. Aside from effects on ECs, Dll4 signaling

from ECs to Notch-expressing mural cells upregulates

PDGFRb expression and thereby stimulates vessel matu-

ration. In accordance, Dll4 expression in patient tumor

biopsies correlates with vessel maturation [60].

ConclusionsThe abnormal structure and function of the tumor vas-

culature help to explain many fundamental hallmarks of

cancer and challenges of anti-cancer treatment. While

anti-angiogenic vessel pruning strategies bear the risk of

rendering the tumor micro-environment further abnormal

and thereby fueling tumor invasiveness and metastasis

[61,62], anti-angiogenic vessel normalization strategies

offer the opportunity of converting a malignant invasive,

metastatic cancer into a more benign, encapsulated,

metabolically less aggressive, and poorly invasive/meta-

static tumor, which also responds better to conventional

anti-cancer chemoradiation therapy. This novel vessel

normalization concept has recently proven its applica-

bility in the clinic. Understanding the molecular basis of

vessel abnormalization not only in malignant but also in

inflammatory or ischemic diseases is therefore of great

importance. This review has highlighted some key prin-

ciples and players of this process, but other factors such as

Rgs5 [63], nitric oxide [64], and EGF receptor [64] also

participate. Discovering additional molecules with vessel

normalization activity therefore promises to offer unpre-

cedented novel opportunities to improve anti-cancer

therapy.

AcknowledgementsKDB is supported by the Fund for Scientific Research in Flanders (FWO).PC is supported by long-term structural funding (Methusalem funding bythe Flemish Government), Interuniversity attraction pole (Grant P60/30,funded by the Belgian Government, BELSPO), FWO G.0692.09 (FlemishGovernment) and a research grant by the Belgian ‘‘Foundation againstCancer’’, GAO 2006/11–K.U.Leuven.

www.sciencedirect.com

References and recommended readingPapers of particular interest, published within the period of review,have been highlighted as:

� of special interest�� of outstanding interest

1. Jain RK: Normalization of tumor vasculature: an emergingconcept in antiangiogenic therapy. Science 2005, 307:58-62.

2. Jain RK: Determinants of tumor blood flow: a review.Cancer Res 1988, 48:2641-2658.

3. De Bock K, De Smet F, Leite De Oliveira R, Anthonis K,Carmeliet P: Endothelial oxygen sensors regulate tumor vesselabnormalization by instructing phalanx endothelial cells.J Mol Med 2009, 87:561-569.

4.��

Mazzone M, Dettori D, Leite de Oliveira R, Loges S, Schmidt T,Jonckx B, Tian YM, Lanahan AA, Pollard P, Ruiz de Almodovar Cet al.: Heterozygous deficiency of PHD2 restores tumoroxygenation and inhibits metastasis via endothelialnormalization. Cell 2009, 136:839-851.

This paper for the first time describes how haplodeficiency of theendothelial PHD2 oxygen sensor redirects the cellular fate of endothelialcells towards quiescence: the phalanx cell. During tumor growth, thephalanx cell forms a tight ‘normal’ layer of cobblestone like endothelialcells thereby improving tumor oxygenation and preventing the develop-ment of metastasis.

5. Rapisarda A, Melillo G: Role of the hypoxic tumormicroenvironment in the resistance to anti-angiogenictherapies. Drug Resist Updat 2009, 12:74-80.

6. Lunt SJ, Chaudary N, Hill RP: The tumor microenvironment andmetastatic disease. Clin Exp Metastasis 2009, 26:19-34.

7. Ferrara N: Pathways mediating VEGF-independent tumorangiogenesis. Cytokine Growth Factor Rev 2010, 21:21-26.

8. Heath VL, Bicknell R: Anticancer strategies involving thevasculature. Nat Rev Clin Oncol 2009, 6:395-404.

9. Ellis LM, Hicklin DJ: VEGF-targeted therapy: mechanisms ofanti-tumour activity. Nat Rev Cancer 2008, 8:579-591.

10. Horowitz A, Simons M: Branching morphogenesis. Circ Res2008, 103:784-795.

11. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L,Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt Cet al.: Abnormal blood vessel development and lethalityin embryos lacking a single VEGF allele. Nature 1996,380:435-439.

12. Crawford Y, Ferrara N: VEGF inhibition: insights from preclinicaland clinical studies. Cell Tissue Res 2009, 335:261-269.

13. Abramovitch R, Dafni H, Smouha E, Benjamin LE, Neeman M:In vivo prediction of vascular susceptibility to vascularsusceptibility endothelial growth factor withdrawal: magneticresonance imaging of C6 rat glioma in nude mice. Cancer Res1999, 59:5012-5016.

14. Dickson PV, Hamner JB, Sims TL, Fraga CH, Ng CY,Rajasekeran S, Hagedorn NL, McCarville MB, Stewart CF,Davidoff AM: Bevacizumab-induced transient remodeling ofthe vasculature in neuroblastoma xenografts results inimproved delivery and efficacy of systemically administeredchemotherapy. Clin Cancer Res 2007, 13:3942-3950.

15. Tong RT, Boucher Y, Kozin SV, Winkler F, Hicklin DJ, Jain RK:Vascular normalization by vascular endothelial growth factorreceptor 2 blockade induces a pressure gradient across thevasculature and improves drug penetration in tumors. CancerRes 2004, 64:3731-3736.

16. Franco M, Man S, Chen L, Emmenegger U, Shaked Y, Cheung AM,Brown AS, Hicklin DJ, Foster FS, Kerbel RS: Targeted anti-vascular endothelial growth factor receptor-2 therapy leads toshort-term and long-term impairment of vascular function andincrease in tumor hypoxia. Cancer Res 2006, 66:3639-3648.

17. Winkler F, Kozin SV, Tong RT, Chae SS, Booth MF, Garkavtsev I,Xu L, Hicklin DJ, Fukumura D, di Tomaso E et al.: Kinetics of

Current Opinion in Genetics & Development 2011, 21:73–79

Page 6: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

78 Genetic and cellular mechanisms of oncogenesis

vascular normalization by VEGFR2 blockade governs braintumor response to radiation: role of oxygenation,angiopoietin-1, and matrix metalloproteinases. Cancer Cell2004, 6:553-563.

18. Greenberg JI, Shields DJ, Barillas SG, Acevedo LM, Murphy E,Huang J, Scheppke L, Stockmann C, Johnson RS, Angle N et al.:A role for VEGF as a negative regulator of pericyte function andvessel maturation. Nature 2008, 456:809-813.

19. Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N,Greenberg JI, Cheresh DA, Johnson RS: Deletion of vascularendothelial growth factor in myeloid cells acceleratestumorigenesis. Nature 2008, 456:814-818.

20. Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG,Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M et al.: AZD2171,a pan-VEGF receptor tyrosine kinase inhibitor, normalizestumor vasculature and alleviates edema in glioblastomapatients. Cancer Cell 2007, 11:83-95.

21.��

Kamoun WS, Ley CD, Farrar CT, Duyverman AM, Lahdenranta J,Lacorre DA, Batchelor TT, di Tomaso E, Duda DG, Munn LL et al.:Edema control by cediranib, a vascular endothelial growthfactor receptor-targeted kinase inhibitor, prolongs survivaldespite persistent brain tumor growth in mice. J Clin Oncol2009, 27:2542-2552.

Using intravital microscopy, the authors show that anti-VEGF alleviatesbrain edema in a mouse model of glioblastoma due to rapid normalizationof vessel function and morphology. Despite persistent growth of thetumor, this improved overall survival.

22. Willett CG, Boucher Y, di Tomaso E, Duda DG, Munn LL, Tong RT,Chung DC, Sahani DV, Kalva SP, Kozin SV et al.: Direct evidencethat the VEGF-specific antibody bevacizumab hasantivascular effects in human rectal cancer. Nat Med 2004,10:145-147.

23. Fischer C, Mazzone M, Jonckx B, Carmeliet P: FLT1 and itsligands VEGFB and PlGF: drug targets for anti-angiogenictherapy? Nat Rev Cancer 2008, 8:942-956.

24. Bais C, Wu X, Yao J, Yang S, Crawford Y, McCutcheon K, Tan C,Kolumam G, Vernes JM, Eastham-Anderson J et al.: PlGFblockade does not inhibit angiogenesis during primary tumorgrowth. Cell 2010, 141:166-177.

25. Fischer C, Jonckx B, Mazzone M, Zacchigna S, Loges S,Pattarini L, Chorianopoulos E, Liesenborghs L, Koch M, De Mol Met al.: Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistanttumors without affecting healthy vessels. Cell 2007,131:463-475.

26.�

Van de Veire S, Stalmans I, Heindryckx F, Oura H, Tijeras-Raballand A, Schmidt T, Loges S, Albrecht I, Jonckx B, Vinckier Set al.: Further pharmacological and genetic evidence for theefficacy of PlGF inhibition in cancer and eye disease. Cell 2010,141:178-190.

The authors show that anti-PlGF treatment, in a model of hepatocellularcarcinoma (HCC), does not alter capillary density, but normalizes thetumor vasculature.

27. Rolny C, Mazzone M, Tugues S, Laoui D, Johannson I, Coulon C,Squadrito ML, Segura I, Li X, Knevels E, et al.: HRG inhibits tumorgrowth and metastasis by skewing macrophage polarizationand vessel normalization through downregulation of PlGF.Cancer Cell 2010, (in press).

28. Gaengel K, Genove G, Armulik A, Betsholtz C: Endothelial–muralcell signaling in vascular development and angiogenesis.Arterioscler Thromb Vasc Biol 2009, 29:630-638.

29. Furuhashi M, Sjoblom T, Abramsson A, Ellingsen J, Micke P, Li H,Bergsten-Folestad E, Eriksson U, Heuchel R, Betsholtz C et al.:Platelet-derived growth factor production by B16 melanomacells leads to increased pericyte abundance in tumors and anassociated increase in tumor growth rate. Cancer Res 2004,64:2725-2733.

30. McCarty MF, Somcio RJ, Stoeltzing O, Wey J, Fan F, Liu W,Bucana C, Ellis LM: Overexpression of PDGF-BB decreasescolorectal and pancreatic cancer growth by increasing tumorpericyte content. J Clin Invest 2007, 117:2114-2122.

31. Hellberg C, Ostman A, Heldin CH: PDGF and vessel maturation.Recent Results Cancer Res 2010, 180:103-114.

Current Opinion in Genetics & Development 2011, 21:73–79

32. Bergers G, Song S, Meyer-Morse N, Bergsland E, Hanahan D:Benefits of targeting both pericytes and endothelial cells in thetumor vasculature with kinase inhibitors. J Clin Invest 2003,111:1287-1295.

33. Nisancioglu MH, Betsholtz C, Genove G: The absence ofpericytes does not increase the sensitivity of tumorvasculature to vascular endothelial growth factor-A blockade.Cancer Res 2010, 70:5109-5115.

34. Gerhardt H, Semb H: Pericytes: gatekeepers in tumour cellmetastasis? J Mol Med 2008, 86:135-144.

35. Augustin HG, Koh GY, Thurston G, Alitalo K: Control of vascularmorphogenesis and homeostasis through the angiopoietin–Tie system. Nat Rev Mol Cell Biol 2009, 10:165-177.

36. Thurston G, Rudge JS, Ioffe E, Zhou H, Ross L, Croll SD, Glazer N,Holash J, McDonald DM, Yancopoulos GD: Angiopoietin-1protects the adult vasculature against plasma leakage.Nat Med 2000, 6:460-463.

37. Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A,Winderlich M, Nottebaum A, Vestweber D, Deutsch U, Koh GYet al.: Angiopoietins assemble distinct Tie2 signallingcomplexes in endothelial cell–cell and cell–matrix contacts.Nat Cell Biol 2008, 10:527-537.

38. Stoeltzing O, Ahmad SA, Liu W, McCarty MF, Wey JS, Parikh AA,Fan F, Reinmuth N, Kawaguchi M, Bucana CD et al.:Angiopoietin-1 inhibits vascular permeability, angiogenesis,and growth of hepatic colon cancer tumors. Cancer Res 2003,63:3370-3377.

39.�

Falcon BL, Hashizume H, Koumoutsakos P, Chou J, Bready JV,Coxon A, Oliner JD, McDonald DM: Contrasting actions ofselective inhibitors of angiopoietin-1 and angiopoietin-2 onthe normalization of tumor blood vessels. Am J Pathol 2009,175:2159-2170.

This paper shows that selective inhibition of Ang-2 normalizes the tumorvasculature by permitting the action of Ang-1. This resulted into tighterendothelial junctions, increased pericyte coverage, and reduced vesselsprouting.

40. Nasarre P, Thomas M, Kruse K, Helfrich I, Wolter V, Deppermann C,Schadendorf D, Thurston G, Fiedler U, Augustin HG: Host-derivedangiopoietin-2 affects early stages of tumor development andvessel maturation but is dispensable for later stages of tumorgrowth. Cancer Res 2009, 69:1324-1333.

41. Helfrich I, Edler L, Sucker A, Thomas M, Christian S,Schadendorf D, Augustin HG: Angiopoietin-2 levels areassociated with disease progression in metastatic malignantmelanoma. Clin Cancer Res 2009, 15:1384-1392.

42. Koh YJ, Kim HZ, Hwang SI, Lee JE, Oh N, Jung K, Kim M, Kim KE,Kim H, Lim NK et al.: Double antiangiogenic protein, DAAP,targeting VEGF-A and angiopoietins in tumor angiogenesis,metastasis, and vascular leakage. Cancer Cell 2010,18:171-184.

43. Kaelin WG Jr, Ratcliffe PJ: Oxygen sensing by metazoans: thecentral role of the HIF hydroxylase pathway. Mol Cell 2008,30:393-402.

44. Semenza GL: Targeting HIF-1 for cancer therapy. Nat RevCancer 2003, 3:721-732.

45. Kearney JB, Kappas NC, Ellerstrom C, DiPaola FW, Bautch VL:The VEGF receptor flt-1 (VEGFR-1) is a positive modulator ofvascular sprout formation and branching morphogenesis.Blood 2004, 103:4527-4535.

46. Carmeliet P, De Smet F, Loges S, Mazzone M: Branchingmorphogenesis and antiangiogenesis candidates: tip cellslead the way. Nat Rev Clin Oncol 2009, 6:315-326.

47. Le Bras A, Lionneton F, Mattot V, Lelievre E, Caetano B, Spruyt N,Soncin F: HIF-2alpha specifically activates the VE-cadherinpromoter independently of hypoxia and in synergy with Ets-1through two essential ETS-binding sites. Oncogene 2007,26:7480-7489.

48. Dejana E, Tournier-Lasserve E, Weinstein BM: The control ofvascular integrity by endothelial cell junctions: molecularbasis and pathological implications. Dev Cell 2009, 16:209-221.

www.sciencedirect.com

Page 7: Vessel abnormalization: another hallmark of cancer?Molecular mechanisms and therapeutic implications

Mechanisms of vessel abnormalization in cancer De Bock, Cauwenberghs and Carmeliet 79

49. Duan LJ, Zhang-Benoit Y, Fong GH: Endothelium-intrinsicrequirement for Hif-2alpha during vascular development.Circulation 2005, 111:2227-2232.

50. Yamashita T, Ohneda K, Nagano M, Miyoshi C, Kaneko N, Miwa Y,Yamamoto M, Ohneda O, Fujii-Kuriyama Y: HIF-2alpha inendothelial cells regulates tumor neovascularizationthrough activation of ephrin A1. J Biol Chem 2008,283:18926-18936.

51.�

Skuli N, Liu L, Runge A, Wang T, Yuan L, Patel S, Iruela-Arispe L,Simon MC, Keith B: Endothelial deletion of hypoxia-induciblefactor-2alpha (HIF-2alpha) alters vascular function and tumorangiogenesis. Blood 2009, 114:469-477.

This paper shows the importance of HIF-2a in vascular function andtumor angiogenesis using a mouse model in which HIF-2a is selectivelydeleted in endothelial cells.

52. Chen JX, Stinnett A: Ang-1 gene therapy inhibits hypoxia-inducible factor-1alpha (HIF-1alpha)-prolyl-4-hydroxylase-2,stabilizes HIF-1alpha expression, and normalizes immaturevasculature in db/db mice. Diabetes 2008, 57:3335-3343.

53. Fraisl P, Mazzone M, Schmidt T, Carmeliet P: Regulation ofangiogenesis by oxygen and metabolism. Dev Cell 2009,16:167-179.

54. Phng LK, Gerhardt H: Angiogenesis: a team effort coordinatedby notch. Dev Cell 2009, 16:196-208.

55. Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y,Kowalski J, Watts RJ, Callahan C, Kasman I et al.: Inhibition ofDll4 signalling inhibits tumour growth by deregulatingangiogenesis. Nature 2006, 444:1083-1087.

56. Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P,Gale NW, Lin HC, Yancopoulos GD, Thurston G: Blockade of Dll4inhibits tumour growth by promoting non-productiveangiogenesis. Nature 2006, 444:1032-1037.

57. Li JL, Sainson RC, Shi W, Leek R, Harrington LS, Preusser M,Biswas S, Turley H, Heikamp E, Hainfellner JA et al.: Delta-like 4Notch ligand regulates tumor angiogenesis, improves tumor

www.sciencedirect.com

vascular function, and promotes tumor growth in vivo. CancerRes 2007, 67:11244-11253.

58.��

Yan M, Callahan CA, Beyer JC, Allamneni KP, Zhang G,Ridgway JB, Niessen K, Plowman GD: Chronic DLL4blockade induces vascular neoplasms. Nature 2010,463:E6-E7.

Using chronic inhibition of Dll4, this study provides initial in vivo evidencethat Dll4-Notch signaling contributes to endothelial cell quiescence.

59. Dou GR, Wang YC, Hu XB, Hou LH, Wang CM, Xu JF, Wang YS,Liang YM, Yao LB, Yang AG et al.: RBP-J, the transcriptionfactor downstream of Notch receptors, is essential for themaintenance of vascular homeostasis in adult mice. FASEB J2008, 22:1606-1617.

60. Patel NS, Dobbie MS, Rochester M, Steers G, Poulsom R, LeMonnier K, Cranston DW, Li JL, Harris AL: Up-regulation ofendothelial delta-like 4 expression correlates with vesselmaturation in bladder cancer. Clin Cancer Res 2006,12:4836-4844.

61. Ebos JML, Lee CR, Cruz-Munoz W, Bjarnason GA,Christensen JG, Kerbel RS: Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis.Cancer Cell 2009, 15:232-239.

62. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F,Inoue M, Bergers G, Hanahan D, Casanovas O: Antiangiogenictherapy elicits malignant progression of tumors to increasedlocal invasion and distant metastasis. Cancer Cell 2009,15:220-231.

63. Hamzah J, Jugold M, Kiessling F, Rigby P, Manzur M, Marti HH,Rabie T, Kaden S, Grone HJ, Hammerling GJ et al.: Vascularnormalization in Rgs5-deficient tumours promotes immunedestruction. Nature 2008, 453:410-414.

64. Kashiwagi S, Tsukada K, Xu L, Miyazaki J, Kozin SV, Tyrrell JA,Sessa WC, Gerweck LE, Jain RK, Fukumura D: Perivascular nitricoxide gradients normalize tumor vasculature. Nat Med 2008,14:255-257.

Current Opinion in Genetics & Development 2011, 21:73–79