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

https://doi.org/10.1016/j.gde.2010.10.008Get rights and content

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.

Introduction

Angiogenesis promotes tumor growth and malignancy. In contrast to the healthy vasculature, tumor vessels are, however, highly abnormal structurally and functionally [1, 2, 3]. This is the result of an uncontrolled, relentless production of angiogenic stimulators, in excess of inhibitors, 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 reprogramming of tumor cell metabolism (Figure 1). In addition, it hampers the anti-tumor immune defense and highjacks inflammatory cells for angiogenesis, enhances tumor tissue swelling (potentially life-threatening in brain tumors) and makes chemoradiotherapy less efficient [1]. Paradoxically thus, even though tumors crave for oxygen, they stimulate a non-productive process of angiogenesis extremely, 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, dissemination and overall malignancy.

Current anti-angiogenic therapies are based on the concept 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 prolongation 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 colorectal) 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 normalizing 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.

Section snippets

Targeting the VEGF-family for vessel stabilization

VEGF stimulates EC migration, proliferation, survival, permeability and lumen formation [10] and is indispensable for physiological angiogenesis [11]. Given its importance in cancer and numerous other angiogenic disorders, VEGF has become a prime target for anti-angiogenic therapy [9, 12]. Apart from its effects on pruning pre-existing vessels and inhibiting growth of new vessels, blockage of VEGF or its signaling pathways also induces vessel normalization [1] (Figure 2). For instance,

Role of PDGFs and angiopoietins in vessel normalization

PDGFB, secreted by sprouting ECs, is a well characterized recruitment signal for PCs, expressing PDGF receptor-β (PDGFRβ) [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 coverage and maturation [29, 30] (Figure 3). Blocking this PC-recruitment signal causes, however, variable effects on tumor

Role of PHD2 oxygen sensor in tumor vessel normalization

One 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-1α in tumor cells by oncogenes, hypoxia and inflammatory signals upregulates VEGF production and thereby promotes vessel abnormalization. Endothelial

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. 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, overexpression of Dll4 by tumor cells reduces vessel density, while enhancing lumen size and perfusion, overall

Conclusions

The abnormal structure and function of the tumor vasculature 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,

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

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

References (64)

  • A. Le Bras et al.

    HIF-2alpha specifically activates the VE-cadherin promoter independently of hypoxia and in synergy with Ets-1 through two essential ETS-binding sites

    Oncogene

    (2007)
  • L.J. Duan et al.

    Endothelium-intrinsic requirement for Hif-2alpha during vascular development

    Circulation

    (2005)
  • J.X. Chen et al.

    Ang-1 gene therapy inhibits hypoxia-inducible factor-1alpha (HIF-1alpha)-prolyl-4-hydroxylase-2, stabilizes HIF-1alpha expression, and normalizes immature vasculature in db/db mice

    Diabetes

    (2008)
  • L.K. Phng et al.

    Angiogenesis: a team effort coordinated by notch

    Dev Cell

    (2009)
  • J. Ridgway et al.

    Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis

    Nature

    (2006)
  • M. Yan et al.

    Chronic DLL4 blockade induces vascular neoplasms

    Nature

    (2010)
  • G.R. Dou et al.

    RBP-J, the transcription factor downstream of Notch receptors, is essential for the maintenance of vascular homeostasis in adult mice

    FASEB J

    (2008)
  • R.K. Jain

    Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy

    Science

    (2005)
  • R.K. Jain

    Determinants of tumor blood flow: a review

    Cancer Res

    (1988)
  • K. De Bock et al.

    Endothelial oxygen sensors regulate tumor vessel abnormalization by instructing phalanx endothelial cells

    J Mol Med

    (2009)
  • A. Rapisarda et al.

    Role of the hypoxic tumor microenvironment in the resistance to anti-angiogenic therapies

    Drug Resist Updat

    (2009)
  • N. Ferrara

    Pathways mediating VEGF-independent tumor angiogenesis

    Cytokine Growth Factor Rev

    (2010)
  • L.M. Ellis et al.

    VEGF-targeted therapy: mechanisms of anti-tumour activity

    Nat Rev Cancer

    (2008)
  • A. Horowitz et al.

    Branching morphogenesis

    Circ Res

    (2008)
  • P. Carmeliet et al.

    Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele

    Nature

    (1996)
  • Y. Crawford et al.

    VEGF inhibition: insights from preclinical and clinical studies

    Cell Tissue Res

    (2009)
  • R. Abramovitch et al.

    In vivo prediction of vascular susceptibility to vascular susceptibility endothelial growth factor withdrawal: magnetic resonance imaging of C6 rat glioma in nude mice

    Cancer Res

    (1999)
  • P.V. Dickson et al.

    Bevacizumab-induced transient remodeling of the vasculature in neuroblastoma xenografts results in improved delivery and efficacy of systemically administered chemotherapy

    Clin Cancer Res

    (2007)
  • R.T. Tong et al.

    Vascular normalization by vascular endothelial growth factor receptor 2 blockade induces a pressure gradient across the vasculature and improves drug penetration in tumors

    Cancer Res

    (2004)
  • M. Franco et al.

    Targeted anti-vascular endothelial growth factor receptor-2 therapy leads to short-term and long-term impairment of vascular function and increase in tumor hypoxia

    Cancer Res

    (2006)
  • F. Winkler et al.

    Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases

    Cancer Cell

    (2004)
  • C. Stockmann et al.

    Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis

    Nature

    (2008)
  • Cited by (168)

    View all citing articles on Scopus
    View full text