Elsevier

Seminars in Cancer Biology

Volume 52, Part 2, October 2018, Pages 107-116
Seminars in Cancer Biology

Review
The reciprocal function and regulation of tumor vessels and immune cells offers new therapeutic opportunities in cancer

https://doi.org/10.1016/j.semcancer.2018.06.002Get rights and content

Abstract

Tumor angiogenesis and escape of immunosurveillance are two cancer hallmarks that are tightly linked and reciprocally regulated by paracrine signaling cues of cell constituents from both compartments. Formation and remodeling of new blood vessels in tumors is abnormal and facilitates immune evasion. In turn, immune cells in the tumor, specifically in context with an acidic and hypoxic environment, can promote neovascularization. Immunotherapy has emerged as a major therapeutic modality in cancer but is often hampered by the low influx of activated cytotoxic T-cells. On the other hand, anti-angiogenic therapy has been shown to transiently normalize the tumor vasculature and enhance infiltration of T lymphocytes, providing a rationale for a combination of these two therapeutic approaches to sustain and improve therapeutic efficacy in cancer. In this review, we discuss how the tumor vasculature facilitates an immunosuppressive phenotype and vice versa how innate and adaptive immune cells regulate angiogenesis during tumor progression. We further highlight recent results of antiangiogenic immunotherapies in experimental models and the clinic to evaluate the concept that targeting both the tumor vessels and immune cells increases the effectiveness in cancer patients.

Introduction

The onset of tumor neovascularization is a well-established hallmark of cancer that is initiated at a certain time during tumor progression depending on the tumor type, grade and stage. Blood vessels not only have to provide oxygen and deplete waste products to oblige the demands of a growing tumor but also form niches that enable cancer stem cell maintenance, protection and entail sites of infiltrating immune cells [[1], [2], [3], [4]]. Various mechanisms for the reinstatement of blood vessel growth have been described of which angiogenesis, the sprouting of new vessels from pre-existing capillaries and postcapillary venules is the most commonly used mode [[5], [6], [7]]. Subsequently, tip cells anastomose with tip cells from neighboring sprouts to connect the newly formed vascular structures with the help of bridging macrophages. With the initiation of blood flow, the establishment of a basement membrane, and the recruitment of vessel-stabilizing pericytes, vascular growth is then terminated [5,6]. Thus, under physiological conditions angiogenesis is tightly regulated by a homeostatic balance of a wealth of proangiogenic and inhibitory factors of which members of the vascular endothelial growth factor (VEGF), and angiopoietin tyrosine kinase receptor families as well as various members of the CXC and CC chemokines are the most prominent molecules to orchestrate this multistep process [1,8]. The balance of angiogenesis-promoting and inhibiting molecules is, however, lost in tumors. Upon initiating neovascularization, tumors continue to promote angiogenic signaling for several reasons [9]. First, tumors express oncogenes and/or lose tumor suppressor genes that result in the activation of pro-angiogenic signaling pathways [[10], [11], [12]]. Second, expanding tumors exhibit a continually and abnormally growing tumor vasculature with a typical display of irregular-shaped, hyperdilated and poorly pericyte-covered tumor vessels that cause a leaky and sluggish blood flow [13]. These typical signs of poor vessel maturation increase interstitial pressure and generate and maintain a hypoxic and acidic environment that continues to elevate proangiogenic factors thereby further aggravating a pro-angiogenic feedback loop that never ceases.

Effects of low-oxygen tension are mediated by hypoxia-inducible factor (HIF) transcription factors that coordinate a transcriptional program to ensure metabolic and vascular adaptations under hypoxic conditions by various mechanisms [[14], [15], [16]]. Hypoxia upregulates various proangiogenic growth factors and chemokines that directly engage in vessel growth like VEGF, PlGF and Ang2 [[17], [18], [19], [20], [21]]. In addition, numerous experimental models have shown that both hypoxic and acidic conditions in the tumor microenvironment change the availability of metabolites and induce the secretion of molecules in tumors (e.g. CSF1, GM-CSF, G-CSF, CX3CL1, CXCL5, CXCL12, CCL17, CCL22, IL6, Sema3) that attract innate immune cells to the tumor site where they become reprogrammed to serve as an additional source of angiogenic factors [[22], [23], [24], [25], [26], [27], [28]]. It is important to note that innate immune cells elicit a high plasticity in order to quickly adapt and serve the homeostatic repair program. Upon injury, first neutrophils and then macrophages are recruited to the wound that are immunosupportive and angiostatic due to their responsibility to phagocytose and kill bacteria and degrade necrotic tissue. In the subsequent resolution phase, myeloid cells then convert to an angiogenic and immunosuppressive phenotype to support tissue and blood vessel restoration [29]. The ability of myeloid cells to display such opposing properties to either eliminate potential infections or hinder excessive inflammation is indeed pivotal to properly regulate tissue injury and maintain tissue homeostasis [30]. In line with the concept that tumors are wounds that never heal [31], tumors take advantage of the myeloid plasticity by reprogramming these cells to exhibit both immunosuppressive and angiogenic features to enable tumors to escape immunosurveillance and facilitate vascular expansion. These two features have been defined as pivotal hallmarks in cancer propagation and progression [32].

There is increasing evidence that tumor-supporting inflammation and angiogenesis, although being distinct and separable processes, are closely related events that are in part regulated by common chemokines [32,33]. VEGF and Ang2 are proangiogenic factors that directly and indirectly convey immunosuppression in endothelial cells as well as innate and adaptive immune cells [[34], [35], [36], [37]]. VEGF induces an immunosuppressive vasculature on different levels (Fig.1). It downregulates the expression of the vascular adhesion molecules I-CAM1 and V-CAM1 that are necessary for T-cells to cross the endothelial layer and transit into the tumor. VEGF and Ang2 have been shown to enhance the expression of the negative checkpoint regulator programmed cell death protein 1 (PD-1)–programmed cell death 1 ligand 1 (PD-L1) in endothelial cells thus disabling the cytotoxic function of PD1+-T cells [[38], [39], [40], [41]]. In addition, elevated FasL levels on tumor ECs is known to trigger apoptosis of Fas-expressing CD8 + T cells [42]. Tumor-associated ECs also preferentially attract immunosuppressive Tregs by upregulating the multifunctional endothelial receptor CLEVER-1/stabilin-1 [43]. All these mechanisms consequently lead to a potent barrier that disables cytotoxic T cell infiltration into the tumor. VEGF, however, not only facilitates VEGFR-2 signaling in endothelial cells but also in VEGFR2+ immune cells. Thereby, it stimulates the expression of PD-L1 in dendritic cells and blocks the maturation and thus, functionality of dendritic cells due to their impaired ability to present tumor antigens [44]. VEGF/VEGFR2 signaling further suppresses proliferation of effector T cells but increases and promotes tumor homing of Tregs [32,44]. This effect is dependent on neuropilin-1 (nrp1) because in an experimental tumor model, nrp1 depletion resulted in reduction of Tregs with a concomitant increase in cytotoxic T cells [45]. VEGF also upregulates several negative immune checkpoint receptors besides PD-1 (e.g. CTLA-4, Lag-3, TIM-3) on T cells contributing to T-cell anergy and exhaustion [46]. Finally, elevated levels of VEGF and/or Ang-2 as well as other tumor-secreted factors promote the recruitment and expansion of innate immune cell populations including TAMs, TEMs, neutrophils, MDSC and immature dendritic cells that secrete molecules fostering an immunosuppressive and angiogenic tumor microenvironment.

Section snippets

Innate immune cells promote angiogenesis

As discussed above, tumor-associated macrophages (TAM) and tumor-associated neutrophils (TAN) can either convey angiostatic and immunosupportive or proangiogenic and immunosuppressive features, but in tumors are commonly found to polarize to an immunosuppressive as well as angiogenic phenotype [[47], [48], [49]]. In addition, immature Gr1+ cells with either a mononuclear or granular morphology have been identified in tumors that promote immunosuppressive functions and are therefore also termed

Metabolic pathways in immune cells regulate angiogenesis

The tumor induces a major disturbance in tissue homeostasis and cellular metabolism by endorsing a hypoxic and acidic microenvironment that strongly affects metabolic availability not only for tumor cells but also other cell constituents within the tumor (Fig.2). Although it is well-established that intratumoral hypoxia induces the recruitment of immunosuppressive and angiogenic myeloid cells to the tumor site, it is less understood how subsequent changes in metabolite availability for immune

Immune cells facilitate resistance to antiangiogenic therapy

Compelling data from multiple laboratories have provided a convincing rationale for the development of VEGF and VEGF receptor inhibitors to block intratumoral vascular growth and subsequent tumor propagation. Subsequently, bevacizumab (Avastin, Genentech/Roche), a monoclonal antibody against VEGF, as well as sorafenib (Nexavar, Bayer) and sunitinib (Sutent, Pfizer), both kinase inhibitors that target VEGF receptor (VEGFR) were the first FDA-approved angiogenic inhibitors followed by other

Antiangiogenic therapy meets immunotherapy

Various studies including those mentioned above, have provided compelling evidence that antiangiogenic therapy is most efficacious when an immunostimulatory environment is generated. This is in agreement with the concept of vascular normalization to reduce hypoxia and polarize innate immune cells to an immunosupportive phenotype. Emerging from these studies is the proposition that angiogenesis and inflammation are reciprocally regulated and that immune cells play a pivotal role in regulating

Conclusions

Currently, various combinatorial treatment modalities of angiogenic inhibitors, specifically those targeting the VEGF/R and Ang2/Tie axis with immune checkpoint inhibitors and other immunotherapies are evaluated in the clinical setting [116]. These trials have been initiated based on compelling evidence from mouse tumor model systems that such a combination modulates both the tumor vasculature and the immune system to foster an immunosupportive environment and thus improves and endures efficacy

Sources of funding

This work was supported by grants from the ERC CoG ImmunoFIT Methusalem (to MM) and from the Flamish government FWO (G066515N) (to MM) and FWO (G0A0818N) (to GB) and the Belgian Association Against Cancer for the project 2014-197 (to MM) and from the National Institute of Health NIH/NCI (to GB).

Competing interests

None.

References (144)

  • M. De Palma et al.

    Tie2-expressing monocytes: regulation of tumor angiogenesis and therapeutic implications

    Trends Immunol.

    (2007)
  • G.T. Motz et al.

    Deciphering and reversing tumor immune suppression

    Immunity

    (2013)
  • Z.G. Fridlender et al.

    Polarization of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN

    Cancer Cell

    (2009)
  • M. Potente et al.

    Basic and therapeutic aspects of angiogenesis

    Cell

    (2011)
  • C. Rolny et al.

    HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PIGF

    Cancer Cell

    (2011)
  • C. Betsholtz

    Insight into the physiological functions of PDGF through genetic studies in mice

    Cytokine Growth Factor Rev.

    (2004)
  • E. De Falco et al.

    SDF-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells

    Blood

    (2004)
  • M. De Palma et al.

    Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors

    Cancer Cell

    (2005)
  • R. Mazzieri et al.

    Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells

    Cancer Cell

    (2011)
  • C.E. Lewis et al.

    The multifaceted role of perivascular macrophages in tumors

    Cancer Cell

    (2016)
  • L. Tamagnone et al.

    Semaphorin signals on the Road of endothelial tip cells

    Dev. Cell

    (2011)
  • L. Tamagnone

    Emerging role of semaphorins as major regulatory signals and potential therapeutic targets in cancer

    Cancer Cell

    (2012)
  • A. Casazza et al.

    Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity

    Cancer Cell

    (2013)
  • S.B. Coffelt et al.

    Immune-mediated mechanisms influencing the efficacy of anticancer therapies

    Trends Immunol.

    (2015)
  • M. Gaudry et al.

    Intracellular pool of vascular endothelial growth factor in human neutrophils

    Blood

    (1997)
  • K. Movahedi et al.

    Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity

    Blood

    (2008)
  • L.B. Rivera et al.

    Intratumoral myeloid cells regulate responsiveness and resistance to antiangiogenic therapy

    Cell Rep.

    (2015)
  • P. Andreu et al.

    FcRgamma activation regulates inflammation-associated squamous carcinogenesis

    Cancer Cell

    (2010)
  • D.G. DeNardo et al.

    CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages

    Cancer Cell

    (2009)
  • V. Huber et al.

    Cancer acidity: an ultimate frontier of tumor immune escape and a novel target of immunomodulation

    Semin. Cancer Biol.

    (2017)
  • P.P. Hsu et al.

    Cancer cell metabolism: Warburg and beyond

    Cell

    (2008)
  • K. De Bock et al.

    Role of endothelial cell metabolism in vessel sprouting

    Cell Metab.

    (2013)
  • G. Bergers et al.

    Tumorigenesis and the angiogenic switch

    Nat. Rev. Cancer

    (2003)
  • P. Carmeliet

    Angiogenesis in life, disease and medicine

    Nature

    (2005)
  • R.H. Adams et al.

    Molecular regulation of angiogenesis and lymphangiogenesis

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • L. Rivera et al.

    Escape mechanisms from antiangiogenic therapy: an immune cell’s perspective

    Adv. Exp. Med. Biol

    (2014)
  • M. Santoni et al.

    CXC and CC chemokines as angiogenic modulators in nonhaematological tumors

    Biomed. Res. Int.

    (2014)
  • Folkman, J. (2000) Tumor angiogenesis in Cancer Medicine (al., H. e., ed), B C Decker, Hamilton,...
  • P. Carmeliet et al.

    Molecular mechanisms and clinical applications of angiogenesis

    Nature

    (2011)
  • R.K. Jain

    Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy

    Science

    (2005)
  • R.K. Jain

    Normalizing tumor microenvironment to treat cancer: bench to bedside to biomarkers

    J. Clin. Oncol.

    (2013)
  • D. Liao et al.

    Hypoxia-inducible factor-1alpha is a key regulator of metastasis in a transgenic model of cancer initiation and progression

    Cancer Res.

    (2007)
  • G.L. Semenza

    Oxygen sensing, homeostasis, and disease

    N. Engl. J. Med

    (2011)
  • B. Keith et al.

    HIF1alpha and HIF2alpha: sibling rivalry in hypoxic tumour growth and progression

    Nat. Rev. Cancer

    (2011)
  • J. LeCouter et al.

    Identification of an angiogenic mitogen selective for endocrine gland endothelium

    Nature

    (2001)
  • M.P. Simon et al.

    The angiopoietin-2 gene of endothelial cells is up-regulated in hypoxia by a HIF binding site located in its first intron and by the central factors GATA-2 and ets-1

    J. Cell. Physiol.

    (2008)
  • J.A. Forsythe et al.

    Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1

    Mol. Cell. Biol.

    (1996)
  • B.D. Kelly et al.

    Cell type-specific regulation of angiogenic growth factor gene expression and induction of angiogenesis in nonischemic tissue by a constitutively active form of hypoxia-inducible factor 1

    Circ. Res.

    (2003)
  • E. Giraudo et al.

    An amino-bisphosphonate targets MMP-9-expressing macrophages and angiogenesis to impair cervical carcinogenesis

    J. Clin. Invest.

    (2004)
  • E.Y. Lin et al.

    Macrophages regulate the angiogenic switch in a mouse model of breast cancer

    Cancer Res.

    (2006)
  • Cited by (65)

    • Inducing vascular normalization: A promising strategy for immunotherapy

      2022, International Immunopharmacology
      Citation Excerpt :

      In addition, increased VGEF directly inhibits cytotoxic T lymphocytes (CTLs) transport, proliferation, and effector functions and also inhibits DCs maturation and antigen presentation, impairing T cell-mediated anti-cancer immune responses[61–64]. The tumor vasculature also further deteriorates the TME to produce immunosuppression[65]. In the TME, cells that promote anti-tumor immunity, such as mature DCs, are relatively deficient, while cells that inhibit anti-tumor immunity are quite abundant, such as tumor-associated macrophages (TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs)[66–68].

    • Strategies of engineering nanomedicines for tumor retention

      2022, Journal of Controlled Release
      Citation Excerpt :

      Because of the rapid proliferation of cancer cells and relative low propagation rate of blood capillaries cells, the tumor vessels are forced apart and the vascular density is reduced [30,43,44]. Moreover, the tumor vessels often exhibit irregular structures with impaired perfusions, and varied with tumor types and stages, which significantly restrict the entry of therapeutic agents into tumor tissues [30,45–49]. The rigid and dense networks of ECM represent a substantial obstacle hampering nanomedicines retention in tumors, which is ubiquitously distributed around and throughout the tumor tissues [24,50–52].

    • The acidic tumour microenvironment: Manipulating the immune response to elicit escape

      2022, Human Immunology
      Citation Excerpt :

      Induction of VEGF expression by ECs also downregulate the expression of adhesion markers, such as intercellular cell adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), on blood vessels in the TME. This prevents CTLs from infiltrating the tumour [31]. VEGF also stimulates PD-L1 expression, inhibiting CTLs [31].

    View all citing articles on Scopus
    View full text