Skip to main content

MINI REVIEW article

Front. Pharmacol., 21 March 2024
Sec. Experimental Pharmacology and Drug Discovery
This article is part of the Research Topic The Pharmacological Modulation of Angiogenesis View all articles

Aberrant tumor vasculature. Facts and pitfalls

  • Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy

Endothelial cells form a single cell layer lining the inner walls of blood vessels and play critical roles in organ homeostasis and disease progression. Specifically, tumor endothelial cells are heterogenous, and highly permeable, because of specific interactions with the tumor tissue environment and through soluble factors and cell–cell interactions. This review article aims to analyze different aspects of endothelial cell heterogeneity in tumor vasculature, with particular emphasis on vascular normalization, vascular permeability, metabolism, endothelial-to-mesenchymal transition, resistance to therapy, and the interplay between endothelial cells and the immune system.

Introduction

The endothelium of large and small vessels, including arteries characterized by continuous endothelium, aligned in the direction of flow and without valves, veins, characterized by continuous endothelium, not aligned in the direction of flow, with valves, and capillaries, characterzied by endothelium adapted to the underlying tissues and with phenotypic differences between different vascular beds (Ribatti et al., 2002; Crivellato et al., 2007). Genetic and environmental factors influence endothelial heterogeneity through the release of specific soluble factors or cell–cell interactions, involved in determining specific vascular structure and function (Ribatti et al., 2002; Crivellato et al., 2007). High permeability and fenestrations are dependent on the secretion of vascular permeability factor/vascular endothelial growth factor (VPF/VEGF) (Dvorak et al., 1992; Esser et al., 1998). Capillary endothelial cells show heterogenic characteristics between different organs (continuous thick capillaries are present in skeletal muscle, cardiac smooth muscle, and testes; thin continuous capillaries are present in the central nervous system and dermis; sinusoids are present in the liver, spleen, and bone marrow; fenestrated capillaries are present in endocrine glands), and also in single organs, such as in the kidney where are present fenestrated endothelial cells in peritubular capillaries, discontinuous endothelial cells in glomerular capillaries, and continuous endothelial cells in other regions (Ribatti et al., 2009). In both between-organ and between-vessel type differences, heterogeneity arises from the necessity for endothelial specialization. Endothelial cells in these differing vascular beds have unique molecular functions that drive their particular structure and molecular phenotype.

Endothelial cells are also able to secrete specific angiocrine factors, such as VEGF, angiopoietin-2 (Ang-2), bone morphogenetic protein-2, -4 (BMP-2, -4), C-X-C motif chemokine-12 (CXCL-12), fibroblast growth factor-2 (FGF-2), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), insulin growth factor-1 (IGF-1), interleukin-3, -6, -8 (IL-3, IL-6, IL-8), pentraxin-3 (PTX-3), placental growth factor (PlGF), platelet-derived growth factor (PDGF), transforming growth factor beta (TGF-β), able to modulate the growth and morphogenesis of specific organs, such as the liver, kidney, and bone marrow (Ribatti et al., 2023a; b, c), and are involved also in cancer progression and metastasis (Maishi et al., 2019; Butler et al., 2010).

A remarkable endothelial cell heterogeneity in different organs has been demonstrated by single-cell RNA sequencing (scRNA-seq) (Kalucha et al., 2020), which is altered in tumors, such as occurs in breast cancer, where subsets of tumor endothelial cells are involved in cancer metabolism and transport (Geldhof et al., 2022). Moreover, endothelial cells from lung cancer compared to normal endothelial cells show a strong signature in signaling pathways such as the MYC and PI3K/Akt/mTOR (Lambrechts et al., 2018). Pericytes in tumor vessels are present but abnormal, lacking an intimate association with endothelial cells (Morikawa et al., 2002), and VEGF inhibitors induce a close association of pericytes with endothelial cells (Inoi et al., 2004).

This review article aims to analyze different aspects of endothelial cell heterogeneity in tumor vasculature, with particular emphasis on vascular normalization, vascular permeability, metabolism, endothelial-to-mesenchymal transition, resistance to therapy, and the interplay between endothelial cells and the immune system.

Tumor endothelial cells

Tumor endothelial cells are characterized by immaturity and leaky, lack of perivascular cell coverage, and loss of basement membrane (Baluk et al., 2003; di Tomaso et al., 2005) favoring the passage of T cells (Di Russo et al., 2017), form arteriovenous shunts (a category of vessels with a very low resistance to flow), show chaotic and sluggish blood flow, which does not follow a unidirectional path, and proceed in alternating directions (Chaplin et al., 1987; Mc Donald and Baluk, 2002; Mc Donald and Choyke, 2003). Only 0.1%–3% of all normal endothelial cells turn over daily declining with age (Schwartz and Benditt, 1973), whereas in tumors, endothelial cell turnover may be 20–2,000 times the rate in normal tissues (Hobson and Denekamp, 1984). Vessels are more numerous at the tumor-host interface, whereas the internal portions are less vascularized. Glomeruloid microvascular proliferations, consisting of poorly organized structures resembling renal glomeruli, have been found in different human tumors (Straume et al., 2003). Alternative vascularization mechanisms, different from classic angiogenesis, have been described in tumors, including vascular co-option, vasculogenic mimicry, and intussusceptive microvascular growth (Ribatti and Pezzella, 2021).

Tumor endothelial cells are genetically unstable with embryonic characteristics promoting pro-tumor and anti-inflammatory behavior (St Croix et al., 2000; Seaman et al., 2007; Huijbers et al., 2022). The gene expression patterns of vascular endothelial cells derived from normal and malignant colorectal tissues have been investigated (St Croix et al., 2000), showing that among 79 transcripts differentially expressed, 46 were elevated and 33 were expressed at lower levels in tumor-associated endothelial cells. The transcriptional profiling results of tumor endothelial cells from multiple studies and multiple tumor types have been compared (Aird, 2009), showing that a few overexpressed genes were shared by different tumors including matrix metalloproteinase 9 (MMP9) (ovary and breast), HEYL (breast and colon), and secreted protein acidic and rich in cysteine (SPARC) (breast and colon and brain), whereas most genes were limited to one tumor type or invasive tumors. The upregulation of several genes, such as lysyl oxidase (Osawa et al., 2013), suprabasin (Alam et al., 2014), and biglycan (Yamamoto et al., 2012) enhances the migration and tube-forming capacity of tumor endothelial cells. Upregulated expression of stemness genes such as stem cell antigen-1 (Sca-1) and MDR-1 (Matsuda et al., 2010) and aldehyde dehydrogenase (ALDH) (Ohmura-Kakutani et al., 2014) has been demonstrated in tumor endothelial cells, as a part of the tumor endothelial cell population (Nagy and Dvorak, 2012; Goveia et al., 2020). CD133+ tumor endothelial cells have a higher frequency of aneuploidy than the CD133- ones, suggesting that tumor endothelial cells originating from progenitor cells are involved in inducing genetic instability in these cells (Akino et al., 2010). Progenitor-derived tumor endothelial cells that express CD133 are undifferentiated, highly proliferative cells (Rafii et al., 2002).

In tumor endothelial cells proangiogenic molecules, including VEGF receptor (VEGFR)-1, −2, −3, VEGF-D, Tie-2, and Ang-1 are upregulated when compared with normal endothelial cells (Bussolati et al., 2003), favoring a proangiogenic phenotype (Matsuda et al., 2010). Moreover, tumor endothelial cells show different responsiveness to epidermal growth factor (EGF) (Amin et al., 2006), adrenomedullin (Tsuchiya et al., 2010), and VEGF (Matsuda et al., 2010) compared with normal endothelial cells.

Endothelial cells from high metastatic tumors show upregulation of VEGF, VEGFR-1, VEGFR-2, MMP-2, MMP-9, and display increased Akt phosphorylation compared with low metastatic ones (Ohga et al., 2012).

Activin-like receptor kinase 1 (ALK1) expression in tumor endothelial cells is a prognostic factor for metastasis of breast cancer, because pharmacologic targeting of ALK1 provided long-term therapeutic benefit in mouse models of mammary carcinoma, accompanied by strikingly reduced metastatic colonization (Cunha et al., 2015). Prolyl hydroxylase domain protein 2 (PHD2) deficiency normalized tumor blood vessels, associated with a reduction of tumor cell intravasation and metastasis (Mazzone et al., 2009). Biglycan is upregulated in tumor endothelial cells of metastatic tumors, facilitating the migration of toll-like receptor 2/4+tumor cells, which increases circulating tumor cells and lung metastasis (Maishi et al., 2016).

Tumor endothelial cells and vessel normalization

Vessel normalization through anti-VEGF agents improves perfusion and more efficient local delivery of oxygen, decreases vascular leakiness, and reduces intratumoral hypoxia improving pericyte recruitment (Jain, 2001; Huang et al., 2012) allowing drug delivery and immune cell infiltration, and increasing the sensitivity of the tumor cells to radiation and chemotherapy.

VEGFR2 blockade induces upregulation of Ang1 which promotes endothelial cell junctions thickening and stabilization of endothelial cells (Winkler et al., 2004). Moreover, VEGF blockade resulted in reduced interstitial fluid pressure, and tissue edema, increased perfusion, and enhanced oxygenation and drug delivery to the tumor core. The transient effect of tumor vascular normalization might be associated either with excessively high and continuous administration of anti-angiogenic drugs or the development of drug resistance due to the activation of other pro-angiogenic factors (Bergers and Hanahan, 2008).

Tumor endothelial cells and vascular permeability

Plasma components extravasate across vascular endothelium by paracellular (through inter-endothelial cell junctions) and transcellular [caveolae, fenestrae and vesiculo-vacuolar organelles (VVOs)] routes. Tumor vessel leakiness occurs through VVOs and trans endothelial cell pores resulting from VVOs activated by VEGF (Feng et al., 1999; Ribatti and Tamma, 2018).

Although tumor vessels can have barrier defects large enough for hemorrhage, plasma leakage in tumors is limited by reduced driving force due to poor vascular perfusion and high interstitial pressure resulting from impaired lymphatic drainage (Mc Donald and Baluk, 2002; Jain et al., 2014.) High tumor interstitial fluid pressure causes blood vessel collapse and impedes blood flow, and delivery of therapeutics to the central region of the tumor, causing hypoxia in tumor tissue (Boucher and Jain, 1992). Hypoxia, in turn, makes tumor cells resistant to radiation therapy, induces numerous genes that make tumor cells resilient to cytotoxic drugs, causes genetic instability within tumor cells, and triggers genetic mutations making the tumor cells more malignant and prone to metastasis. Low permeability tumors may overexpress Ang-1 and under express VEGF or PlGF, whereas those with high permeability may lack Ang-1 or overexpress its antagonist Ang-2 (Jain and Munn, 2000). Hypoxia and the secretion of angiogenic cytokines, favor tumor revascularization through the mobilization of bone marrow-derived endothelial progenitor cells (Gao et al., 2009).

Tumor endothelial cells and metabolism

Emerging evidence has suggested that endothelial metabolism allows endothelial cells to adapt to the tissue-specific functions are supply the tissue with the necessary nutrients that it imports from the circulating blood. Dysregulation of endothelial cell metabolism has been associated with many diseases including atherosclerosis, diabetes, neovascular eye disease, and cancer. Glycolysis-related genes are overexpressed in the transcriptomic signature of tumor endothelial cells (Rohlenova et al., 2020). Glucose uptake and glycolysis are higher in tumor endothelial cells (Garcia-Caballero et al., 2022). Dysfunctional metabolism in tumor endothelial cells produces excessive lactate and 2-hydroxyglutarate (2-HG), inhibiting the cytotoxic functions of T cells (Tyrakis et al., 2016). Altered glycolysis in tumor endothelial cells due to an upregulated expression of glycolysis genes, contributes to structural deformities observed in tumor blood vessels (Cantelmo et al., 2016). Tumor endothelial cells proliferate under lactic acidosis caused by tumor cell glycolytic metabolism, and the pH regulator, carbonic anhydrase 2 (CAII), is involved in resistance to low pH in tumor endothelial cells (Annan et al., 2020).

Tumor endothelial cells and endothelial to mesenchymal transition

Endothelial cells may de-differentiate into mesenchymal stem-like cells (Medici and Kalluri, 2012), a process named endothelial to mesenchymal transition (EndoMT) (Ribatti, 2022). During EndoMT, endothelial cells lose endothelial markers, including platelet endothelial cell adhesion molecule-1 (PECAM-1), Tie-2, and vascular endothelial (VE)-cadherin, and acquire mesenchymal markers, including N-cadherin, fibroblast specific protein-1 (FSP-1), alpha-smooth muscle actin (αSMA), types I/III collagen, and vimentin. The endothelial cytoskeleton rearrangement associated with EndoMT promotes intravasation and extravasation of tumor endothelial cells (Reymond et al., 2013).

Tumor-induced EndoMT is associated with the activation of pro-inflammatory pathways in endothelial cells (Nie et al., 2014). Endothelial cells undergoing tumor induced EndoMT express higher levels of the VEGF gene (Hong et al., 2018), and EndoMT contributes to metastatic extravasation and intravasation (Dudley et al., 2012). In glioblastoma, tumor endothelial cells secrete extracellular vesicles which induce mesenchymal reprogramming of cancer cells (Adnani et al., 2002).

Tumor endothelial cells and resistance to therapy

Renal carcinoma endothelial cells are resistant to vincristine (Bussolati et al., 2003), and hepatocellular carcinoma endothelial cells are resistant to 5-fluorouracil and Adriamycin (Xiong et al., 2009; Ohga et al., 2012). Endothelial cells of metastatic melanoma have a higher expression of MDR-1 (Akiyama et al., 2012) and ALDH and are resistant to paclitaxel (Hida et al., 2017). IGFBP7 expressed by tumor endothelial cells suppresses IGF1R signaling and the stem-cell-like property of tumor cells. Chemotherapy triggers tumor endothelial cells to suppress IGFBP7, and the upregulation of IGF1 activates the FGF4-FGFR1-ETS2 pathway and accelerates the conversion of tumor cells to chemo-resistant tumor stem-like cells (Cao et al., 2017).

Vasculogenic mimicry and vascular co-option are involved in intrinsic and acquired resistance. Vasculogenic mimicry is associated with poor prognosis, reduced survival, and a high risk of cancer recurrence (Li et al., 2016). Histological examination of glioma bioptic specimens of patients who died after receiving treatment with cediranib, an inhibitor of VEGFR-2 (di Tomaso et al., 2011), or bevacizumab (de Groot et al., 2010) demonstrated that glioma cells grow around pre-existing vessels in a non-angiogenic fashion.

Tumor endothelial cells and the immune system

Tumor endothelial cells promote the loss of protective anti-cancer immunity, the so-called “endothelial anergy” (De Sanctis et al., 2018), corresponding to the unresponsiveness of tumor endothelial cells to pro-inflammatory stimulation, impeding the adhesion and migration of immune cells (Griffioen et al., 1996; Lambrechts et al., 2018). Endothelial anergy is reversible (several therapeutic approaches have been developed to reverse tumor endothelial cell anergy and thus favor the intra-tumoral recruitment of anti-tumor immune cells) and may be used as a therapeutic strategy, suggesting that blocking this mechanism in tumor endothelial cells favors the influx of immune cells (Facciabene et al., 2017). It has been demonstrated that anti-angiogenic therapy could revert endothelial cell anergy, allow leukocytes to infiltrate tumors, and stimulate anti-tumor immunity (Nowak-Sliwinska et al., 2023).

Endothelial cells regulate leukocyte extravasation through the expression of adhesion molecules, such as selectins, intercellular cellular adhesion molecule-1 (ICAM-1), vascular cellular adhesion molecule-1 (V-CAM-1), PECAM-1, and through the weakening of endothelial cell-cell contacts allowing transmigration of immune cells (Wettschurek et al., 2019). Tumor endothelial cells fail to express proper ICAM-1 and VCAM-1 levels (Huijbers et al., 2022). The glycosylation of surface molecules modulates the adhesive properties of tumor endothelial cells and can either enhance or reduce immune cell migration (Chandler et al., 2019). The vasoconstrictive peptide endothelin 1 (ET1) is associated with ICAM-1 expression and the decreased presence of tumor-infiltrating leukocytes (Buckanovich et al., 2008).

Tumor endothelial cells secrete IL-6 and CSF-1 which promote anti-tumor alternative macrophage polarization by triggering Akt1/mTOR pathway, resulting in anti-inflammatory and pro-tumorigenic macrophage activation (Wang et al., 2018). Endothelial cell-derived CXCL-12 promotes monocyte recruitment and macrophage education by tumor cells (Alsina-Sanchis et al., 2022). Endothelial cell-derived PGE2 and IL-10 restrict T-cell activity (Mulligan and Young, 2010).

Tumor endothelial cells downregulate genes responsible for major histocompatibility complex (MHC) expression impeding their antigen-presenting functions, thus contributing to tumor immune evasion (Goveia et al., 2020). The binding of inhibitory immune checkpoints (e.g., PD-1) on CD8+ cells with their ligands (e.g., PD-L1 and PD-L2) on tumor endothelial cells inhibits T cell activation and these ligands can be upregulated by tumor endothelial cells on proinflammatory factors (Georganaki et al., 2018).

Discussion

In this review, we addressed the abnormality and heterogeneity of tumor endothelial cells through the analysis of different aspects of this heterogeneity, including vascular normalization, vascular permeability, metabolism, endothelial-to-mesenchymal transition, resistance to therapy, and the interplay between endothelial cells and the immune system.

Endothelial cell heterogeneity can be quantified through epigenomic, transcriptomic, and proteomic studies. The characterization and understanding of endothelial cell heterogeneity have advanced in the past years, due to the development of single-cell OMICs approaches. ScRNA-seq methods for tissue-derived cell suspensions and cultured cell populations have been an area of intense development. Single-cell transcriptional sequencing (scRNA-seq) techniques enable gene expression analyses at a single cell level, investigating the transcriptional output of cells in both normal and tumoral tissue samples. Thanks to the identification of preferentially expressed genes, gene expression study permits to identification of not only different cell types, but also various cell states progression along the cell cycle, different metabolic states, or rather the diversity within each of the clusters defined as “cell types.” More work on vascular single-cell analysis is required to establish the principles of endothelial activation and their interpretation for the different tissue challenges that require vascular adaptations (Pasut et al., 2021; Becker et al., 2023).

The knowledge on tumor endothelial cell phenotypes is under continuous development, even if their role in immune escape and the response to immune and anti-angiogenic therapies should be further analyzed and clarified. Notably, most human tumor types contain varying numbers but only a small population of angiogenic tumor endothelial cells, the targets of anti-angiogenic therapies, contributing to the limited efficacy of and resistance to these therapies.

Author contributions

DR: Writing–original draft, Writing–review and editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. Associazione Italiana Leucemie e Linfomi (AIL).

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Adnani, L., Kassouf, J., Meehan, B., Tawil, N., Nakano, I., et al. (2022). Angiocrine extracellular vesicles impose mesenchymal reprogramming upon proneural glioma stem cells. Nat. Commun. 13, 5494. doi:10.1038/s41467-022-33235-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Aird, W. C. (2009). Molecular heterogeneity of tumor endothelium. Cell Tissue Res. 335, 271–281. doi:10.1007/s00441-008-0672-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Akino, T., Hida, K., Hida, Y., Tsuchiya, K., Freedman, D., Muraki, C., et al. (2010). Cytogenetic abnormalities of tumor-associated endothelial cells in human malignant tumors. Am. J. Pathol. 175, 2657–2667. doi:10.2353/ajpath.2009.090202

PubMed Abstract | CrossRef Full Text | Google Scholar

Akiyama, K., Ohga, N., Hida, Y., Kawamoto, T., Sadamoto, Y., Ishikawa, S., et al. (2012). Tumor endothelial cells acquire drug resistance by MDR1 up-regulation via VEGF signaling in tumor microenvironment. Am. J. Pathol. 180, 1283–1293. doi:10.1016/j.ajpath.2011.11.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Alam, M. T., Nagao-Kitamoto, H., Ohga, N., Akiyama, K., Maishi, N., Kawamoto, T., et al. (2014). Suprabasin as a novel tumor endothelial cell marker. Cancer Sci. 105, 1533–1540. doi:10.1111/cas.12549

PubMed Abstract | CrossRef Full Text | Google Scholar

Alsina-Sanchis, E., Mülfarth, R., Moll, I., Böhn, S., Wiedmann, L., Jordana-Urriza, L., et al. (2022). Endothelial RBPJ is essential for the education of tumor-associated macrophages. Cancer Res. 82, 4414–4428. doi:10.1158/0008-5472.CAN-22-0076

PubMed Abstract | CrossRef Full Text | Google Scholar

Amin, D. N., Hida, K., Bielenberg, D. R., and Klagsbrun, M. (2006). Tumor endothelial cells express epidermal growth factor receptor (EGFR) but not ErbB3 and are responsive to EGF and to EGFR kinase inhibitors. Cancer Res. 66, 2173–2180. doi:10.1158/0008-5472.CAN-05-3387

PubMed Abstract | CrossRef Full Text | Google Scholar

Annan, D. A., Kikuchi, H., Maishi, N., Hida, Y., and Hida, K. (2020). Tumor endothelial cell. A biological tool for translational cancer research. Int. J. Mol. Sci. 21, 3238. doi:10.3390/ijms21093238

PubMed Abstract | CrossRef Full Text | Google Scholar

Baluk, P., Morikawa, S., Haskell, A., and Mancuso, M. (2003). Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 163, 1801–1815. doi:10.1016/S0002-9440(10)63540-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Becker, L. M., Chen, S. H., Rodir, J., de Rooij, L. P. M. H., Baker, A. H., and Carmeliet, P. (2023). Deciphering endothelial heterogeneity in health and disease at single-cell resolution: progress and perspectives. Cardiovasc. Res. 119, 6–27. doi:10.1093/cvr/cvac018

PubMed Abstract | CrossRef Full Text | Google Scholar

Bergers, G., and Hanahan, D. (2008). Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 8, 592–603. doi:10.1038/nrc2442

PubMed Abstract | CrossRef Full Text | Google Scholar

Boucher, Y., and Jain, R. K. (1992). Microvascular pressure is the principal driving force for interstitial hypertension in solid tumors: implications for vascular collapse. Cancer Res. 52, 5110–5114.

PubMed Abstract | Google Scholar

Buckanovich, R. J., Facciabene, A., Kim, S., Benencia, F., Sasaroli, D., Balint, K., et al. (2008). Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat. Med. 14, 28–36. doi:10.1038/nm1699

PubMed Abstract | CrossRef Full Text | Google Scholar

Bussolati, B., Deambrosis, I., Russo, S., Deregibus, M. C., and Camussi, G. (2003). Altered angiogenesis and survival in human tumor-derived endothelial cells. FASEB J. 17, 1159–1161. doi:10.1096/fj.02-0557fje

PubMed Abstract | CrossRef Full Text | Google Scholar

Butler, J. M., Kobayashi, H., and Rafii, S. (2010). Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 10, 138–146. doi:10.1038/nrc2791

PubMed Abstract | CrossRef Full Text | Google Scholar

Cantelmo, A. R., Conradi, L. C., Brajic, A., Goveia, J., Kalucka, J., Pircher, A., et al. (2016). Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy. Cancer Cell 30, 968–985. doi:10.1016/j.ccell.2016.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, Z., Scandura, J. M., Inghirami, G. G., Shido, K., Ding, B. S., and Rafii, S. (2017). Molecular checkpoint decisions made by subverted vascular niche transform indolent tumor cells into chemoresistant cancer stem cells. Cancer Cell 31, 110–126. doi:10.1016/j.ccell.2016.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Chandler, K. B., Costello, C. E., and Rahimi, N. (2019). Glycosylation in the tumor microenvironment: implications for tumor angiogenesis and metastasis. Cells 8, 544. doi:10.3390/cells8060544

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaplin, D. J., Olive, P. L., and Durand, R. E. (1987). Intermittent blood flow in a murine tumor: radiobiological effects. Cancer Res. 47, 597–601.

PubMed Abstract | Google Scholar

Crivellato, E., Nico, B., and Ribatti, D. (2007). Contribution of endothelial cells to organogenesis: a modern reappraisal of an old Aristotelian concept. J. Anat. 211, 415–427. doi:10.1111/j.1469-7580.2007.00790.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cunha, S. I., Bocci, M., Lovrot, J., Eleftheriou, N., Roswall, P., Cordero, E., et al. (2015). Endothelial ALK1 is a therapeutic target to block metastatic dissemination of breast cancer. Cancer Res. 75, 2445–2456. doi:10.1158/0008-5472.CAN-14-3706

PubMed Abstract | CrossRef Full Text | Google Scholar

de Groot, J. F., Fuller, G., Kumar, A. J., Piao, Y., Eterovic, K., Ji, Y., et al. (2010). Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro. Oncol. 12, 233–242. doi:10.1093/neuonc/nop027

PubMed Abstract | CrossRef Full Text | Google Scholar

De Sanctis, F., Ugel, S., Facciponte, J., and Facciabene, A. (2018). The dark side of tumor-associated endothelial cells. Semin. Immunol. 35, 35–47. doi:10.1016/j.smim.2018.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Russo, J., Luik, A. L., Song, J., Zhang, X., et al. (2017). Vascular laminins in physiology and pathology. Matrix Biol. 57-58, 140–148. doi:10.1016/j.matbio.2016.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Tomaso, E., Capen, D., Haskell, A., Hart, J., Logie, J. J., Jain, R. K., et al. (2005). Mosaic tumor vessels: cellular basis and ultrastructure of focal regions lacking endothelial cell markers. Cancer Res. 65, 5740–5749. doi:10.1158/0008-5472.CAN-04-4552

PubMed Abstract | CrossRef Full Text | Google Scholar

Di Tomaso, E., Snuderl, M., Kamoun, W. S., Duda, D. G., Auluck, P. K., Fazlollahi, L., et al. (2011). Glioblastoma recurrence after cediranib therapy in patients: lack of “rebound” revascularization as mode of escape. Cancer Res. 71, 19–28. doi:10.1158/0008-5472.CAN-10-2602

PubMed Abstract | CrossRef Full Text | Google Scholar

Dvorak, H. F., Nagy, J. A., Berse, B., Brown, L. F., Yeo, K. T., Yeo, T. K., et al. (1992). Vascular permeability factor, fibrin, and the pathogenesis of tumor stroma formation. Ann. N.Y. Acad. Sci. 667, 101–111. doi:10.1111/j.1749-6632.1992.tb51603.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Dudley, A. C. (2012). Tumor endothelial cells. Cold Sprin Harb Perspect Med. 2 (3), a006536. doi:10.1101/cshperspect.a006536

PubMed Abstract | CrossRef Full Text | Google Scholar

Esser, S., Wolburg, K., Wolbirg, H., Breier, G., Kurzchalia, T., and Risau, W. (1998). Vascular endothelial growth factor induces endothelial fenestrations in vitro. J. Cell Biol. 140, 947–959. doi:10.1083/jcb.140.4.947

PubMed Abstract | CrossRef Full Text | Google Scholar

Facciabene, A., De Sanctis, F., Pierini, S., Reis, E. S., Balint, K., Facciponte, J., et al. (2017). Local endothelial complement activation reverses endothelial quiescence, enabling t-cell homing, and tumor control during T-cell immunotherapy. Oncoimmunology 6, e1326442. doi:10.1080/2162402X.2017.1326442

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, D., Nagy, J. A., Pyne, K., Dvorak, H. F., and Dvorak, A. M. (1999). Pathways of macromolecular extravasation across microvascular endothelium in response to VPF/VEGF and other vasoactive mediators. Microcirculation 6, 23–44. doi:10.1080/mic.6.1.23.44

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, D., Nolan, D., McDonnell, K., Vahdat, L., Benezra, R., Altorki, N., et al. (2009). Bone marrow-derived endothelial progenitor cells contribute to the angiogenic switch in tumor growth and metastatic progression. Biochim. Biophys. Acta 1796, 33–40. doi:10.1016/j.bbcan.2009.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Caballero, M., Sokol, L., Cuypers, A., and Carmeliet, P. (2022). Metabolic reprogramming in tumor endothelial cells. Int. J. Mol. Sci. 23, 11052. doi:10.3390/ijms231911052

PubMed Abstract | CrossRef Full Text | Google Scholar

Geldhof, V., Sokol, L., Amersfoort, J., De Schepper, M., et al. (2002). Single cell atlas identifies lipid-processing and immunomodulatory endothelial cells in healthy and malignant breast. Nat. Commun. 13, 5511. doi:10.1038/s41467-022-33052-y

CrossRef Full Text | Google Scholar

Georganaki, M., van Hooren, L., and Dimberg, A. (2018). Vascular targeting to increase the efficiency of immune checkpoint blockade in Cancer. Front. Immunol. 9, 3081. doi:10.3389/fimmu.2018.03081

PubMed Abstract | CrossRef Full Text | Google Scholar

Goveia, J., Rohlenova, K., Taverna, F., Treps, L., Conradi, L. C., Pircher, A., et al. (2020). An integrated gene expression landscape profiling approach to identify lung tumor endothelial cell heterogeneity and angiogenic candidates. Cancer Cell 37, 421–436. doi:10.1016/j.ccell.2020.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Griffioen, A. W., Damen, C. A., Blijham, G. H., and Groenewegen, G. (1996). Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood 88, 667–673. doi:10.1182/blood.v88.2.667.bloodjournal882667

PubMed Abstract | CrossRef Full Text | Google Scholar

Hida, K., Maishi, N., Akiyama, K., Ohmura-Kakutani, H., Torii, C., Ohga, N., et al. (2017). Tumor endothelial cells with high aldehyde dehydrogenase activity show drug resistance. Cancer Sci. 108, 2195–2203. doi:10.1111/cas.13388

PubMed Abstract | CrossRef Full Text | Google Scholar

Hobson, B., and Denekamp, J. (1984). Endothelial proliferation in tumours and normal tissues: continuous labelling studies. Br. J. Cancer 49, 405–413. doi:10.1038/bjc.1984.66

PubMed Abstract | CrossRef Full Text | Google Scholar

Hong, L., Du, X., Li, W., Mao, Y., Sun, L., and EndMT, L. X. (2018). EndMT: a promising and controversial field. Eur. J. Cell Biol. 97, 493–500. doi:10.1016/j.ejcb.2018.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, Y., Yuan, J., Righi, E., Kamoun, W. S., Ancukiewicz, M., et al. (2012). Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy. Proc. Natl. Acad. Sci. U. S. A. 109, 17561–17566. doi:10.1073/pnas.1215397109

PubMed Abstract | CrossRef Full Text | Google Scholar

Huijbers, E. J. M., Khan, K. A., and Kerbel, R. S., Tumors resurrect an embryonic vascular program to escape immunity. Sci. Immunol., (2022) 7, eabm 6388, doi:10.1126/sciimmunol.abm6388

CrossRef Full Text | Google Scholar

Inai, T. (2004). Inhibition of vascular endothelial growth factor (VEGF) signaling in cancer causes loss of endothelial fenestrations, regression of tumor vessels, and appearance of basement membrane ghosts. Am. J. Pathol. 165, 35–53.

PubMed Abstract | CrossRef Full Text | Google Scholar

Jain, R. K. (2001). Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat. Med. 7, 987–989. doi:10.1038/nm0901-987

PubMed Abstract | CrossRef Full Text | Google Scholar

Jain, R. K., Martin, J. D., and Stylianopoulos, T. (2014). The role of mechanical forces in tumor growth and therapy. Annu. Rev. Biomed. Eng. 16, 321–346. doi:10.1146/annurev-bioeng-071813-105259

PubMed Abstract | CrossRef Full Text | Google Scholar

Jain, R. K., and Munn, L. L. (2000). Leaky vessels? Call Ang 1. Nat. Med. 6, 131–132. doi:10.1038/72212

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalucha, J., de Rooij, L. P. M. H., Goveia, J., Rohlenova, K., Dumas, S. J., Meta, E., et al. (2020). Single-cell transcriptome atlas of murine endothelial cells. Cell 180, 764–779. doi:10.1016/j.cell.2020.01.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Lambrechts, D., Wauters, E., Boeckx, B., Aibar, S., Nittner, D., Burton, O., et al. (2018). Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289. doi:10.1038/s41591-018-0096-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, S., Meng, W., Guan, Z., Guo, Y., and Han, X. (2016). The hypoxia-related signaling pathways of vasculogenic mimicry in tumor treatment. Biomed. Pharm. 80, 127–135. doi:10.1016/j.biopha.2016.03.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Maishi, N., Annan, D. A., Kikuchi, H., Hida, Y., and Hida, K. (2019). Tumor endothelial heterogeneity in cancer progression. Cancers 11, 1511. doi:10.3390/cancers11101511

PubMed Abstract | CrossRef Full Text | Google Scholar

Maishi, N., Ohba, Y., Akiyama, K., Ohga, N., Hamada, J. I., Nagao-Kitamoto, H., et al. (2016). Tumour endothelial cells in high metastatic tumours promote metastasis via epigenetic dysregulation of biglycan. Sci. Rep. 6, 28039–28113. doi:10.1038/srep28039

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsuda, K., Ohga, N., Hida, Y., Muraki, C., Tsuchiya, K., Kurosu, T., et al. (2010). Isolated tumor endothelial cells maintain specific character during long-term culture. Biochem. Biophys. Res. Commun. 394, 947–954. doi:10.1016/j.bbrc.2010.03.089

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazzone, M., Dettori, D., de Oliveira, R. L., Loges, S., Schmidt, T., Jonckx, B., et al. (2009). Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits metastasis via endothelial normalization. Cell 136, 839–851. doi:10.1016/j.cell.2009.01.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Mc Donald., D. M., and Baluk, P. (2002). Significance of blood vessel leakiness in cancer. Cancer Res. 62, 5381–5385.

PubMed Abstract | Google Scholar

Mc Donald, D. M., and Choyke, P. L. (2003). Imaging of angiogenesis: from microscope to clinic. Nat. Med. 9, 713–725. doi:10.1038/nm0603-713

PubMed Abstract | CrossRef Full Text | Google Scholar

Medici, D., and Kalluri, R. (2012). Endothelial–mesenchymal transition and its contribution to the emergence of stem cell phenotype. Semin. Cancer Biol. 22, 379–384. doi:10.1016/j.semcancer.2012.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Morikawa, S., Kaidoh, T., Haskell, A., and McDonald, D. M. (2002). Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 160, 985–1000. doi:10.1016/S0002-9440(10)64920-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Mulligan, J. K., and Young, M. R. (2010). Tumors induce the formation of suppressor endothelial cells in vivo. Cancer Immunol. Immunother. 59, 267–277. doi:10.1007/s00262-009-0747-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagy, J. A., and Dvorak, H. F. (2012). Heterogeneity of the tumor vasculature: the need for new tumor blood vessel type-specific targets. Clin. Exp. Metastasis 29, 657–662. doi:10.1007/s10585-012-9500-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Nie, L., Lyros, O., Medda, R., Jovanovic, N., Schmidt, J. L., Otterson, M. F., et al. (2014). Endothelial-mesenchymal transition in normal human esophageal endothelial cells cocultured with esophageal adenocarcinoma cells: role of IL-1β and TGF-β2. Am. J. Physiol. Cell Physiol. 307, C859–C877. doi:10.1152/ajpcell.00081.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

Nowak-Sliwinska, P., van Beijnum, J. R., Griffioen, C. J., Huinen, Z. R., Sopesens, N. G., Schulz, R., et al. (2023). Proinflammatory activity of VEGF-targeted treatment through reversal of tumor endothelial cell anergy. Angiogenesis 26, 279–293. doi:10.1007/s10456-022-09863-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohga, N., Akiyama, K., and Hida, Y. (2012). Heterogeneity of tumor endothelial cells: comparison between tumor endothelial cells isolated from high-and low-metastatic tumors. Am. J. Pathol. 180, 1294–1307. doi:10.1016/j.ajpath.2011.11.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohmura-Kakutani, H., Akiyama, K., Maishi, N., Ohga, N., Hida, Y., Kawamoto, T., et al. (2014). Identification of tumor endothelial cells with high aldehyde dehydrogenase activity and a highly angiogenic phenotype. PloS one 9, e113910–e113917. doi:10.1371/journal.pone.0113910

PubMed Abstract | CrossRef Full Text | Google Scholar

Osawa, T., Ohga, N., Akiyama, K., Hida, Y., Kitayama, K., Kawamoto, T., et al. (2013). Lysyl oxidase secreted by tumour endothelial cells promotes angiogenesis and metastasis. Brit. J. Cancer 109, 2237–2247. doi:10.1038/bjc.2013.535

PubMed Abstract | CrossRef Full Text | Google Scholar

Pasut, A., Becker, L. M., Cuypers, A., and Carmeliet, P. (2021). Endothelial cell plasticity at the single-cell level. Angiogenesis 24, 311–326. doi:10.1007/s10456-021-09797-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Rafii, S., Lyden, D., Benezra, R., Hattori, K., and Heissig, B. (2002). Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat. Rev. Cancer 2, 826–835. doi:10.1038/nrc925

PubMed Abstract | CrossRef Full Text | Google Scholar

Reymond, N., and d'Água, B. B. (2013). Crossing the endothelial barrier during metastasis. Nat. Rev. Cancer 13, 858–870. doi:10.1038/nrc3628

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D. (2022). Epithelial-endothelial transition and endothelial-mesenchymal transition. Int. J. Dev. Biol. 66, 311–316. doi:10.1387/ijdb.210234dr

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D. (2023a). Liver angiocrine factors. Tissue and Cell 81, 102027. doi:10.1016/j.tice.2023.102027

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D., and d’Amati, A. (2023c). Bone angiocrine factors. Front. Cell. Dev. Biol. 11, 1244372. doi:10.3389/fcell.2023.1244372

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D., Ligresti, G., and Nicosia, R. F. (2023b). Kidney endothelial cell heterogeneity, angiocrine activity and paracrine regulatory mechanisms. Vasc. Pharmacol. 148, 107139. doi:10.1016/j.vph.2022.107139

CrossRef Full Text | Google Scholar

Ribatti, D., Nico, B., and Crivellato, E. (2009). Morphological and molecular aspects of physiological vascular morphogenesis. Angiogenesis 12, 101–111. doi:10.1007/s10456-008-9125-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D., Nico, B., Vacca, A., Roncali, L., and Dammacco, F. (2002). Endothelial cell heterogeneity and organ specificity. J. Hematother. Stem Cell Res. 11, 81–90. doi:10.1089/152581602753448559

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D., and Pezzella, F. (2021). Overview on the different patterns of tumor vascularization. Cells 10, 639. doi:10.3390/cells10030639

PubMed Abstract | CrossRef Full Text | Google Scholar

Ribatti, D., and Tamma, R. (2018). A revisited concept. Tumors: wounds that do not heal. Crit. Rev. Oncol. Hematol. 128, 65–69. doi:10.1016/j.critrevonc.2018.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Rohlenova, K., Goveia, J., García-Caballero, M., Subramanian, A., Kalucka, J., Treps, L., et al. (2020). Single-cell RNA sequencing maps endothelial metabolic plasticity in pathological angiogenesis. Cell Metab. 31, 862–877. doi:10.1016/j.cmet.2020.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwartz, S. M., and Benditt, E. P. (1973). Cell replication in the aortic endothelium: a new method for study of the problem. Lab. Invest. 28, 699–707.

PubMed Abstract | Google Scholar

Seaman, S., Stevens, J., Yang, M. Y., Logsdon, D., and St Croix, B. (2007). Genes that distinguish physiological and pathological angiogenesis. Cancer Cell 11, 539–554. doi:10.1016/j.ccr.2007.04.017

PubMed Abstract | CrossRef Full Text | Google Scholar

St Croix, B., Rago, C., Velculescu, V., Traverso, G., Romans, K. E., Montgomery, E., et al. (2000). Genes expressed in human tumor endothelium. Science 289, 1197–1202. doi:10.1126/science.289.5482.1197

PubMed Abstract | CrossRef Full Text | Google Scholar

Straume, O., Chappuis, P. O., Salvesen, H. B., Halvorsen, O. J., Haukaas, S. A., Goffin, J. R., et al. (2002). Prognostic importance of glomeruloid microvascular proliferation indicates an aggressive angiogenic phenotype in human cancers. Cancer Res. 62, 6808–6811.

PubMed Abstract | Google Scholar

Tsuchiya, K., Hida, K., Hida, Y., Ohga, N., Muraki, C., Akino, T., et al. (2010). Adrenomedullin antagonist suppresses tumor formation in renal cell carcinoma through inhibitory effects on tumor endothelial cells and endothelial progenitor mobilization. Int. J. Oncol. 36, 1379–1386. doi:10.3892/ijo_00000622

PubMed Abstract | CrossRef Full Text | Google Scholar

Tyrakis, P. A., Palazon, A., Macias, D., Lee, K. L., Phan, A. T., Veliça, P., et al. (2016). S-2-hydroxyglutarate regulates CD8+ T-lymphocyte fate. Nature 540, 236–241. doi:10.1038/nature20165

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Q., He, Z., Huang, M., Liu, T., Wang, Y., Xu, H., et al. (2018). Vascular niche IL-6 induces alternative macrophage activation in glioblastoma through HIF-2α. Nat. Commun. 9, 559. doi:10.1038/s41467-018-03050-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Wettschurek, N., Strilic, B., and Offermanns, S. (2019). Passing the vascular barrier: endothelial signaling processes controlling extravasation. Physiol. Rev. 99, 1467–1525. doi:10.1152/physrev.00037.2018

PubMed Abstract | CrossRef Full Text | Google Scholar

Winkler, F., Kozin, S. V., Tong, R. T., Chae, S. S., Booth, M. F., Garkavtsev, I., et al. (2004). Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 6, 553–563. doi:10.1016/j.ccr.2004.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiang, Y. Q., Sun, H. C., Zhang, W., Zhu, X. D., Zhuang, P. Y., Zhang, J. B., et al. (2009). Human hepatocellular carcinoma tumor-derived endothelial cells manifest increased angiogenesis capability and drug resistance compared with normal endothelial cells. Clin. Cancer Res. 15, 4838–4846. doi:10.1158/1078-0432.CCR-08-2780

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, K., Ohga, N., Hida, Y., Maishi, N., Kawamoto, T., Kitayama, K., et al. (2012). Biglycan is a specific marker and an autocrine angiogenic factor of tumour endothelial cells. Brit. J. Cancer 106, 1214–1223. doi:10.1038/bjc.2012.59

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: angiocrine factors, angiogenesis, endothelial cells, tumor growth, resistance

Citation: Ribatti D (2024) Aberrant tumor vasculature. Facts and pitfalls. Front. Pharmacol. 15:1384721. doi: 10.3389/fphar.2024.1384721

Received: 10 February 2024; Accepted: 11 March 2024;
Published: 21 March 2024.

Edited by:

Simona Federica Spampinato, University of Turin, Italy

Reviewed by:

Gabriela D’Amico, Queen Mary University of London, United Kingdom
Raffaella Giavazzi, Mario Negri Institute for Pharmacological Research (IRCCS), Italy

Copyright © 2024 Ribatti. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Domenico Ribatti, domenico.ribatti@uniba.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.