Category: Family

Angiogenesis and cancer

Angiogenesis and cancer

Anx entire process is carefully regulated Sports nutrition proteins Antiogenesis can tip the cancef in Angiogenesid way; either activating or Sports nutrition angiogenesis. Enhancing mental clarity experiment was performed again, the treatment cander was Meal planning for the whole family. into sensitive Angiogenesis and cancer insensitive Angilgenesis according to their initially Sports nutrition cut-off values, and histopathological analyses were performed. Xu J et al Vascular CXCR expression promotes vessel sprouting and sensitivity to sorafenib treatment in hepatocellular carcinoma. Tumor progression is often accompanied by ingrowth of blood vessels, consistent with a need for malignant cells to have access to the circulation system to thrive. In melanoma cells, both IL-1α and IL-1β can promote tumor angiogenesis by activating NF-κB signaling pathways to induce the expression of IL-6, IL-8, intercellular adhesion molecule-1, and tissue factor [ ].

Angiogenesis and cancer -

Recruitment of stromal cells, immune cells and progenitors is another potential mechanism for resistance to anti-angiogenic therapy. In particular, many studies have pointed out an important role of bone marrow derived cells BMDCs in this aspect.

Recruitment of BMDCs in glioblastoma can cause resistance to vatalanib treatment and the depletion of BMDCs can potentiate the effects of this anti-angiogenic drug [ ]. Release of proangiogenic factors and increased hypoxia in response to vascularization blockade can lead to recruitment of endothelial progenitor cells EPC from the bone marrow, which contribute to tumor vascularization and have been linked to development of resistance to anti-VEGF therapy [ ].

Moreover, recruitment of pro-angiogenic myeloid cells is also considered to be a mechanism whereby tumors bypass the inhibitory effects of anti-angiogenics drugs. Tumors can recruit different populations of myeloid cells with pro-angiogenic properties which in turn can be used as an alternative source of pro-angiogenic chemokines and cytokines [ ].

In addition, alternative pro-angiogenic signaling pathways including ANGPT-2, FGF-2, IL-8 can be induced by tumor cells in response to a pharmacological inhibition of the VEGF signaling pathway [ ].

In recent years, progress has been made towards understanding the mechanism of action of anti-angiogenic drugs through evaluating the effects of anti-angiogenic inhibitors on tumor vessels in preclinical and clinical studies.

An important aspect that have emerged is the broad spectrum of effects covered by the angiogenic inhibitors and the diversity in terms of therapeutic response [ ].

Although anti-angiogenic drugs were initially designed to block blood vessel formation, their ability to control tumor growth may be due to several different mechanism, which are not mutually exclusive.

To improve vascular targeting, a thorough understanding of the cellular and molecular mechanisms that hinder tumor progression in response to anti-angiogenic therapy in specific tumors is necessary. The possible mechanism of actions of angiogenesis inhibitors on tumor blood vessels can be broadly classified into three categories: a vessel depletion, b vessel normalization, and c immune activation Fig.

Effects of anti-angiogenic therapy. The mechanism of actions of angiogenesis inhibitors on tumor blood vessels can be classified into three categories: a vessel depletion, b vessel normalization, and c immune activation.

a Vessel depletion result in tumor cell starvation and an increased tissue hypoxia. Enhanced hypoxia may promote the recruitment of pro-angiogenic myeloid cells and the mobilization of tumor cell from the hypoxic tissue to the normal tissue as well as co-option of normal vessels.

In addition, the depletion of tumor vessels results in an inefficient delivery of anti-cancer drugs. b Normalization of tumor blood vessels achieved by restored endothelial cell junctions, increased pericytes coverage and re-established blood flow result in decrease tissue hypoxia and increased drugs delivery.

In addition, vessel normalization promote the expression of endothelial adhesion molecule facilitating immune cell infiltration. c Immune activation, induced by anti-angiogenic drugs include dendritic cell DC maturation, activation and infiltration of T-cell as well as the polarization of tumor associated macrophages TAM towards an M1-like phenotype.

In addition, a decrease in regulatory T-cells Treg , myeloid derived suppressor cells MDSCs and mast cells have been observed in response to anti-angiogenic therapy. The development of anti-angiogenic drugs was initiated by the hypothesis that starving tumors by blocking angiogenesis would slow tumor progression and improve patient survival [ 1 ].

Early preclinical studies were promising and demonstrated a significant tumor growth delay and reduced metastasis. However, the effects of anti-angiogenic agents administrated as monotherapy in cancer patients during clinical trials often failed to show significant survival benefits.

These observations suggest that anti-angiogenic therapy alone is insufficient to induce substantial tumor shrinkage in most cancer patients. Particular attention must be placed on the effects of tumor vessel depletion on the tumor microenvironment as well as the development of anti-angiogenic resistance.

Indeed, as mentioned above, hypoxia induced by vessel depletion can activate several mechanisms used by tumor cells to counteract the lack of oxygen and nutrients such as increased tumor invasiveness and co-option of normal vessels resulting in ineffective anti-angiogenic therapy.

Several studies demonstrate that before reaching complete depletion of the vascular bed, anti-VEGF drugs induce an early and transient phase in which vessels assume normal shape and function [ , ]. This vessel normalization window is characterized by a rescue of the balance between pro- and anti-angiogenic factors and it can promote increase tumor drug delivery and efficacy.

Despite a high vascular density, tumors are usually hypoxic and nutrient-deprived since the tumor vessels are abnormal, leaky and malfunction. Such abnormal vasculature significantly compromises the efficacy of most anti-cancer therapies by limiting the delivery of drugs as well as promoting resistance to treatment.

The vessel normalization hypothesis, introduced by Rakesh Jain in [ ] suggests that rather than depleting vessels, a sub-maximal doses of anti-angiogenic therapy can restore the normal function and structure of tumor vessels and improve drug delivery. This hypothesis could explain the increased progression-free survival observed in patients treated with anti-angiogenic drugs combined with chemotherapy as compared to treatment with chemotherapy alone [ ].

Evidence supporting the idea that vessel normalization can improve cancer therapy has been obtained in mouse models. These studies show that improving tumor vessel perfusion and oxygenation ameliorates the efficacy of conventional therapies such as radiotherapy, chemotherapy and immunotherapy and reduces metastatic dissemination [ , ].

Evidence that support the notion that vessel normalization occur in response to anti-angiogenic therapy has also been obtained from clinical studies.

The functionality of the tumor vasculature in glioblastoma patients treated with anti-VEGF therapies has been evaluated by magnetic resonance imaging MRI. MRI analysis of patients treated with cediranib revealed a decrease in vessel diameter, vascular permeability, and edema.

More importantly, survival of patients with recurrent glioblastoma following cediranib-treatment was found to correlate with a vascular normalization index [ ]. Improved perfusion occurred only in a subset of glioblastoma patients treated with cediranib, and was associated with improved patient overall survival [ ].

These observations suggest that the degree of vessel normalization in terms of improved perfusion may be used as a tool to distinguish responders to anti-angiogenic therapy from non-responding patients [ , ].

Pro-angiogenic factors in tumors induce down-regulation of adhesion molecules on endothelial cells in the tumor vasculature and induce anergy to inflammatory signals such as TNFα and IL Hereby, tumors with an angiogenic phenotype may escape the infiltration of cytotoxic leukocytes [ ].

Using anti-angiogenic agents can potentially overcome the down-regulation of adhesion molecules and the unresponsiveness to inflammatory signals [ ]. Consistent with this, normalization of tumor vasculature through anti-VEGF therapy in combination with adoptive T-cell transfer was found to increase tumor T-cell infiltration and improve survival in murine melanoma model [ ].

Inhibition of VEGF signaling in the tumor microenvironment may be beneficial not only in terms of improving immune cell recruitment, but can also directly improve immune cell activation.

Normalization of the tumor vascular network and decreased hypoxia can promote T cell infiltration and induce polarization of TAM to an M1-like phenotype [ ]. Anti-angiogenic therapy can also reduce the prevalence of immunosuppressive cells.

Decreases in Treg recruitment as well as MDSC has been reported after sunitinib treatment in tumor-bearing mice and in patients with metastatic renal carcinoma [ , ].

In addition, inhibition of angiogenic signaling may improve T-cell priming and activation by improving dendritic cell DC maturation. Anti-anigogenic therapy using the VEGF-neutralizing antibody bevacizumab was found to increase the number and the maturation of DCs in patients with metastatic non-small cell lung carcinoma [ ].

These observations indicate that immune activation is an additional mechanism that can contribute to response to anti-angiogenic therapy.

Tumor vessels are often dysfunctional and anergic to inflammatory stimuli, leading to a hostile tumor microenvironment that fuel cancer progression and aggravate therapeutic approaches.

Current vascular targeting strategies are based on inhibition of key angiogenic signaling pathways known to promote tumor angiogenesis. Although several anti-angiogenic drugs have been approved, intrinsic and acquired resistance to therapy limit their efficacy.

An increased understanding of tumor vessel phenotype and mechanisms involved in treatment response and resistance to therapy is necessary to overcome the hurdles that prevent successful control of the angiogenic response in tumors. Alternatively, vascular targeting should instead be designed to target the tumor vessels in new ways that are conceptually different from inhibition of angiogenesis.

This may involve altering the timing and dosing of already existing anti-angiogenic therapy in combination with other drugs, or development of novel therapeutics to either directly target the tumor vessels or optimize their function to fit the cancer therapy at hand.

The fact that tumor vessels differ molecularly from their normal counterparts can be used to develop treatment strategies that specifically target malignant cells and tumor vasculature. Therapeutic vaccination strategies to raise endogenous antibodies against antigens specifically expressed by tumor vasculature have shown efficacy in pre-clinical cancer models [ ].

Prophylactic immunization of the alternatively spliced extra domain ED -B of fibronectin efficiently reduced growth of syngeneic subcutaneous tumors [ ], and therapeutic vaccination against ED-A after tumor development reduced metastatic dissemination in the MMTV-PyMT model of metastatic mammary carcinoma [ ].

Antibodies targeting tumor vessel markers have also been used. Conjugating TEM8-targeting antibodies with cytotoxic monomethyl auristatin E was successful in specifically directing the drug to the tumor microenvironment of orthotopic tumors and patient derived xenografts, significantly inducing regression or eradication of tumor growth in pre-clinical models [ ].

Using an alternative strategy, targeting tumor endothelium and TEM8-positive malignant cells by employing TEM8-specific CAR T cells was effective in treating triple negative breast cancer TNBC patient derived xenograft PDX models and metastatic TNBC cell-line xenografts [ ].

Peptides that specifically bind tumor endothelial cells have also been used to target either therapeutic antibodies or chemokines to the tumor microenvironment to improve efficacy and decrease toxicity [ , ].

Going beyond anti-angiogenesis and vascular normalization, strategies that can alter vessel phenotype to optimize specific types of cancer therapy are quickly emerging. gov [ , ]. To provide an even more efficient gateway for T-cells to enter the tumor microenvironment, tumor vessels can be induced to differentiate to high-endothelial venules HEV.

HEV have a distinct morphology, built up by cuboidal endothelial cells, and they express chemokine and adhesion molecules that mediate efficient recruitment of lymphocytes into the tissue [ ]. Depletion of Tregs in a model of fibrosarcoma led to HEV neogenesis, enabling recruitment of T-cells into the tumor [ ].

The presence of HEV within the tumor was a pre-requisite for tumor control after Treg depletion. Consistent with a role of activated T-cells in HEV neogenesis, combining anti-angiogenic therapy with anti-PD-L1 immunotherapy was sufficient to induce HEVs in several orthotopic and genetically engineered mouse models of cancer, stimulating tumor immunity [ ].

With respect to brain tumors, strategies that transiently open the blood brain barrier to enable delivery of drugs are of considerable interest [ ]. The observation that paracrine signaling in WNT-medulloblastoma was associated with fenestrated tumor vessels that lack ABC transporters suggests that brain tumor vessels can indeed be modulated to allow a better penetration of drugs [ ].

This exciting possibility has yet to be explored therapeutically. It is necessary to gain a deeper understanding of how tumor vessel function is altered in specific cancer types, and how vessel phenotype can be modulated. This may lead to new vascular targeting strategies aimed at tailoring vessel function to optimize drug response.

Folkman J Tumor angiogenesis: therapeutic implications. N Engl J Med 21 — CAS PubMed Google Scholar. Folkman J et al Isolation of a tumor factor responsible for angiogenesis. J Exp Med 2 — CAS PubMed PubMed Central Google Scholar.

Cao Y et al Forty-year journey of angiogenesis translational research. Sci Transl Med 3 rv3. PubMed PubMed Central Google Scholar. Hurwitz H et al Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer.

N Engl J Med 23 — Kuczynski EA et al Vessel co-option in cancer. Nat Rev Clin Oncol 16 8 — Hanahan D, Folkman J Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 86 3 — Nature — Nowak-Sliwinska P et al Consensus guidelines for the use and interpretation of angiogenesis assays.

Angiogenesis 21 3 — Jakobsson L, Bentley K, Gerhardt H VEGFRs and Notch: a dynamic collaboration in vascular patterning. Biochem Soc Trans 37 Pt 6 — Tammela T et al Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation.

Strasser GA, Kaminker JS, Tessier-Lavigne M Microarray analysis of retinal endothelial tip cells identifies CXCR11 as a mediator of tip cell morphology and branching. Blood 24 — Shawber CJ et al Notch alters VEGF responsiveness in human and murine endothelial cells by direct regulation of VEGFR-3 expression.

J Clin Invest 11 — Jakobsson L et al Endothelial cells dynamically compete for the tip cell position during angiogenic sprouting. Nat Cell Biol 12 10 — Hellstrom M et al Dll4 signalling through Notch1 regulates formation of tip cells during angiogenesis.

PubMed Google Scholar. Lobov IB et al Delta-like ligand 4 Dll4 is induced by VEGF as a negative regulator of angiogenic sprouting. Proc Natl Acad Sci USA 9 — Harrington LS et al Regulation of multiple angiogenic pathways by Dll4 and Notch in human umbilical vein endothelial cells.

Microvasc Res 75 2 — Funahashi Y et al Notch regulates the angiogenic response via induction of VEGFR J Angiogenes Res 2 1 Gerhardt H et al VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J Cell Biol 6 — Fantin A et al NRP1 acts cell autonomously in endothelium to promote tip cell function during sprouting angiogenesis.

Blood 12 — Segarra M et al Semaphorin 6A regulates angiogenesis by modulating VEGF signaling. Blood 19 — Phng LK et al Nrarp coordinates endothelial Notch and Wnt signaling to control vessel density in angiogenesis.

Dev Cell 16 1 — Herwig L et al Distinct cellular mechanisms of blood vessel fusion in the zebrafish embryo.

Curr Biol 21 22 — Kochhan E et al Blood flow changes coincide with cellular rearrangements during blood vessel pruning in zebrafish embryos.

PLoS One 8 10 :e Lenard A et al Endothelial cell self-fusion during vascular pruning. PLoS Biol 13 4 :e Lenard A et al In vivo analysis reveals a highly stereotypic morphogenetic pathway of vascular anastomosis. Dev Cell 25 5 — Phng LK, Stanchi F, Gerhardt H Filopodia are dispensable for endothelial tip cell guidance.

Development 19 — Blum Y et al Complex cell rearrangements during intersegmental vessel sprouting and vessel fusion in the zebrafish embryo. Dev Biol 2 — Betz C et al Cell behaviors and dynamics during angiogenesis. Development 13 — Patan S et al Intussusceptive microvascular growth: a common alternative to capillary sprouting.

Arch Histol Cytol 55 Suppl — Burri PH, Tarek MR A novel mechanism of capillary growth in the rat pulmonary microcirculation. Anat Rec 1 — Hellstrom M et al Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse.

Development 14 — Wilting J et al VEGF induces proliferation of vascular endothelial cells and expression of flk-1 without affecting lymphatic vessels of chorioallantoic membrane.

Dev Biol 1 — Crivellato E et al Recombinant human erythropoietin induces intussusceptive microvascular growth in vivo. Leukemia 18 2 — Ribatti D et al Microvascular density, vascular endothelial growth factor immunoreactivity in tumor cells, vessel diameter and intussusceptive microvascular growth in primary melanoma.

Oncol Rep 14 1 — Nico B et al Intussusceptive microvascular growth in human glioma. Clin Exp Med 10 2 — Patan S, Munn LL, Jain RK Intussusceptive microvascular growth in a human colon adenocarcinoma xenograft: a novel mechanism of tumor angiogenesis.

Microvasc Res 51 2 — Djonov V et al MMP cellular localization of a novel metalloproteinase within normal breast tissue and mammary gland tumours. J Pathol 2 — Risau W et al Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies.

Development 3 — Risau W, Lemmon V Changes in the vascular extracellular matrix during embryonic vasculogenesis and angiogenesis. Choi K Hemangioblast development and regulation. Biochem Cell Biol 76 6 — Asahara T et al Isolation of putative progenitor endothelial cells for angiogenesis.

Science — Bussolati B, Grange C, Camussi G Tumor exploits alternative strategies to achieve vascularization. FASEB J 25 9 — Kioi M et al Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice.

J Clin Invest 3 — Ahn JB et al Circulating endothelial progenitor cells EPC for tumor vasculogenesis in gastric cancer patients. Cancer Lett 1 — Greenfield JP, Cobb WS, Lyden D Resisting arrest: a switch from angiogenesis to vasculogenesis in recurrent malignant gliomas.

Chopra H et al Insights into endothelial progenitor cells: origin, classification, potentials, and prospects. Stem Cells Int Schmidt A, Brixius K, Bloch W Endothelial precursor cell migration during vasculogenesis. Circ Res 2 — Circ Res 97 4 — Shin JW et al Isolation of endothelial progenitor cells from cord blood and induction of differentiation by ex vivo expansion.

Yonsei Med J 46 2 — Urbich C, Dimmeler S Endothelial progenitor cells: characterization and role in vascular biology. Circ Res 95 4 — Reale A et al Functional and biological role of endothelial precursor cells in tumour progression: a new potential therapeutic target in haematological malignancies.

Asahara T et al VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J 18 14 — Hattori K et al Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells.

J Exp Med 9 — Kopp HG, Ramos CA, Rafii S Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue.

Curr Opin Hematol 13 3 — Chang EI et al Hypoxia, hormones, and endothelial progenitor cells in hemangioma. Lymphat Res Biol 5 4 — Spring H et al Chemokines direct endothelial progenitors into tumor neovessels.

Proc Natl Acad Sci USA 50 — FEBS Lett 15 — Maniotis AJ et al Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry.

Am J Pathol 3 — Ricci-Vitiani L et al Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Upile T et al Vascular mimicry in cultured head and neck tumour cell lines. Head Neck Oncol Williamson SC et al Vasculogenic mimicry in small cell lung cancer.

Nat Commun Baeten CI et al Prognostic role of vasculogenic mimicry in colorectal cancer. Dis Colon Rectum 52 12 — Sharma N et al Prostatic tumor cell plasticity involves cooperative interactions of distinct phenotypic subpopulations: role in vasculogenic mimicry.

Prostate 50 3 — Fausto N Vasculogenic mimicry in tumors. Fact or artifact? Am J Pathol 2 Seftor RE et al Tumor cell vasculogenic mimicry: from controversy to therapeutic promise. Am J Pathol 4 — Folberg R, Maniotis AJ Vasculogenic mimicry.

APMIS 7—8 — Angara K, Borin TF, Arbab AS Vascular mimicry: a novel neovascularization mechanism driving anti-angiogenic therapy AAT resistance in glioblastoma. Transl Oncol 10 4 — Valyi-Nagy K et al Stem cell marker CD is expressed by vasculogenic mimicry-forming uveal melanoma cells in three-dimensional cultures.

Mol Vis — Lin AY et al Distinguishing fibrovascular septa from vasculogenic mimicry patterns. Arch Pathol Lab Med 7 — Comito G et al HIF-1alpha stabilization by mitochondrial ROS promotes Met-dependent invasive growth and vasculogenic mimicry in melanoma cells.

Free Radic Biol Med 51 4 — Angara K et al Vascular mimicry in glioblastoma following anti-angiogenic and antiHETE therapies. Histol Histopathol 32 9 — Li M et al Vasculogenic mimicry: a new prognostic sign of gastric adenocarcinoma.

Pathol Oncol Res 16 2 — Wang R et al Glioblastoma stem-like cells give rise to tumour endothelium. Mei X et al Glioblastoma stem cell differentiation into endothelial cells evidenced through live-cell imaging.

Neuro Oncol 19 8 — J Cell Mol Med 13 2 — Alvero AB et al Stem-like ovarian cancer cells can serve as tumor vascular progenitors. Stem Cells 27 10 — Zhao Y et al Endothelial cell transdifferentiation of human glioma stem progenitor cells in vitro.

Brain Res Bull 82 5—6 — Kulla A et al Analysis of the TP53 gene in laser-microdissected glioblastoma vasculature.

Acta Neuropathol 4 — Rodriguez FJ et al Neoplastic cells are a rare component in human glioblastoma microvasculature. Oncotarget 3 1 — De Palma M et al Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors.

Cancer Cell 8 3 — Cheng L et al Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth.

Cell 1 — Baluk P, Hashizume H, McDonald DM Cellular abnormalities of blood vessels as targets in cancer. Curr Opin Genet Dev 15 1 — McDonald DM, Baluk P Imaging of angiogenesis in inflamed airways and tumors: newly formed blood vessels are not alike and may be wildly abnormal: Parker B.

Francis lecture. Chest 6 Suppl S—S. Kimura H et al Fluctuations in red cell flux in tumor microvessels can lead to transient hypoxia and reoxygenation in tumor parenchyma.

Cancer Res 56 23 — Bennewith KL, Durand RE Quantifying transient hypoxia in human tumor xenografts by flow cytometry. Cancer Res 64 17 — Hashizume H et al Openings between defective endothelial cells explain tumor vessel leakiness.

Padera TP et al Pathology: cancer cells compress intratumour vessels. Nature Abramsson A et al Analysis of mural cell recruitment to tumor vessels. Circulation 1 — Morikawa S et al Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors.

Baluk P et al Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol 5 — St Croix B et al Genes expressed in human tumor endothelium.

Zhang L et al Tumor-derived vascular endothelial growth factor up-regulates angiopoietin-2 in host endothelium and destabilizes host vasculature, supporting angiogenesis in ovarian cancer. Cancer Res 63 12 — Carson-Walter EB et al Cell surface tumor endothelial markers are conserved in mice and humans.

Cancer Res 61 18 — Huang X et al Lymphoma endothelium preferentially expresses Tim-3 and facilitates the progression of lymphoma by mediating immune evasion. J Exp Med 3 — Dieterich LC et al Transcriptional profiling of human glioblastoma vessels indicates a key role of VEGF-A and TGFbeta2 in vascular abnormalization.

J Pathol 3 — Roudnicky F et al Endocan is upregulated on tumor vessels in invasive bladder cancer where it mediates VEGF-A-induced angiogenesis. Cancer Res 73 3 — Zhao Q et al Single-cell transcriptome analyses reveal endothelial cell heterogeneity in tumors and changes following antiangiogenic treatment.

Cancer Res 78 9 — Buckanovich RJ et al Tumor vascular proteins as biomarkers in ovarian cancer. J Clin Oncol 25 7 — Zhang L et al IDH mutation status is associated with distinct vascular gene expression signatures in lower-grade gliomas.

Neuro Oncol 20 11 — Masiero M et al A core human primary tumor angiogenesis signature identifies the endothelial orphan receptor ELTD1 as a key regulator of angiogenesis.

Cancer Cell 24 2 — Hanly AM, Winter DC The role of thrombomodulin in malignancy. Semin Thromb Hemost 33 7 — Maruno M et al Expression of thrombomodulin in astrocytomas of various malignancy and in gliotic and normal brains.

J Neurooncol 19 2 — Mura M et al Identification and angiogenic role of the novel tumor endothelial marker CLEC14A. Oncogene 31 3 — Langenkamp E et al Elevated expression of the C-type lectin CD93 in the glioblastoma vasculature regulates cytoskeletal rearrangements that enhance vessel function and reduce host survival.

Cancer Res 75 21 — Lugano R et al CD93 promotes beta1 integrin activation and fibronectin fibrillogenesis during tumor angiogenesis.

J Clin Invest 8 — Christian S et al Endosialin Tem1 is a marker of tumor-associated myofibroblasts and tumor vessel-associated mural cells. Am J Pathol 2 — Khan KA et al Multimerin-2 is a ligand for group 14 family C-type lectins CLEC14A, CD93 and CD spanning the endothelial pericyte interface.

Oncogene 36 44 — Galvagni F et al Dissecting the CDMultimerin 2 interaction involved in cell adhesion and migration of the activated endothelium. Matrix Biol — Mogler C et al Hepatic stellate cell-expressed endosialin balances fibrogenesis and hepatocyte proliferation during liver damage.

EMBO Mol Med 7 3 — Viski C et al Endosialin-expressing pericytes promote metastatic dissemination. Cancer Res 76 18 — Griffioen AW et al Tumor angiogenesis is accompanied by a decreased inflammatory response of tumor-associated endothelium. Blood 88 2 — Griffioen AW et al Endothelial intercellular adhesion molecule-1 expression is suppressed in human malignancies: the role of angiogenic factors.

Cancer Res 56 5 — Dirkx AE et al Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression.

Cancer Res 63 9 — Huang H et al VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NF-kappaB-induced endothelial activation. FASEB J 29 1 — Motz GT et al Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors.

Nat Med 20 6 — Buckanovich RJ et al Endothelin B receptor mediates the endothelial barrier to T cell homing to tumors and disables immune therapy. Nat Med 14 1 — Phoenix TN et al Medulloblastoma genotype dictates blood brain barrier phenotype.

Cancer Cell 29 4 — Ferrara N, Gerber HP, LeCouter J The biology of VEGF and its receptors. Nat Med 9 6 — Apte RS, Chen DS, Ferrara N VEGF in signaling and disease: beyond discovery and development. Cell 6 — Ferrara N Vascular endothelial growth factor: basic science and clinical progress.

Endocr Rev 25 4 — Claesson-Welsh L, Welsh M VEGFA and tumour angiogenesis. J Intern Med 2 — EMBO J 20 11 — Adv Cancer Res — Lamalice L, Le Boeuf F, Huot J Endothelial cell migration during angiogenesis.

Circ Res 6 — van Hinsbergh VW, Koolwijk P Endothelial sprouting and angiogenesis: matrix metalloproteinases in the lead.

Cardiovasc Res 78 2 — Weis SM, Cheresh DA Pathophysiological consequences of VEGF-induced vascular permeability. Azzi S, Hebda JK, Gavard J Vascular permeability and drug delivery in cancers.

Front Oncol Weis S et al Endothelial barrier disruption by VEGF-mediated Src activity potentiates tumor cell extravasation and metastasis. J Cell Biol 2 — Hofer E, Schweighofer B Signal transduction induced in endothelial cells by growth factor receptors involved in angiogenesis.

Thromb Haemost 97 3 — Autiero M et al Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1.

Nat Med 9 7 — Schomber T et al Placental growth factor-1 attenuates vascular endothelial growth factor-A-dependent tumor angiogenesis during beta cell carcinogenesis. Cancer Res 67 22 — Fischer C et al Anti-PlGF inhibits growth of VEGF R -inhibitor-resistant tumors without affecting healthy vessels.

Cell 3 — Bais C et al PlGF blockade does not inhibit angiogenesis during primary tumor growth. Turner N, Grose R Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 10 2 — Ornitz DM, Itoh N The fibroblast growth factor signaling pathway.

Wiley Interdiscip Rev Dev Biol 4 3 — Cytokine Growth Factor Rev 16 2 — Compagni A et al Fibroblast growth factors are required for efficient tumor angiogenesis. Cancer Res 60 24 — Yu P et al FGF-dependent metabolic control of vascular development.

Incio J et al Obesity promotes resistance to anti-VEGF therapy in breast cancer by up-regulating IL-6 and potentially FGF Sci Transl Med 10 :eaag Heldin CH, Westermark B Mechanism of action and in vivo role of platelet-derived growth factor.

Physiol Rev 79 4 — Franco M et al Pericytes promote endothelial cell survival through induction of autocrine VEGF-A signaling and Bcl-w expression.

Blood 10 — Betsholtz C Insight into the physiological functions of PDGF through genetic studies in mice. Cytokine Growth Factor Rev 15 4 — Guo P et al Platelet-derived growth factor-B enhances glioma angiogenesis by stimulating vascular endothelial growth factor expression in tumor endothelia and by promoting pericyte recruitment.

Davis S et al Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning.

Cell 87 7 — Maisonpierre PC et al Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Kiss EA, Saharinen P Anti-angiogenic targets: angiopoietin and angiopoietin-receptors.

In: Marmé D ed Tumor angiogenesis: a key target for cancer therapy. Springer, Cham, pp 1— Google Scholar. Reiss Y et al Switching of vascular phenotypes within a murine breast cancer model induced by angiopoietin J Pathol 4 — Shim WS, Ho IA, Wong PE Angiopoietin: a TIE d balance in tumor angiogenesis.

Mol Cancer Res 5 7 — Fiedler U et al Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med 12 2 — Chae SS et al Angiopoietin-2 interferes with anti-VEGFR2-induced vessel normalization and survival benefit in mice bearing gliomas.

Clin Cancer Res 16 14 — Peterson TE et al Dual inhibition of Ang-2 and VEGF receptors normalizes tumor vasculature and prolongs survival in glioblastoma by altering macrophages. Proc Natl Acad Sci USA 16 — Wu FT et al Efficacy of cotargeting angiopoietin-2 and the VEGF pathway in the adjuvant postsurgical setting for early breast, colorectal, and renal cancers.

Cancer Res 76 23 — Lisle JE et al Eph receptors and their ligands: promising molecular biomarkers and therapeutic targets in prostate cancer. Biochim Biophys Acta 2 — Kullander K, Klein R Mechanisms and functions of Eph and ephrin signalling.

Nat Rev Mol Cell Biol 3 7 — Holder N, Klein R Eph receptors and ephrins: effectors of morphogenesis. Development 10 — Adams RH, Klein R Eph receptors and ephrin ligands.

Essential mediators of vascular development. Trends Cardiovasc Med 10 5 — Surawska H, Ma PC, Salgia R The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 15 6 — Dodelet VC, Pasquale EB Eph receptors and ephrin ligands: embryogenesis to tumorigenesis.

Oncogene 19 49 — Dong Y et al Downregulation of EphA1 in colorectal carcinomas correlates with invasion and metastasis.

Mod Pathol 22 1 — Hafner C et al Loss of EphB6 expression in metastatic melanoma. Int J Oncol 23 6 — Ogawa K et al The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19 52 — Dobrzanski P et al Antiangiogenic and antitumor efficacy of EphA2 receptor antagonist.

Cancer Res 64 3 — Brantley DM et al Soluble Eph A receptors inhibit tumor angiogenesis and progression in vivo. Oncogene 21 46 — Cheng N et al Inhibition of VEGF-dependent multistage carcinogenesis by soluble EphA receptors.

Neoplasia 5 5 — Noren NK et al Interplay between EphB4 on tumor cells and vascular ephrin-B2 regulates tumor growth. Proc Natl Acad Sci USA 15 — Uhl C et al EphB4 mediates resistance to antiangiogenic therapy in experimental glioma.

Angiogenesis 21 4 — Krusche B et al EphrinB2 drives perivascular invasion and proliferation of glioblastoma stem-like cells. Elife 5:e Wang Y et al Ephrin-B2 controls VEGF-induced angiogenesis and lymphangiogenesis. Sawamiphak S et al Ephrin-B2 regulates VEGFR2 function in developmental and tumour angiogenesis.

Tatemoto K et al Isolation and characterization of a novel endogenous peptide ligand for the human APJ receptor. Biochem Biophys Res Commun 2 — Devic E et al Expression of a new G protein-coupled receptor X-msr is associated with an endothelial lineage in Xenopus laevis.

Mech Dev 59 2 — Cox CM et al Apelin, the ligand for the endothelial G-protein-coupled receptor, APJ, is a potent angiogenic factor required for normal vascular development of the frog embryo.

Kalin RE et al Paracrine and autocrine mechanisms of apelin signaling govern embryonic and tumor angiogenesis. Wysocka MB, Pietraszek-Gremplewicz K, Nowak D The role of apelin in cardiovascular diseases, obesity and cancer.

Front Physiol Berta J et al Apelin expression in human non-small cell lung cancer: role in angiogenesis and prognosis.

J Thorac Oncol 5 8 — Tolkach Y et al Apelin and apelin receptor expression in renal cell carcinoma. Br J Cancer 6 — Seaman S et al Genes that distinguish physiological and pathological angiogenesis.

Cancer Cell 11 6 — Feng M et al Tumor apelin, not serum apelin, is associated with the clinical features and prognosis of gastric cancer. BMC Cancer 16 1 Lacquaniti A et al Apelin beyond kidney failure and hyponatremia: a useful biomarker for cancer disease progression evaluation.

Clin Exp Med 15 1 — Heo K et al Hypoxia-induced up-regulation of apelin is associated with a poor prognosis in oral squamous cell carcinoma patients.

Oral Oncol 48 6 — Cancer Lett — Macaluso NJ et al Discovery of a competitive apelin receptor APJ antagonist.

Chem Med Chem 6 6 — Lv D et al PAK1-cofilin phosphorylation mediates human lung adenocarcinoma cells migration induced by apelin Clin Exp Pharmacol Physiol 43 5 — Berta J et al Apelin promotes lymphangiogenesis and lymph node metastasis.

Oncotarget 5 12 — Sorli SC et al Apelin is a potent activator of tumour neoangiogenesis. Oncogene 26 55 — Sorli SC et al Therapeutic potential of interfering with apelin signalling. Drug Discov Today 11 23—24 — Uribesalgo I et al Apelin inhibition prevents resistance and metastasis associated with anti-angiogenic therapy.

EMBO Mol Med 11 8 :e Cancer Res 79 9 — Harford-Wright E et al Pharmacological targeting of apelin impairs glioblastoma growth. Brain 11 — Le Y et al Chemokines and chemokine receptors: their manifold roles in homeostasis and disease.

Cell Mol Immunol 1 2 — Heidemann J et al Angiogenic effects of interleukin 8 CXCL8 in human intestinal microvascular endothelial cells are mediated by CXCR J Biol Chem 10 — Keane MP et al Depletion of CXCR inhibits tumor growth and angiogenesis in a murine model of lung cancer.

J Immunol 5 — Kitadai Y et al Regulation of disease-progression genes in human gastric carcinoma cells by interleukin 8.

Clin Cancer Res 6 7 — Ijichi H et al Inhibiting Cxcr2 disrupts tumor-stromal interactions and improves survival in a mouse model of pancreatic ductal adenocarcinoma.

J Clin Invest 10 — Yang G et al CXCR promotes ovarian cancer growth through dysregulated cell cycle, diminished apoptosis, and enhanced angiogenesis. Clin Cancer Res 16 15 — Smith ML, Olson TS, Ley K CXCR and E-selectin-induced neutrophil arrest during inflammation in vivo.

J Exp Med 7 — Smith DR et al Inhibition of interleukin 8 attenuates angiogenesis in bronchogenic carcinoma. J Exp Med 5 — Theranostics 7 6 — J Immunol 8 — Zhao X et al ELR-CXC chemokine receptor antagonism targets inflammatory responses at multiple levels.

Li A et al Autocrine role of interleukin-8 in induction of endothelial cell proliferation, survival, migration and MMP-2 production and angiogenesis.

Angiogenesis 8 1 — Kobayashi Y The role of chemokines in neutrophil biology. Front Biosci — Sozzani S et al Chemokines as effector and target molecules in vascular biology.

Cardiovasc Res 3 — Xu J et al Vascular CXCR expression promotes vessel sprouting and sensitivity to sorafenib treatment in hepatocellular carcinoma.

Clin Cancer Res 23 15 — Ceradini DJ et al Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF Nat Med 10 8 — Wolf MJ et al Endothelial CCR signaling induced by colon carcinoma cells enables extravasation via the JAK2-Stat5 and p38MAPK pathway.

Depending on the experimental cancer model and the type of the tumor, vasculogenesis contributes to tumor vessel formation processes ranging from 0. As an example, the tumor ecosystem of hematopoietic and lymphoid tissues is more dependent on EPCs.

Besides its role in primary tumor growth, vasculogenesis is also involved in dissemination and metastasis. SDF-1 produced by immune cells might attract EPCs to distant sites and once there spontaneously induce SDF-1 production, generating a gradient of this molecule that will serve as a chemoattractant of tumor cells.

The interaction between SDF-1, secreted by EPCs, and its CXCR4 receptor, mainly expressed by tumor cells, would promote extravasation and development of the pre-metastatic niche Moreover, the activation of MMP9 by EPCs is also related to an increase in tumor cell migration and invasion, confirming the role of vasculogenesis in metastatic niche formation Vasculogenic mimicry refers to the ability of some malignant cells to start the dedifferentiation process to adopt multiple cellular phenotypes, including endothelial-like properties 41 Figure 2 C.

Those cells finally converge in de novo vasculogenic-like networks composed of red blood cells that are able to contribute to circulation In this way, cells undergoing VM are able to reproduce the pattern of an early embryonic vascular plexus, providing the tumor ecosystem with an additional circulatory system independent of angiogenesis.

The process of VM was observed in highly invasive melanoma cells, whose phenotype reverted to an embryonic-like state and increased cell plasticity, including expression of endothelium-associated genes such as Ephrin-A2 and VE-cadherin Release of ECM components, hypoxia, and activation of transmembrane metalloproteinases has been described as VM promoters Although the occurrence of VM is relatively infrequent within tumors, it has been related to aggressive tumors, an increased risk of metastasis and poor prognosis Vessel intussusception or intussusceptive microvascular growth IMG is defined as a developmental intravascular growth mechanism consisting of the splitting of preexisting vessels into two new vascular structures.

This was first described in postnatal remodeling of lung capillaries 46 Figure 2 D. During intussusception, endothelial cell proliferation is not required, which ultimately makes it a rapid process that occurs within hours or minutes if compared with sprouting angiogenesis. Furthermore, IMG does not rely on endothelial cell proliferation, but it is rather a remodeling process of the endothelial cells that happens as a consequence of both their narrowing and volume increase.

IMG is described to occur after sprouting angiogenesis or vasculogenesis, as a mean of expanding the capillary plexus without the need of a high-metabolic demand To reinforce the transendothelial cell bridge, the endothelial bilayer is formed with cell—cell junctions and the interstitial pillar is formed.

Pericytes and other mural cells are recruited to cover the interstitial wall, which is later widened, allowing endothelial cell retraction and the creation of two independent vessels By using this mechanism, a large vessel is able to split into many smaller functional vessels.

Although the precise mechanism underlying IMG is not fully described, alterations in blood flow dynamics, wall stress over pericytes, changes in shear stress on endothelial cells sensed by absence of CD31 and VEGF are some of the possible events that result in IMG initiation Intussusceptive microvascular growth has been reported in mammary, colorectal, and melanoma tumors In human melanomas, a correlation between VEGF and intussusceptive angiogenesis was found, together with a higher number of intraluminal tissue folds This scenario suggests that sprouting angiogenesis inhibition could stimulate IMG.

Taking into account that intussusceptive angiogenesis only occurs in preexisting vascular structures, its most important contribution to tumor malignization is its ability to augment the number and complexity of tumor microvessel networks already created by other angiogenic mechanisms.

Ultimately, the creation of new vessel structures also provides additional surface for further activation of sprouting angiogenesis. Inside the tumor ecosystem, tumor cells are the main producers of the proangiogenic molecules that switch on the angiogenic program.

Among the molecules that regulate this process, PDGF, HGF, FGF, and, particularly, VEGF and its cognate receptors VEGFRs are the driving force, owing to their specific expression on tumor and endothelial cells. Nevertheless, other cells composing the tumor ecosystem also contribute to tumor angiogenesis and their role must be considered throughout an integrative approach Figure 1.

Cancer-associated fibroblasts normally originate from tumor or resident stroma, even though they can also differentiate from bone marrow precursors. While CAF-mediated secretion of proteases contributes to ECM degradation, CAFs also produce and deposit ECM, remarking a dual role for these cells in ECM remodeling.

Besides, CAFs also secrete multiple angiogenic cues, participating in tumor growth and progression Due to their primary localization at the leading edge of the tumor, where expanded vessel supply is demanded, the contribution to angiogenesis by stromal fibroblasts becomes crucial 52 , One of the most important molecules secreted by stromal CAFs is VEGF-A, which was found to be induced in the stroma of both spontaneously arising and implanted tumors of genetically engineered mice with a reporter for VEGF-A Actually, in ovarian carcinomas, most angiogenic growth factors are provided by CAFs rather than by malignant cells CAFs also supply other factors such as angiopoietin-1 and -2, which are needed for neovascular stabilization The tumor ecosystem constitutes a crucible of heterogenous immune cell populations, resulting in tangled interactions between tumor cells and stroma.

Immune cells have a remarkable role during the regulation of different aspects of tumor growth, such as modulation of angiogenesis and immune system evasion Particularly, the contribution of macrophages, DCs, and mast cells is further explored in this section.

Tumor-associated macrophages TAMs represent one of the most abundant leukocyte population in the tumor ecosystem and their presence correlates with a reduction in survival in most tumor types Regarding their phenotype, macrophages can be classified into the classically activated M1 and alternative activated M2 subsets.

Whereas M2 macrophages show a proangiogenic phenotype, M1 macrophages have been described as antitumor effectors TAMs often shift toward the M2 phenotype, becoming an important supplier of angiogenic cytokines and ECM remodeling molecules 60 — Indeed, in different types of tumors, macrophage presence has been correlated with high vascularity 63 , Apart from the canonical signaling pathways, alternative proangiogenic molecules such as semaphorins and plexins have been also described as mediators of the macrophage—endothelial cell cross talk Dendritic cells, due to their potent antigen-presenting ability, are considered a critical factor in antitumor immunity Nevertheless, defective myelopoiesis inside the tumor ecosystems renders DCs incompetent A role for DCs in tumor angiogenesis has been described after the finding that immature DCs increased neovascularization in implanted tumor models, while depletion of DCs revoked angiogenesis Mast cells were found more than 30 years ago to be accumulated in tumors before the onset of angiogenesis, residing in close proximity to blood vessels Those granulocytes participate in tumor rejection by IL1, IL4, IL6, and TNF-α production.

However, mast cells also promote tumor growth by increasing the angiogenic supply, degradation of the ECM and immunosuppression In detail, mast cells release angiogenic cytokines, such as VEGF, FGF-2, and TGF-β, among others Even though endothelial cells are the main players of the angiogenic tumor ecosystem, other components of the vascular system, such as platelets and pericytes, are also necessary for the proangiogenic switch.

For instance, platelets, best known for their role in assisting the blood clotting process, have also been described as proangiogenic cells.

Upon interaction with tumor cells, platelets are able to release VEGF from α granules 72 , The contractile cells that surround the basement membrane of vessels are known as pericytes. In absence of angiogenesis, pericytes commonly express proteins such as PDGFRβ, NG2, and desmin and lack expression of α-SMA.

Upon the activation of angiogenic signaling via PDGF, TGF-β, angiopoietin, and Notch, tumor pericytes loosen their attachment to the vessel, leading to a higher permeability of blood vessels 74 , Particularly, the recruitment of pericytes to the tumors highly depends on PDGF-B ligand production by endothelial cells 76 , Nevertheless, the ultimate outcome of pericyte-derived signaling remains to be fully elucidated, since it seems to be context dependent.

On the one hand, ectopic expression of PDGF-B in a mouse melanoma model increased tumor growth, indicating that a more stable and functional neovasculature was achieved through pericytes 78 , On the other hand, PDGF-B transfection into colorectal and pancreatic tumor cell lines inhibited tumor growth as a consequence of the angiostatic effect of recruited pericytes Pericytes are also involved in the control of the metastatic spread of tumor cells In fact, an increased rate of metastasis was described in a pancreatic neuroendocrine tumor mouse model genetically designed to be pericyte-poor.

It remains to be elucidated whether their protective effect against metastasis is due to their active participation or as a consequence of their passive role as a physical barrier to extravasation. The organization and composition of the matrix that supports the cells of the tumor ecosystem is essential for the regulation of angiogenesis.

In fact, mice bearing alterations in ECM molecules such as collagen, laminin, and fibronectin exhibit vascular abnormalities Vessel ECM is constituted by the basement membrane BM, which is mainly composed of collagen IV and laminin 83 and provides a broad binding surface for other ECM proteins, integrin receptors, and growth factors.

Those interactions lead to the activation of many signaling pathways, such as PI3K, AKT, and MAPK, which are involved in adhesion, migration, invasion, and proliferation, thus contributing to tumor angiogenesis The interstitial matrix that surrounds the BM, which comprises collagen I, II, and III, as well as fibronectin and fibrinogen, also contributes to tumor angiogenesis.

It primarily functions as a reservoir of regulatory molecules, such as angiogenic growth factors, cytokines, and proteolytic enzymes Moreover, binding of VEGF to fibronectin has been found to enhance the activity of VEGF.

Concomitantly, tumor and stromal cells produce proteolytic enzymes, such as MMPs, that release fragments with promigratory and proangiogenic properties 86 , besides the activation of ECM-sequestered growth factors In the absence of new vasculature, during the avascular phase, tumor growth is normally limited to no more than 1—2 mm 3.

Tumors obtain nutrients and oxygen from nearby blood vessels and angiogenic processes are not observed. The avascular tumors reach a stable state characterized by a balance between proliferation and apoptosis.

To grow beyond the restricted size and sustain unlimited proliferation, tumors require their vascular network to be extended. In pursuance of angiogenic activation, tumor cells need to undergo numerous genetic and epigenetic rearrangements that grant them the angiogenic potential for both tumor growth and latter metastasis.

Indeed, a plethora of experiments have shown that the lack of a functional vascular network leads to tumor apoptosis or necrosis, reinforcing the importance of tumor vasculature for tumor thriving The angiogenic switch depends on a dynamic balance between positive proangiogenic and negative antiangiogenic factors controlling vascular homeostasis Under physiological conditions, this balance is shifted toward negative regulation of angiogenic processes, thus maintaining the quiescence of the vasculature.

Once tumor progression is started, different mechanisms, such as the loss of tumor suppressor genes and oncogene upregulation, revert this balance. During the first steps of tumorigenesis, high levels of strong angiogenic inducers, such as VEGF and FGF, are released to the tumor ecosystem.

VEGF is regarded as the canonical angiogenesis initiator and has been found to be expressed in most types of cancer in response to different stimuli. Besides hypoxia, hypoglycemia, and growth factors, overexpression of the oncogene Myc produces a fold increase in VEGF levels Apart from VEGF, other proangiogenic molecules upregulated for the engagement of tumor angiogenesis are PDGF, EGF, TGF-β, FGF, MMPs, and angiopoietins.

Aiming at evading the ECM-associated endogenous inhibitors, tumor cells are able to further upregulate proangiogenic factors and even lose the expression of tumor suppressor genes such as p53 92 , Moreover, tumor cell metabolism shifts and becomes highly acidic, as a consequence of the Warburg effect The net increase in glucose consumption produces an abnormal lactic acid release that turns lowers extracellular pH High levels of lactate have been correlated with EMT, dissemination, and metastases of several types of human cancer, such as melanoma and Lewis lung carcinoma 96 — In detail, acidification further promotes angiogenesis through the increased expression of VEGF Lack of oxygen inside the tumor occurs as an inevitable consequence of the rapid expansion of the tumor mass.

Neoplasms have been generally described as highly hypoxic structures, bearing distorted, and abnormal vascular networks, inefficient in oxygen transportation Hypoxia is known to upregulate proangiogenic inducers and endothelial—pericyte destabilizing molecules Ang-2 and downregulate inhibitors.

Furthermore, mobilization of bone marrow-derived precursor cells and recruitment of immune cells to the tumor ecosystem is also positively controlled by hypoxia By changing the cytokine milieu, hypoxia can also induce an immunosuppressive microenvironment, allowing immune system evasion by cancer cells Hypoxia also produces a metabolic switch to apoptosis inhibition, anaerobic metabolism, increased invasiveness, EMT, and metastasis A stem-like phenotype is induced concomitantly with the release of cytokines like IL Consistently, hypoxia-driven expression of VEGF, MMPs, and ANGPTL4 is crucial for intravasation In detail, ANGPTL4 expression disrupts vascular endothelial tight junctions and augments permeability, thereby altering transendothelial barriers Aside from the role in primary tumor ecosystem maintenance, tumor angiogenesis enables tumor cell invasion and dissemination and favors the creation of new secondary tumor ecosystems at metastasized sites.

VEGF-mediated stimulation of blood and lymphatic endothelial cells provides a wide vascular area for intravasation of tumor cells, apart from increasing vascular permeability. In tumor endothelial cells, VEGF upregulates protease secretion, contributing to basement membrane degradation, and increasing the expression of molecules that mediate in tumor—endothelial cell interactions Other stromal cells also participate in the angiogenic-driven metastasis process.

Pericytes covering tumor vessels are more loosely attached to endothelial cells, affecting endothelial cell survival, and increasing the number of intercellular gaps that permit easy access for tumor cell intravasation 81 , As a consequence of the increased vascular leakiness, passive escape of tumor cells is highly induced Fighting neovascularization to halt tumor progression has become a critical step of the long-established theory of angiogenic activation for tumor growth.

In fact, more than 40 years have passed since tumor angiogenesis inhibition was first introduced as a potential therapeutic strategy 21 , Since then, many drugs targeting tumor vascularization have proven successful in the treatment of different tumors.

Such is the case for the first FDA-approved angiogenesis inhibitors sunitinib Sutent ® and bevacizumab Avastin ® , which demonstrated promising results in the treatment of kidney and colorectal cancers , Currently, using standard chemotherapy alone for cancer treatment has proven inefficient due to low selectivity of tumor cells, producing toxicity in normal tissues with high-proliferation rates e.

Besides, tumor cells become resistant, whereas the abnormality of tumor vasculature impairs efficient drug delivery On the contrary, with thousands of people being treated with VEGF inhibitors around the world, antiangiogenic targeting surely serves as an example of specific tumor ecosystem disruption for efficient cancer treatment.

There are different reasons underlying the success of tumor vascular targeting, involving both tumor and stromal cell interplay.

First, the concept that tumors are dependent on multiple factors extrinsic to themselves, so rendering them without a functional vasculature that delivers oxygen and nutrients should kill them.

Second, stromal cells, unlike neoplastic cells, are genetically more stable, being less likely to develop resistance to therapy. This makes angiogenesis a really attractive target for drug development. Third, tumors have always been described as highly vascular structures, meaning that anti-vascular targeting could be aimed at the treatment of a wide range of solid tumors , Taking into account the abundance of mechanisms involved in tumor angiogenesis, blood vessel formation processes can be inhibited at many different levels Figure 3.

Actually, distinct types of compounds, such as antibodies and small molecules, have been developed as antiangiogenic drugs.

Production of antibodies presents some disadvantages for the pharma companies regarding the expensive requirement of mammalian cell production systems, dependence on disulfide bonds for stability, overcoming the tendency to aggregation, and low expression yields.

Consequently, other promising molecules such as small globular proteins, aptamers, and peptides are currently being investigated Noteworthy, not all antiangiogenic compounds have the same cellular effects nor the same therapeutic relevance.

The main effects of angiogenic inhibitors can be classified according to their effects on: inhibition, regression, or normalization of tumor blood vessels. In this section, some of the main mechanisms to inhibit vascular malignization will be highlighted. Figure 3. Tumor angiogenesis inhibition strategies.

Due to the complexity of tumor angiogenesis, it can be inhibited at different levels. Direct vessel signaling inhibition approaches include VEGF ligand inhibitors, VEGFR receptor inhibitors, and other growth factors inhibitors released by stromal or tumor cells. Other examples are tyrosine kinase TK inhibitors, that block endothelial and pericyte cell activation, thus blocking their proliferation, migration, and survival.

Endothelial cell activation is commonly initiated upon stimulation of tyrosine kinase TK receptors by growth factors. As previously stated, VEGF is the most important growth factor involved in tumor angiogenesis, and its inhibition influences endothelial cell survival, growth, migration, blood flow, and stromal cell recruitment , Some of the VEGF-inhibiting approaches imply neutralization of the ligand or the receptor by specific antibodies, soluble receptors, small-molecule inhibitors of TK phosphorylation, and the direct inhibition of its intracellular signaling pathway Figure 3.

Thus far, 10 molecules that target VEGF or VEGFR have been approved for the treatment of various malignancies Since TK receptors are expressed both in tumor and vascular cells, TK inhibitors TKIs are regarded as a useful drugging strategy for their potentially dual effect Figure 3.

They are capable of blocking tumor cell proliferation and proangiogenic signaling simultaneously However, the efficacy of TKIs varies depending on the different expression levels of the targeted ligands and effectors depending on the tumor type.

Some strategies include compounds that block the binding site of the ATP in the TK receptor, causing the blockade of the receptor. Other TKIs aim at preventing the binding of the TK ligand with antibodies that block the growth factor or the binding site of the receptor The best known TKIs that block VEGFR and PDGF signaling are sorafenib, sunitinib, and pazopanib.

Sorafenib is a synthetic compound that inhibits both Raf signaling, involved in cell division and proliferation, and VEGFR-2 and PDGFRβ signaling, modulators of angiogenesis Its use is approved in the treatment of hepatocellular, thyroid, and renal cell carcinomas. Similarly, sunitinib is a TKI that, apart from blocking VEGFR-2 and PDGFRβ, is able to inhibit c-kit.

The FDA approved the use of sunitinib for the treatment of imatinib-resistant gastrointestinal stromal tumor and renal cell carcinoma Recently, anti-VEGFR2 antibody ramucirumab has received the FDA approval for second-line gastric cancer treatment Another example includes pazopanib, a VEGFR-1, -2, -3, c-kit, and PDGFR inhibitor, approved for renal cell carcinoma and soft tissue sarcoma Considering the contribution of EPCs to tumor angiogenesis and metastasis, blocking of EPC recruitment is a recently explored strategy for new blood vessel and metastatic niche abrogation Figure 3.

To achieve so, specific targeting of molecules involved in EPC homing and recruitment from the bone marrow is an interesting approach.

The action of these compounds is based on their ability to prevent the chemokine gradient that permits the homing of EPCs to the tumor ecosystem. Besides, VEGF is also a key modulator of EPC recruitment and preclinical studies have shown that VEGF blockade negatively modulates EPC-driven vasculogenesis Given that interactions between cells composing the tumor ecosystem and their surrounding ECM are crucial for angiogenesis regulation, modifying the structural and biochemical properties of the stroma should also impair vessel growth Figure 3.

Among all the molecules that compose the ECM, MMPs are critically relevant for angiogenesis and tumor invasion, as demonstrated by genetic ablation studies where their absence impeded angiogenic tumor growth In this context, tissue inhibitors of MMPs, together with synthetic inhibitors of serine proteases, such as urokinase type plasminogen activator, are regarded as potential antiangiogenics Importantly, there are many endogenous angiogenesis inhibitors composing the ECM that are inactivated during the angiogenic switch.

Many laboratories are trying to reproduce these natural angiogenesis inhibitors that act through binding αvβ3 and β1 integrins in endothelial cells. Some examples include arrestin, canstatin, and tumstatin Since the combination of immune checkpoint inhibitors with VEGF targeted agents shows a strong preclinical rationale, several undergoing studies are exploring its potential clinical exploitance [as reviewed in Ref.

In a recent study, the use of axitinib, a multireceptor inhibitor that targets VEGFR, PDGF, and c-kit, demonstrated a depletion of mast cells together with an improved T-cell response, pivotal for the therapeutic efficacy In comparison with physiologic tissue vasculature, tumor vasculature is characterized by aberrant, dilated, disorganized, and tortuous blood vessels.

Lack of pericyte association and vascular immaturity produce excessive permeability, increased hypoxia, and poor perfusion, resulting in decreased antitumor treatment efficacy.

For instance, chemotherapeutic drugs and immunotherapies are not able to reach all regions of the tumor , To overcome this challenge, combination of antitumor treatments and low doses of vascular targeting agents are used.

Careful dosage of antiangiogenics are able to restore normal levels of angiogenic signals in different types of tumors, provoking decreased permeability by recruiting pericytes and tightening cell—cell junctions Benefits of vascular normalization have been observed in different types of tumors.

The combination of bevacizumab, together with chemotherapy, produced a positive outcome in a subset of breast cancer patients Furthermore, combined inhibition of VEGFR and angiopoietin-2 improves survival of mouse glioblastoma tumor models, by increasing vessel normalization and reprogramming TAMs Another example of the benefits of vessel normalization include the use of trebananib, a fusion protein that inhibits angiogenesis by blocking binding of angiopoietin-1 and -2 to Tie 2 receptor.

In a recent study, combination of trebananib and chemotherapy demonstrated benefits in progression-free survival in epithelial ovarian cancer patients Far ahead from the traditional idea that neoplasms are merely characterized by the tumor cells, tumors are now regarded as a heterogeneous association of both tumor and stromal cells that contribute in an interconnected fashion to malignant progression.

The tumor ecosystem remains a bustling interchange of tumor cells, secreted molecules, and native tissue elements that, acting together, control the balance toward a proangiogenic program activation. In this way, the correct interaction between the components of the tumor ecosystem is critical for the success of the malignant lesion.

Tumor stroma acts as a co-director for the development of vascularized growing mass, becoming the rationale driving the development of new antitumor therapies with antiangiogenic drugs.

Several years after the establishment of tumor angiogenesis as a cancer hallmark, the clinical exploitation of antiangiogenic therapies has reached a certain level of maturity 6. From the archetypal sprouting angiogenesis to describing less known mechanisms such as VM, the understanding of angiogenic mechanisms has become imperative for successful therapeutic targeting.

The focus on the importance of these processes and the achievements in the clinical setting are reflected in the increasing number of drugs available to target angiogenesis mediators. Undoubtedly, the normalization of the tumor ecosystem is an important new aspect for cancer treatment.

Even though the tumor microenvironment holds many different cell types and components, the severity of the disease can be reduced by using a single effective drug, as demonstrated with antiangiogenics. Based on this observation, the combination of different therapies targeting different stromal components, together with traditional antitumor agents, could hold the key to impair cancer progression.

Despite the rapid progress achieved in tumor ecosystem targeting, only a modest clinical success has been so far observed Ongoing studies in the field which focus on studying the tumor ecosystem from an integrative point of view bear the potential to significantly control tumor angiogenesis and broaden the spectrum of current anticancer treatments.

OC declares that has been economically compensated with his assistance to advisory boards and conferences from Novartis, Pfizer, Ipsen, and Teva.

Apart from this, there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported. Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer — PubMed Abstract CrossRef Full Text Google Scholar.

Radisky D, Hagios C, Bissell MJ. Tumors are unique organs defined by abnormal signaling and context. Semin Cancer Biol — Rak JW, St Croix BD, Kerbel RS. Consequences of angiogenesis for tumor progression, metastasis and cancer therapy. Anticancer Drugs — Liotta LA, Kohn EC. The microenvironment of the tumour-host interface.

Nature —9. Shojaei F, Ferrara N. Drug Resist Updat — Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell — CrossRef Full Text Google Scholar. Polyak K, Haviv I, Campbell IG. Co-evolution of tumor cells and their microenvironment. Trends Genet —8.

Polyak K, Weinberg RA. Transitions between epithelial and mesenchymal states: acquisition of malignant and stem cell traits. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med — Mittal K, Ebos J, Rini B. Angiogenesis and the tumor microenvironment: vascular endothelial growth factor and beyond.

Semin Oncol — ElShamy WM, Sinha A, Said N. Aggressiveness niche: can it be the foster ground for cancer metastasis precursors? Stem Cells Int —7. Muz B, de la Puente P, Azab F, Azab AK. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy.

Hypoxia Auckl Yang Y, Sun M, Wang L, Jiao B. HIFs, angiogenesis, and cancer. J Cell Biochem — Vandekeere S, Dewerchin M, Carmeliet P. Angiogenesis revisited: an overlooked role of endothelial cell metabolism in vessel sprouting. Microcirculation — Nowak-Sliwinska P, Alitalo K, Allen E, Anisimov A, Aplin AC, Auerbach R, et al.

Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis :1— Ribatti D. The chick embryo chorioallantoic membrane as a model for tumor biology. Exp Cell Res — Vogel HB, Berry RG.

Chorioallantoic membrane heterotransplantation of human brain tumors. Int J Cancer —8. Tulotta C, He S, van der Ent W, Chen L, Groenewoud A, Spaink HP, et al.

Imaging Cancer Angiogenesis and Metastasis in a Zebrafish Embryo Model. Cham: Springer Google Scholar. Gengenbacher N, Singhal M, Augustin HG.

Preclinical mouse solid tumour models: status quo, challenges and perspectives. Soleimani S, Shamsi M, Ghazani MA, Modarres HP, Valente KP, Saghafian M, et al.

Translational models of tumor angiogenesis: a nexus of in silico and in vitro models. Biotechnol Adv — Sherwood LM, Parris EE, Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med —6. Carmeliet P, Jain RK.

Molecular mechanisms and clinical applications of angiogenesis. Nature — Paku S, Paweletz N. First steps of tumor-related angiogenesis. Lab Invest — PubMed Abstract Google Scholar.

Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Jain RK. Molecular regulation of vessel maturation. Iruela-Arispe ML, Dvorak HF. Angiogenesis: a dynamic balance of stimulators and inhibitors. Thromb Haemost —7. Gerhardt H, Golding M, Fruttiger M, Ruhrberg C, Lundkvist A, Abramsson A, et al.

VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J Cell Biol — Dejana E, Orsenigo F, Molendini C, Baluk P, McDonald DM. Organization and signaling of endothelial cell-to-cell junctions in various regions of the blood and lymphatic vascular trees.

Cell Tissue Res — Leslie JD, Ariza-McNaughton L, Bermange AL, McAdow R, Johnson SL, Lewis J. Endothelial signalling by the notch ligand delta-like 4 restricts angiogenesis.

Development — Bautch VL. Endothelial cells form a phalanx to block tumor metastasis. Cell —2. Geudens I, Gerhardt H. Coordinating cell behaviour during blood vessel formation. Risau W. Mechanisms of angiogenesis. Nature —4. Rafii DC, Psaila B, Butler J, Jin DK, Lyden D.

Regulation of vasculogenesis by platelet-mediated recruitment of bone marrow-derived cells. Arterioscler Thromb Vasc Biol — Brown JM. Vasculogenesis: a crucial player in the resistance of solid tumours to radiotherapy.

Br J Radiol Benezra R, Rafii S, Lyden D. The Id proteins and angiogenesis. Oncogene — Heissig B, Hattori K, Dias S, Friedrich M, Ferris B, Hackett NR, et al.

Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Deb A, Skelding KA, Wang S, Reeder M, Simper D, Caplice NM. Integrin profile and in vivo homing of human smooth muscle progenitor cells. Circulation —7.

Urbich C, Dimmeler S. Endothelial progenitor cells. Trends Cardiovasc Med — Jin F, Brockmeier U, Otterbach F, Metzen E. Mol Cancer Res — Kopp H-G, Hooper AT, Broekman MJ, Avecilla ST, Petit I, Luo M, et al. Thrombospondins deployed by thrombopoietic cells determine angiogenic switch and extent of revascularization.

J Clin Invest — Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol — Frenkel S, Barzel I, Levy J, Lin AY, Bartsch D-U, Majumdar D, et al. Demonstrating circulation in vasculogenic mimicry patterns of uveal melanoma by confocal indocyanine green angiography.

Eye — Hendrix MJC, Seftor EA, Hess AR, Seftor REB. Angiogenesis: vasculogenic mimicry and tumour-cell plasticity: lessons from melanoma. Seftor EA, Brown KM, Chin L, Kirschmann DA, Wheaton WW, Protopopov A, et al. Epigenetic transdifferentiation of normal melanocytes by a metastatic melanoma microenvironment.

Cancer Res —9. Sun B, Zhang S, Zhang D, Du J, Guo H, Zhao X, et al. Vasculogenic mimicry is associated with high tumor grade, invasion and metastasis, and short survival in patients with hepatocellular carcinoma.

Oncol Rep —8. Caduff JH, Fischer LC, Burri PH. Scanning electron microscope study of the developing microvasculature in the postnatal rat lung. Anat Rec — Burri PH, Hlushchuk R, Djonov V. Intussusceptive angiogenesis: its emergence, its characteristics, and its significance.

Dev Dyn — Djonov V, Makanya AN. New insights into intussusceptive angiogenesis. EXS — Dome F, Taziaux P, Boniver J, Fridman V, Delbecque K. Rev Med Liege — Ribatti D, Nico B, Floris C, Mangieri D, Piras F, Ennas MG, et al. Microvascular density, vascular endothelial growth factor immunoreactivity in tumor cells, vessel diameter and intussusceptive microvascular growth in primary melanoma.

Oncol Rep —4. Watnick RS. The role of the tumor microenvironment in regulating angiogenesis. Cold Spring Harb Perspect Med 2:a Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF, Harrington K, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells.

Nat Cell Biol — Granot D, Addadi Y, Kalchenko V, Harmelin A, Kunz-Schughart LA, Neeman M. In vivo imaging of the systemic recruitment of fibroblasts to the angiogenic rim of ovarian carcinoma tumors. Fukumura D, Xavier R, Sugiura T, Chen Y, Park EC, Lu N, et al. Tumor induction of VEGF promoter activity in stromal cells.

Thijssen VLJL, Brandwijk RJMGE, Dings RPM, Griffioen AW. Angiogenesis gene expression profiling in xenograft models to study cellular interactions. Gilad AA, Israely T, Dafni H, Meir G, Cohen B, Neeman M.

Functional and molecular mapping of uncoupling between vascular permeability and loss of vascular maturation in ovarian carcinoma xenografts: the role of stroma cells in tumor angiogenesis.

Int J Cancer — Albini A, Bruno A, Noonan DM, Mortara L. Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: implications for immunotherapy.

Front Immunol Petty AJ, Yang Y.

Tumor vascularization occurs Angiogenesis and cancer several canver Sports nutrition processes, which not Angiogenesiis vary between tumor type and anatomic location, but also occur simultaneously camcer the same cancer Angiogenesis and cancer. These processes are orchestrated cabcer a Sports nutrition Natural antifungal remedies for candida secreted cajcer and signaling Sports nutrition and can involve participation of non-endothelial cells, such as progenitors or cancer stem cells. Anti-angiogenic therapies using either antibodies or tyrosine kinase inhibitors have been approved to treat several types of cancer. However, the benefit of treatment has so far been modest, some patients not responding at all and others acquiring resistance. It is becoming increasingly clear that blocking tumors from accessing the circulation is not an easy task to accomplish. Tumor vessel functionality and gene expression often differ vastly when comparing different cancer subtypes, and vessel phenotype can be markedly heterogeneous within a single tumor. Angiovenesis Angiogenesis and cancer and resources for Angiogenesis and cancer and returning patients. Learn about clinical Enhance mental acuity at MD Anderson and search Angiogenesis and cancer database cxncer open studies. The Lyda Hill Performance feedback and analysis Prevention Center provides cancer risk Angioogenesis, screening and diagnostic Angiogenesis and cancer. Your gift will help support our mission to end cancer and make a difference in the lives of our patients. Our personalized portal helps you refer your patients and communicate with their MD Anderson care team. As part of our mission to eliminate cancer, MD Anderson researchers conduct hundreds of clinical trials to test new treatments for both common and rare cancers. Choose from 12 allied health programs at School of Health Professions. Angiogenesis and cancer

Author: Malrajas

3 thoughts on “Angiogenesis and cancer

  1. Ich entschuldige mich, aber meiner Meinung nach irren Sie sich. Geben Sie wir werden es besprechen. Schreiben Sie mir in PM.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com