Angiogenesis in Cancer and Its Therapeutic Targeting: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Vascular endothelial growth factors (VEGFs) are the key regulators of vasculogenesis in normal and oncological development. VEGF-A is the most studied angiogenic factor secreted by malignant tumor cells under hypoxic and inflammatory stress, which made VEGF-A a rational target for anticancer therapy. However, inhibition of VEGF-A by monoclonal antibody drugs led to the upregulation of VEGF-D. VEGF-D was primarily described as a lymphangiogenic factor; however, VEGF-D’s blood angiogenic potential comparable to VEGF-A has already been demonstrated in glioblastoma and colorectal carcinoma. These findings suggested a role for VEGF-D in facilitating malignant tumor growth by bypassing the anti-VEGF-A antiangiogenic therapy. Owing to its high mitogenic ability, higher affinity for VEGFR-2, and higher expression in cancer, VEGF-D might even be a stronger angiogenic driver and, hence, a better therapeutic target than VEGF-A.

  • VEGF-D
  • angiogenesis
  • lymphangiogenesis
  • growth factors

1. Introduction

In order to meet the increased nutrient and oxygen demand, growing malignant tumors require blood supply, and hence, promote angiogenesis from existing vessels by inducing sprouting [1]. The malignant tumor vasculature has a significantly different structure and physiology as compared to normal blood vessels [2]. Vascular endothelial growth factors (VEGFs) are the primary growth factors regulating angiogenesis. Upon binding their corresponding vascular endothelial growth factor receptors (VEGFRs), VEGFs promote the proliferation and migration of endothelial cells, tube formation, increase vascular permeability and vascular endothelial cell survival, altogether angiogenesis [3]. Under pathologic conditions such as malignant tumor development, poor vasculature, and hypoxia trigger the expression of VEGFs, which activate ECs both in an autocrine and paracrine fashion, thereby increasing EC proliferation and migration [4]. Secreted VEGFs also increase vascular permeability and facilitate the transfer of plasma proteins into the extracellular matrix, where these proteins provide provisional support to the incoming ECs and facilitate vasculogenesis [3][5].
VEGFs belong to the Platelet Derived Growth Factor (PDGF) family, an important mitogenic family that modulates various biochemical pathways involved in cell growth, proliferation, and survival, as well as maintaining the structural integrity of the cell [6]. VEGFs are particularly important for blood vessel formation during embryogenesis. In mammals, members of the VEGF family include VEGF-A, VEGF-B, VEGF-C, VEGF-D, and Placental Growth Factor (PIGF). There are three types of VEGF receptors to which the members of the VEGF family can bind with varying affinity and specificity, thereby eliciting different responses. VEGFR-1 and VEGFR-2 are expressed on vascular endothelial cells (VECs) and, in some instances, on non-endothelial cells [7]. VEGFR-3 is expressed particularly on lymphatic endothelial cells (LECs) [3][8][9].

2. Angiogenesis in Cancer and Its Therapeutic Targeting

A tumor cell’s ability to induce angiogenesis is one of the hallmarks of cancer, as angiogenesis is essential for tumor cells to survive [10]. Furthermore, the autocrine production of growth factors and cytokines by the active ECs promotes tumor progression [11]. Tumor angiogenesis (the angiogenic switch) is initiated in response to hypoxic or inflammatory stimuli in the tumor microenvironment. During the angiogenic switch, ECs are activated and start to produce angiogenic growth factors such as Fibroblast Growth Factors (FGFs), TGF-β, PDGF, and VEGFs (A and D) [12][13]. The binding of these growth factors to their EC receptors leads to the production of matrix-degrading enzymes, enabling blood vessel growth. VEGF-A mediated activation of VEGFR-2 leads to degradation of the basement membrane followed by the transformation of ECs to motile tip cells at the sprouting end of the vessel [13][14] endothelial cell migration [15], and EC proliferation [16][17][18]. Angiogenesis is mediated by the coordination of dynamic tip cells with filopodia and stalk cells. Tip cells sense the pro or antiangiogenic factors in the tumor microenvironment and migrate, while the stalk cells have less filopodia and a faster proliferation rate to facilitate tube formation [14]. The new ECs establish tight junctions, and hence, a luminal vessel is produced [19][20]. The newly formed vessels are then covered by tumor-recruited pericytes, which provide support to the vessels from outside (maturation step) [21]. Unlike normal blood vessels, blood vessels in tumors are more permeable and tortuous, which is attributed to the altered endothelial cells (ECs) referred to as Tumor Endothelial Cells (TECs) [22][23]. Furthermore, besides the autocrine production of proangiogenic factors by tumor cells, tumor cells can prime immune cells to produce more angiogenic factors, which results in a loss of balance between the pro and antiangiogenic factors, rendering the tumor vasculature unruly and poorly developed [24]. Tumour blood vessels thereby have loose junctions, an incomplete basement membrane distribution, and reduced pericyte support. This deformed vascular physiology leads to poor blood supply and, therefore, hypoxia and reduced drug delivery [12][25].
Tumor vasculature and angiogenesis are important prerequisites of tumor cell proliferation, survival, and progression. Therefore, it is a feasible approach to inhibit tumor angiogenesis to target cancer proliferation. In the early 1990s, a murine anti-human VEGF-A antibody was developed that could reduce angiogenesis in vivo. A few years later, the antibody was humanized and was termed Bevacizumab (Avastin), which had the ability to bind and neutralize all VEGF-A isoforms [26]. Bevacizumab was approved by the U.S. Food and Drug Administration (FDA) as a first-line antiangiogenic therapy for colorectal cancer (CRC) in 2004 and is a common drug of choice for various cancers today [27]. Ever since the discovery of Avastin, tumor angiogenesis and factors that might be involved in the regulation of the process have been extensively researched. There have been several candidates, but only a few were approved for antiangiogenic therapy to treat cancer, age-related macular degeneration (AMD), and diabetic retinopathy. Approved VEGF-A specific monoclonal antibodies for the treatment of cancer include Bevacizumab (Avastin for non-small cell lung cancer (NSCLC), glioblastoma, metastatic renal cell-, and cervical cancer) and ramucirumab (Cyramza for gastric, NSCLC and metastatic colorectal cancer). Further anti-VEGF-A antibodies are approved for macular edema (Ranibizumab (Lucentis) and Aflibercept (Eylea)). In addition, the targeted therapy, antibodies with a broader target range targeting tyrosine kinase receptor, i.e., sorafenib (Nexavar), regorafenib (Stivarga), and sunitinib (Sutent) are also used to inhibit VEGFR activation and hence angiogenesis [28][29].

2.1. VEGF-D and Blood Capillary Angiogenesis

Like many other types of anticancer treatments, tumor cells developed resistance to antiangiogenic therapies [30][31][32]. Altered gene expression of angiogenesis-related genes in tumor cells was demonstrated in response to antiangiogenic drugs. Particularly, the expression of VEGFs was altered both in tumor cells and the surrounding stroma [28][32][33][34]. Since other members of the VEGF family could activate VEGFR-2 in the absence of VEGF-A, this could be a potential mechanism of antiangiogenic therapy resistance [35]. An interesting finding was that Avastin-mediated VEGF-A inhibition led to an upregulation of VEGF-D expression in gliosarcomas, and although the tumor growth was slower, the tumors when established, had better vascularization [36]. The normalization of this neo vasculature post-VEGF-A inhibition resulted in morphological changes: increased pericyte coverage, less leaking, less dilation and functional changes; decreased interstitial fluid pressure, increased tumor oxygenation, and improved penetration of drugs into these tumors [37]. VEGF-D can, therefore, execute all steps of tumor angiogenesis in the absence of VEGF-A with comparable efficiency [17][38].
Similarly, upregulated VEGF-D expression was reported in inflammatory breast cancer after treatment with celecoxib (non-steroidal anti-inflammatory drug) and VEGFR-2 inhibitor SU5416 [39]. Furthermore, patients with nonresectable hepatocellular carcinoma, already receiving bevacizumab, had a higher VEGF-D expression and promoted disease progression [40]. The ability of VEGF-D as an inducer of blood capillary angiogenesis was first characterized in rabbit retinal tissue [41] and was confirmed in human embryonic kidney cell line: induced VEGF-D expression resulted in the formation of highly vascularized and non-oedemic tumors as compared to tumors of wild type cells in mice [42]. VEGF-D can induce CD-31, as well as LYVE-1 positive vessels, by activating either VEGFR-2 or VEGFR-3, respectively, in the absence of VEGF-A [42]. Upon adenoviral vector delivery to skeletal muscles, mature VEGF-D was an excellent inducer of both lymphangiogenesis and angiogenesis, promoting pericyte recruitment and vascular permeability and a more diffused angiogenesis pattern [43]. A similar dose-dependent angiogenic response was observed in pig hearts [44]. VEGF-D also induced angiogenesis in the brain after blood–brain barrier breakdown by activating both VEGFR-2 and VEGFR-3 [45]. VEGF-D modulates angiogenesis by inducing EC proliferation and migration to branching points and facilitating tube formation [43][46][47]. Based on these findings, VEGF-D expressed by tumor cells could be a way for cancer to circumvent VEGF-A targeting antiangiogenic therapy [48][49][50]. Interestingly, other forms of anticancer therapies, such as docetaxel and vinorelbine, also upregulate VEGF-D expression and induced angiogenesis in vitro in HUVEC and BC cells in relation to melatonin signaling [51]; this further indicates that VEGF-D has a unique yet unidentified expression and signaling pattern which should be explored to facilitate multi-targeted antiangiogenic therapy.

2.2. VEGF-D Mediated Angiogenic Signaling in Cancer

Since VEGF-D can activate VEGFR-2 mediated signaling, it mainly activates signaling pathways involved in the proliferation, migration, survival, and physiology of EC cells [29], essentially maintaining the vascular system in development and disease [52]. Although researchers are in agreement that upregulated VEGF-D in response to antiangiogenic drugs might be one of the key factors enabling antiangiogenic drug resistance in cancers [28], little is known about the signaling pathways and expression regulators involved.
One of the main factors promoting VEGF-D expression in response to antiangiogenic therapy would be therapy-related hypoxia [53]. Achen et al. reported that VEGF-D was secreted exclusively by tumor cells in two independent model systems, acting in a paracrine fashion to activate VEGFR-2 positive blood vessels in tumors [54]. Furthermore, VEGF-D secreted by the tumor cells was localized in metastatic melanoma blood vessels endothelial cells near the immune-positive tumor cells but not in the distant blood vessels, which further supports that VEGF-D promotes angiogenesis in a paracrine fashion [55][56][57]. However, due to the localized production of VEGF-D by tumors and its role in promoting tumor cell progression, migration, and metastasis, it might also have an autocrine function in addition to its paracrine signaling as reported in endometrial carcinoma [58] and invasive cervical carcinoma [59]. Furthermore, VEGF-D could induce proangiogenic phenotype in HUVECs by upregulating genes involved in matrix modulation and cell membrane alteration in an autocrine manner [60]. Interestingly, in Gastric cancer (GC), VEGF-D transcription was facilitated by proteolytic activation of transcriptional factor CDP/Cux p200 by protease cathepsin L (CTSL). This upregulated VEGF-D translated to higher angiogenesis in GC [61]. Recently, in GC, a long noncoding RNA (LncRNA) CRART16 was found to downregulate miR-122-5p, thereby upregulating transcriptional factor FOS and hence upregulated VEGF-D expression and angiogenesis both in vitro and in vivo [62].
Some interesting points for research would include investigating if VEGF-D activates the same downstream signaling cascades as VEGF-A when activating VEGFR-2. Based on the bioinformatics analysis, VEGF-D potentially activates the MAPK signaling pathway, promoting cell proliferation, differentiation, and survival [63]. Secondly, as in glioblastoma, vascular, breast, and lung cancer, the tumor tissue has high expression of VEGFR-3 that unconventionally contributes to maintaining endothelial integrity in tumor blood vascular angiogenesis [64][65][66][67]. It would be interesting to investigate how VEGF-D/VEGFR-3 signaling is stimulating angiogenesis.

This entry is adapted from the peer-reviewed paper 10.3390/ijms241713317

References

  1. Hanahan, D.; Folkman, J. Patterns and Emerging Mechanisms of the Angiogenic Switch during Tumorigenesis. Cell 1996, 86, 353–364.
  2. Ruoslahti, E. Specialization of tumour vasculature. Nat. Rev. Cancer 2002, 2, 83–90.
  3. Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)—Key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467.
  4. Raza, A.; Franklin, M.J.; Dudek, A.Z. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am. J. Hematol. 2010, 85, 593–598.
  5. Miron, L.; Gafton, B.; Marinca, M. Angiogeneza Tumorală-Implicaţii în Terapia Cancerelor. J. Chir. 2010, 6, 2.
  6. La Mendola, D.; Trincavell, M.L.; Martini, C. Angiogenesis in Disease. Int. J. Mol. Sci. 2022, 23, 10962.
  7. Shibuya, M. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: A crucial target for anti- and pro-angiogenic therapies. Genes Cancer 2011, 2, 1097–1105.
  8. Dakowicz, D.; Zajkowska, M.; Mroczko, B. Relationship between VEGF Family Members, Their Receptors and Cell Death in the Neoplastic Transformation of Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 3375.
  9. Hamrah, P.; Chen, L.; Cursiefen, C.; Zhang, Q.; Joyce, N.C.; Dana, M.R. Expression of vascular endothelial growth factor receptor-3 (VEGFR-3) on monocytic bone marrow-derived cells in the conjunctiva. Exp. Eye Res. 2004, 79, 553–561.
  10. Suresh, R.; Diaz, R.J. The remodelling of actin composition as a hallmark of cancer. Transl. Oncol. 2021, 14, 101051.
  11. Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int. J. Mol. Sci. 2018, 19, 1272.
  12. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770.
  13. Al-Ostoot, F.H.; Salah, S.; Khamees, H.A.; Khanum, S.A. Tumor Angiogenesis: Current Challenges and Therapeutic Opportunities (Review). Cancer Treat. Res. Commun. 2021, 28, 100422.
  14. Mazurek, R.; Dave, J.M.; Chandran, R.R.; Misra, A.; Sheikh, A.Q.; Greif, D.M. Vascular cells in blood vessel wall development and disease. Adv. Pharmacol. 2017, 78, 323–350.
  15. Cai, H.; Gong, L.; Liu, J.; Zhou, Q.; Zheng, Z. Diosgenin inhibits tumor angiogenesis through regulating GRP78-mediated HIF-1α and VEGF/VEGFR signaling pathways. Die Pharm. Int. J. Pharm. Sci. 2019, 74, 680–684.
  16. Domigan, C.K.; Ziyad, S.; Iruela-Arispe, M.L. Canonical and noncanonical vascular endothelial growth factor pathways: New developments in biology and signal transduction. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 30–39.
  17. Vimalraj, S. A concise review of VEGF, PDGF, FGF, Notch, angiopoietin, and HGF signalling in tumor angiogenesis with a focus on alternative approaches and future directions. Int. J. Biol. Macromol. 2022, 221, 1428–1438.
  18. Qi, S.; Deng, S.; Lian, Z.; Yu, K. Novel Drugs with High Efficacy against Tumor Angiogenesis. Int. J. Mol. Sci. 2022, 23, 6934.
  19. Iruela-Arispe, M.L.; Davis, G.E. Cellular and Molecular Mechanisms of Vascular Lumen Formation. Dev. Cell 2009, 16, 222–231.
  20. Dejana, E.; Tournier-Lasserve, E.; Weinstein, B.M. The Control of Vascular Integrity by Endothelial Cell Junctions: Molecular Basis and Pathological Implications. Dev. Cell 2009, 16, 209–221.
  21. Lees, D.M.; Reynolds, L.E.; Pedrosa, A.R.; Roy-Luzarraga, M.; Hodivala-Dilke, K.M. Phosphorylation of pericyte FAK-Y861 affects tumour cell apoptosis and tumour blood vessel regression. Angiogenesis 2021, 24, 471–482.
  22. Maishi, N.; Annan, D.A.; Kikuchi, H.; Hida, Y.; Hida, K. Tumor Endothelial Heterogeneity in Cancer Progression. Cancers 2019, 11, 1511.
  23. Taleb, M.; Mohammadkhani, N.; Bahreini, F.; Ovais, M.; Nie, G. Modulation of Tumor Vasculature Network: Key Strategies. Small Struct. 2022, 3, 2100164.
  24. Ozel, I.; Duerig, I.; Domnich, M.; Lang, S.; Pylaeva, E.; Jablonska, J. The Good, the Bad, and the Ugly: Neutrophils, Angiogenesis, and Cancer. Cancers 2022, 14, 536.
  25. Zhou, W.; Yang, L.; Nie, L.; Lin, H. Unraveling the molecular mechanisms between inflammation and tumor angiogenesis. Am. J. Cancer Res. 2021, 11, 301–317.
  26. Ferrara, N.; Hillan, K.J.; Gerber, H.-P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400.
  27. Garcia, J.; Hurwitz, H.I.; Sandler, A.B.; Miles, D.; Coleman, R.L.; Deurloo, R.; Chinot, O.L. Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat. Rev. 2020, 86, 102017.
  28. Huang, M.; Lin, Y.; Wang, C.; Deng, L.; Chen, M.; Assaraf, Y.G.; Chen, Z.-S.; Ye, W.; Zhang, D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist. Updat. 2022, 64, 100849.
  29. Liu, X.-J.; Zhao, H.-C.; Hou, S.-J.; Zhang, H.-J.; Cheng, L.; Yuan, S.; Zhang, L.-R.; Song, J.; Zhang, S.-Y.; Chen, S.-W. Recent development of multi-target VEGFR-2 inhibitors for the cancer therapy. Bioorg. Chem. 2023, 133, 106425.
  30. Welti, J.; Loges, S.; Dimmeler, S.; Carmeliet, P. Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer. J. Clin. Investig. 2013, 123, 3190–3200.
  31. Jain, R.K. Antiangiogenesis Strategies Revisited: From Starving Tumors to Alleviating Hypoxia. Cancer Cell 2014, 26, 605–622.
  32. Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603.
  33. Huijbers, E.J.; van Beijnum, J.R.; Thijssen, V.L.; Sabrkhany, S.; Nowak-Sliwinska, P.; Griffioen, A.W. Role of the tumor stroma in resistance to anti-angiogenic therapy. Drug Resist. Updat. 2016, 25, 26–37.
  34. Cascone, T.; Herynk, M.H.; Xu, L.; Du, Z.; Kadara, H.; Nilsson, M.B.; Oborn, C.J.; Park, Y.-Y.; Erez, B.; Jacoby, J.J.; et al. Upregulated stromal EGFR and vascular remodeling in mouse xenograft models of angiogenesis inhibitor–resistant human lung adenocarcinoma. J. Clin. Investig. 2011, 121, 1313–1328.
  35. Mashima, T.; Wakatsuki, T.; Kawata, N.; Jang, M.-K.; Nagamori, A.; Yoshida, H.; Nakamura, K.; Migita, T.; Seimiya, H.; Yamaguchi, K. Neutralization of the induced VEGF-A potentiates the therapeutic effect of an anti-VEGFR2 antibody on gastric cancer in vivo. Sci. Rep. 2021, 11, 15125.
  36. Moffat, B.A.; Chen, M.; Kariaapper, M.S.; Hamstra, D.A.; Hall, D.E.; Stojanovska, J.; Johnson, T.D.; Blaivas, M.; Kumar, M.; Chenevert, T.L.; et al. Inhibition of Vascular Endothelial Growth Factor (VEGF)-A Causes a Paradoxical Increase in Tumor Blood Flow and Up-Regulation of VEGF-D. Clin. Cancer Res. 2006, 12, 1525–1532.
  37. Jain, R.K. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science 2005, 307, 58–62.
  38. Rajabi, M.; Mousa, S.A. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017, 5, 34.
  39. Astin, J.W.; Haggerty, M.J.L.; Okuda, K.S.; Le Guen, L.; Misa, J.P.; Tromp, A.; Hogan, B.M.; Crosier, K.E.; Crosier, P.S. Vegfd can compensate for loss of Vegfc in zebrafish facial lymphatic sprouting. Development 2014, 141, 2680–2690.
  40. Yang, Z.; Suda, G.; Maehara, O.; Ohara, M.; Yoda, T.; Sasaki, T.; Kohya, R.; Yoshida, S.; Hosoda, S.; Tokuchi, Y.; et al. Changes in Serum Growth Factors during Resistance to Atezolizumab Plus Bevacizumab Treatment in Patients with Unresectable Hepatocellular Carcinoma. Cancers 2023, 15, 593.
  41. Marconcini, L.; Marchiò, S.; Morbidelli, L.; Cartocci, E.; Albini, A.; Ziche, M.; Bussolino, F.; Oliviero, S. c-fos-induced growth factor/vascular endothelial growth factor D induces angiogenesis in vivo and in vitro. Proc. Natl. Acad. Sci. USA 1999, 96, 9671–9676.
  42. Stacker, S.A.; Caesar, C.; Baldwin, M.E.; Thornton, G.E.; Williams, R.A.; Prevo, R.; Jackson, D.G.; Nishikawa, S.-I.; Kubo, H.; Achen, M.G. VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nat. Med. 2001, 7, 186–191.
  43. Rissanen, T.T.; Markkanen, J.E.; Gruchala, M.; Heikura, T.; Puranen, A.; Kettunen, M.I.; Kholová, I.; Kauppinen, R.A.; Achen, M.G.; Stacker, S.A.; et al. VEGF-D Is the Strongest Angiogenic and Lymphangiogenic Effector Among VEGFs Delivered into Skeletal Muscle via Adenoviruses. Circ. Res. 2003, 92, 1098–1106.
  44. Rutanen, J.; Rissanen, T.T.; Markkanen, J.E.; Gruchala, M.; Silvennoinen, P.; Kivelä, A.; Hedman, A.; Hedman, M.; Heikura, T.; Ordén, M.-R.; et al. Adenoviral Catheter-Mediated Intramyocardial Gene Transfer Using the Mature Form of Vascular Endothelial Growth Factor-D Induces Transmural Angiogenesis in Porcine Heart. Circulation 2004, 109, 1029–1035.
  45. Nag, S.; Manias, J.; Eubanks, J.H.; Stewart, D.J. Increased Expression of Vascular Endothelial Growth Factor-D Following Brain Injury. Int. J. Mol. Sci. 2019, 20, 1594.
  46. Bower, N.I.; Vogrin, A.J.; Le Guen, L.; Chen, H.; Stacker, S.A.; Achen, M.G.; Hogan, B.M. Vegfd modulates both angiogenesis and lymphangiogenesis during zebrafish embryonic development. Development 2017, 144, 507–518.
  47. Gangadaran, P.; Rajendran, R.L.; Oh, J.M.; Oh, E.J.; Hong, C.M.; Chung, H.Y.; Lee, J.; Ahn, B.C. Identification of Angiogenic Cargo in Extracellular Vesicles Secreted from Human Adipose Tis-sue-Derived Stem Cells and Induction of Angiogenesis In Vitro and In Vivo. Pharmaceutics 2021, 13, 495.
  48. Hanrahan, V.; Currie, M.J.; Gunningham, S.P.; Morrin, H.R.; Scott, P.A.; Robinson, B.A.; Fox, S.B. The angiogenic switch for vascular endothelial growth factor (VEGF)-A, VEGF-B, VEGF-C, and VEGF-D in the adenoma–carcinoma sequence during colorectal cancer progression. J. Pathol. 2003, 200, 183–194.
  49. Debinski, W.; Slagle-Webb, B.; Achen, M.G.; Stacker, S.A.; Tulchinsky, E.; Gillespie, G.Y.; Gibo, D.M. VEGF-D is an X-linked/AP-1 Regulated Putative Onco-angiogen in Human Glioblastoma Multiforme. Mol. Med. 2001, 7, 598–608.
  50. Badodekar, N.; Sharma, A.; Patil, V.; Telang, G.; Sharma, R.; Patil, S.; Vyas, N.; Somasundaram, I. Angiogenesis induction in breast cancer: A paracrine paradigm. Cell Biochem. Funct. 2021, 39, 860–873.
  51. González-González, A.; González, A.; Rueda, N.; Alonso-González, C.; Menéndez, J.M.; Martínez-Campa, C.; Mitola, S.; Cos, S. Usefulness of melatonin as complementary to chemotherapeutic agents at different stages of the angiogenic process. Sci. Rep. 2020, 10, 4790.
  52. Wang, X.; Bove, A.M.; Simone, G.; Ma, B. Molecular Bases of VEGFR-2-Mediated Physiological Function and Pathological Role. Front. Cell Dev. Biol. 2020, 8, 599281.
  53. Teng, X.; Li, D.; Johns, R.A. Hypoxia up-regulates mouse vascular endothelial growth factor D promoter activity in rat pulmonary microvascular smooth-muscle cells. Chest 2002, 121, 82S–83S.
  54. Achen, M.G.; Williams, R.A.; Baldwin, M.E.; Lai, P.; Roufail, S.; Alitalo, K.; Stacker, S.A. The Angiogenic and Lymphangiogenic Factor Vascular Endothelial Growth Factor-D Exhibits a Paracrine Mode of Action in Cancer. Growth Factors 2002, 20, 99–107.
  55. Baldwin, M.E.; Catimel, B.; Nice, E.C.; Roufail, S.; Hall, N.E.; Stenvers, K.L.; Karkkainen, M.J.; Alitalo, K.; Stacker, S.A.; Achen, M.G. The Specificity of Receptor Binding by Vascular Endothelial Growth Factor-D Is Different in Mouse and Man. J. Biol. Chem. 2001, 276, 19166–19171.
  56. Achen, M.G.; Williams, R.A.; Minekus, M.P.; Thornton, G.E.; Stenvers, K.; Rogers, P.A.W.; Lederman, F.; Roufail, S.; Stacker, S.A. Localization of vascular endothelial growth factor-D in malignant melanoma suggests a role in tumour angiogenesis. J. Pathol. 2000, 193, 147–154.
  57. Vacca, A.; Ria, R.; Ribatti, D.; Semeraro, F.; Djonov, V.; Di Raimondo, F.; Dammacco, F. A paracrine loop in the vascular endothelial growth factor pathway triggers tumor angiogenesis and growth in multiple myeloma. Haematologica 2003, 88, 176–185.
  58. Yokoyama, Y.; Charnock-Jones, D.S.; Licence, D.; Yanaihara, A.; Hastings, J.M.; Holland, C.M.; Emoto, M.; Sakamoto, A.; Sakamoto, T.; Maruyama, H.; et al. Expression of vascular endothelial growth factor (VEGF)-D and its receptor, VEGF receptor 3, as a prognostic factor in endometrial carcinoma. Clin. Cancer Res. 2003, 9, 1361–1369.
  59. Yu, H.; Zhang, S.; Zhang, R.; Zhang, L. The role of VEGF-C/D and Flt-4 in the lymphatic metastasis of early-stage invasive cervical carcinoma. J. Exp. Clin. Cancer Res. 2009, 28, 96–98.
  60. Papiewska-Pajak, I.; Boncela, J.; Przygodzka, P.; Cierniewski, C.S. Autocrine effects of VEGF-D on endothelial cells after transduction with AD-VEGF-DΔNΔC. Exp. Cell Res. 2010, 316, 907–914.
  61. Pan, T.; Jin, Z.; Yu, Z.; Wu, X.; Chang, X.; Fan, Z.; Li, F.; Wang, X.; Li, Z.; Zhou, Q.; et al. Cathepsin L promotes angiogenesis by regulating the CDP/Cux/VEGF-D pathway in human gastric cancer. Gastric Cancer 2020, 23, 974–987.
  62. Zhang, J.; Pang, X.; Lei, L.; Zhang, J.; Zhang, X.; Chen, Z.; Zhu, J.; Jiang, Y.; Chen, G.; Wu, Y.; et al. LncRNA CRART16/miR-122-5p/FOS axis promotes angiogenesis of gastric cancer by upregulating VEGFD expression. Aging 2022, 14, 4137–4157.
  63. Guo, Y.J.; Pan, W.W.; Liu, S.B.; Shen, Z.F.; Xu, Y.; Hu, L.L. ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med. 2020, 19, 1997–2007.
  64. Partanen, T.A.; Alitalo, K.; Miettinen, M. Lack of lymphatic vascular specificity of vascular endothelial growth factor receptor 3 in 185 vascular tumors. Cancer 1999, 86, 2406–2412.
  65. Kubo, H.; Fujiwara, T.; Jussila, L.; Hashi, H.; Ogawa, M.; Shimizu, K.; Awane, M.; Sakai, Y.; Takabayashi, A.; Alitalo, K.; et al. Involvement of vascular endothelial growth factor receptor-3 in maintenance of integrity of endothelial cell lining during tumor angiogenesis. Blood 2000, 96, 546–553.
  66. Valtola, R.; Salven, P.; Heikkilä, P.; Taipale, J.; Joensuu, H.; Rehn, M.; Pihlajaniemi, T.; Weich, H.; Dewaal, R.; Alitalo, K. VEGFR-3 and Its Ligand VEGF-C Are Associated with Angiogenesis in Breast Cancer. Am. J. Pathol. 1999, 154, 1381–1390.
  67. Niki, T.; Iba, S.; Yamada, T.; Matsuno, Y.; Enholm, B.; Hirohashi, S. Expression of vascular endothelial growth factor receptor 3 in blood and lymphatic vessels of lung adenocar-cinoma. J. Pathol. 2001, 193, 450–457.
More
This entry is offline, you can click here to edit this entry!
Video Production Service