Next Article in Journal
A Functional Genomics Review of Non-Small-Cell Lung Cancer in Never Smokers
Previous Article in Journal
Human lncRNA SUGCT-AS1 Regulates the Proinflammatory Response of Macrophage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors

by
Syeda Mahak Zahra Bokhari
and
Peter Hamar
*
Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(17), 13317; https://doi.org/10.3390/ijms241713317
Submission received: 6 July 2023 / Revised: 17 August 2023 / Accepted: 23 August 2023 / Published: 28 August 2023
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
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. In this review, we summarized the angiogenic role of VEGF-D in blood vasculogenesis and its targetability as an antiangiogenic therapy in cancer.

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] (Figure 1).

1.1. VEGF-A and Its Receptors

VEGF-A, also often referred to as VEGF, and its isoforms bind VEGFR-1 and VEGFR-2 and regulate the differentiation and development of the vascular system [7]. VEGF-A is an essential protein for embryonic development and angiogenesis in adults. VEGF-A can exist both in soluble and matrix-bound isoforms. The soluble isoform stimulates vessel enlargement, whereas the matrix-bound isoform stimulates vascular branching [13]. VEGF-A activation of VEGFR-2 leads to EC mitogenesis and enhanced vascular permeability [14]. The indispensability of VEGF-A in vasculogenesis was proven as genetic deletion of VEGF-A resulted in the disruption of vascular development and, consequently, early embryonic lethality in mice [15]. VEGF-A produced by normal ECs leads to vascular homeostasis in an autocrine fashion, while VEGF-A production by cancer cells leads to vascular sprouting and abnormal tumor angiogenesis in an autocrine fashion [16].
Although VEGF-A can bind both VEGFR-1 and VEGFR-2, Canonical angiogenesis is achieved by the activation of VEGFR-2, not VEGFR-1. The mechanism of VEGFR-1 signaling is not well understood, as during embryonic development, VEGFR-1 acts as a decoy and keeps VEGF-A from binding VEGFR-2. This decoy action of VEGFR-1 fine-tunes the actions of VEGF-A on stalk cells of growing blood vessels and by recruiting pericytes for vascular stability [17]. In adult life, VEGFR-1 activation by VEGF-A, PIGF, or VEGF-B stimulates inflammation by attuning activation and migration of macrophage cells [18,19]. Likewise, in ischemic and tumor tissues, VEGFR-1 can be activated by VEGF-A and promote angiogenesis, extracellular matrix invasion, and immune evasion [20,21]. The physiologic role of VEGFR-2 is the modulation of signaling pathways leading to EC proliferation, survival, migration, vascular permeability, and angiogenesis [22]. In tumor cells, VEGFR-2 activation promotes VECs’ proliferation and recruitment. Under hypoxic conditions, tumor cells produce several growth factors that influence angiogenesis, including VEGFs (mainly VEGF-A and VEGF-D) that activate VEGFR-2 and mediate angiogenesis to counteract oxygen shortage in the tumor core [20,23,24].
Furthermore, the interaction of VEGF-A with VEGFR-2 is facilitated by NeuroPilin (NP) [25]. NP 1,2 are small membrane glycoproteins originally identified in neurons with pleiotropic functions [26]. They regulate the modulation of growth factor signaling in various processes, such as development, angiogenesis (binding VEGFs), lymphangiogenesis, Immune regulation, cardiovascular disease, and cancer progression, including tumor angiogenesis, invasion, metastasis, and therapy resistance [27,28,29,30,31,32]. They are also involved in enhancing other VEGF family members’ interaction and activation of corresponding receptors [11,25,33,34].

1.2. VEGF-C and Its Receptors

All members of the VEGF family share a VEGF homology domain (VHD). The VHD is the central domain of all VEGF family members that binds the receptor. However, there are amino acid-based structural differences and additional domains that affect the VHD’s binding to the receptor. Two members of the family VEGF-C and VEGF-D have additional N- and C- terminal domains flanking the VHD and are secreted as preproproteins, later proteolytically cleaved to their mature form [8,11,12]. The flanking domains have complex modulatory roles. For example, like mature VEGF-C, prepro VEGF-C can bind VEGFR-3, but it cannot activate the receptor, thereby acting as a competitive blocker. On the other hand, VEGF-C, with its C terminal propeptide processed, can activate the Neuropilin (NP-1) co-receptor, promoting lymphangiogenesis [11,35,36]. The proteolytic cleavage of VEGF-C to its mature form can be initiated by ADAMTS3, plasmin, Cathepsin D, thrombin, and Prostate Specific Antigen (PSA) [11,37,38,39]. Different proteases can result in different mature forms of VEGF-C or VEGF-D, leading to different receptor affinity and activation potentials [11].
Although both VEGF-C and VEGF-D can bind and activate VEGFR-3, VEGF-C is the main member associated with lymphangiogenesis. The role of VEGF-D is much more subtle in lymphangiogenesis as compared to VEGF-C, as VEGF-C is crucial for embryonic lymphangiogenic growth, but VEGF-D is rather redundant. VEGF-D knockout mice experience no significant phenotypical alteration and undergo normal lymphangiogenesis [40]. Thus, the role of VEGF-C is better characterized as a lymphangiogenic factor. Nevertheless, VEGF-D can initiate lymphangiogenic sprouting in the absence of VEGF-C [41]. It, therefore, has a supporting or compensatory role in lymphangiogenesis by activating VEGFR-3.
Upon binding and activating VEGFR-3, VEGF-C promotes proliferation, migration, and survival of LECs and initiates lymphatic vessel sprouting from large veins [42,43]. The indispensability of VEGF-C was confirmed by knockout studies when VEGF-C null mice encountered pre-natal death owing to a lack of lymphatic development [42]. VEGF-C has also demonstrated a pro-lymphangiogenic role in cancer. Human breast carcinoma cells with high VEGF-C expression promoted tumor lymphangiogenesis and, hence, metastasis in a mouse model; however, this spread could be halted by trapping VEGF-C with soluble VEGFR-3 [44]. High VEGF-C expression can facilitate metastasis by increasing the overall number of lymphatic vessels and the interaction between tumor cells and LECs, thereby promoting the entry of tumor cells into the lymph vessels [45,46].

1.3. VEGF-D and Its Receptors

Vascular endothelial growth factor-D (VEGF-D) is a secreted glycoprotein that, according to its first descriptions [47], binds and activates VEGFR-3, promoting the proliferation of lymphatic endothelial cells and hence promoting lymphangiogenesis. It was identified first as a mitogenic growth factor, secreted as an effector protein of the proto-oncogene c-fos, which is an early response transcription factor involved in cell proliferation and differentiation in cultured fibroblasts. Hence, VEGF-D was originally named c-Fos-Induced Growth Factor (FIGF) [48]. In murine embryos, VEGF-D was expressed in all major organs, including the kidney, lungs, adrenal glands, vertebral column, etc. [49]. Soon after its discovery in mice, VEGF-D was characterized in humans. In healthy humans, VEGF-D expression has been demonstrated in the lungs, heart, skeletal muscle, colon, and small intestine [50].
Human VEGF-D has 85% sequence homology to its mouse counterpart. There are structural similarities with VEGF-C (48%) and, to a lesser extent, with VEGF-A (31%). As already mentioned, VEGF-D is secreted as a preproprotein, which is cleaved to its active form. Different proteases can produce different cleaved products with unique receptor-binding abilities and functions. Similarly, there is a partially processed VEGF-D form with the C-terminal flanking peptide removed, but the N-terminal flanking peptide is still attached via disulfide bridges [51]. Classically, VEGF-D is associated with lymphangiogenesis by activation of VEGFR-3 [52]. However, VEGF-D has the ability to activate various receptors and mediate complex crosstalk between them (Figure 2). Mature VEGF-D is a non-covalent dimer, which has a 290- and 40-fold higher affinity to VEGFR-2 and VEGFR-3, respectively, as compared to its long unprocessed form [47,53]. The inability to achieve the proteolytic cleavage renders VEGF-D ineffective as an angiogenic protein as the unprocessed form cannot bind VEGFR-2 [12,38]. Mature VEGF-D induces both angiogenesis and lymphangiogenesis in vitro and in vivo via binding and activating VEGFR-2 or VEGFR-3, depending on the availability of ECs expressing one or the other receptor [54].
Interestingly, VEGF-D is the only member of the family that has different receptor affinity in mouse and human species. VEGF-D binds and activates VEGFR-3 in mice while activating both VEGFR-2 and VEGFR-3 in humans. However, in mice, VEGF-D induces blood vessel proliferation by activating VEGFR-3 instead of VEGFR-2 [55]. Thus, despite the differences in receptor affinity, the physiologic effects (stimulation of both angio- and lymphangiogenesis) are similar. However, owing to the presence of VEGFR-3 in the blood vascular endothelial cells, VEGF-D can promote angiogenesis via VEGFR-3 activation as well [56]. Like VEGF-A, VEGF-D induces the proliferation of vascular endothelial cells, promoting the growth of VEGFR-2 positive blood vessels. The response is strong but delayed [57], which can explain why VEGF-D has not been classically affiliated with blood vessel angiogenesis. There might be a feedback loop where different receptors are upregulated in response to ligand binding; hence, VEGF members have a cooperative role in vasculogenesis.
VEGF-D also has the ability to induce VEGFR-2/3 hetero-dimerization in both vascular and lymphatic endothelial cells [58]. ProVEGF-D, with either its N or C terminal removed, has the ability to initiate VEGFR-2/VEGFR-3 heterodimerization [59]. As the heterodimer has an altered trans-phosphorylation tyrosine residue in the VEGFR-3 unit, the binding of partially or fully processed VEGF-D elicits distinct signaling and responses [59]. VEGFR-2/VEGFR-3 heterodimer activation can promote sprouting angiogenesis [58]. This ability of partially processed and fully mature VEGF-D to activate VEGFR2/VEGFR-3 heterodimer could be one of the signaling mechanisms by which VEGF-D can induce angiogenesis.
Both mature and C-terminal proVEGF-D have the ability to bind NeuroPilin (NP) NP-1 and NP-2 [59]. Upon activation by VEGF-D, VEGFR-3 is co-localized and internalized with NP-2, which indicates that NP-2 acts as a facilitating co-receptor in VEGF-D mediated signaling [25,33,34]. VEGF-D can modulate angiogenesis in an NP-1-dependent fashion as well, with NP-1 either acting as a co-receptor for efficient receptor activation or by NP-1-mediated phosphorylation of downstream VEGFR-2 effectors in breast and other cancers [60,61]. We can, therefore, infer that the angiogenic signaling mediated by VEGF-D could be NP-dependent.
Figure 2. VEGF-D mediated angiogenic signaling in lymphatic or blood capillary endothelial cells. PreproVEGF-D can bind VEGFR-3 and activate lymphangiogenesis. However, proteolytic cleavage of the preproVEGF-D provides mature VEGF-D, which can bind and activate VEGFR-3 but also VEGFR-2 and induce VEGFR-2/3 heterodimerization [47,58,59]. Created with BioRender.com accessed on 27 July 2023.
Figure 2. VEGF-D mediated angiogenic signaling in lymphatic or blood capillary endothelial cells. PreproVEGF-D can bind VEGFR-3 and activate lymphangiogenesis. However, proteolytic cleavage of the preproVEGF-D provides mature VEGF-D, which can bind and activate VEGFR-3 but also VEGFR-2 and induce VEGFR-2/3 heterodimerization [47,58,59]. Created with BioRender.com accessed on 27 July 2023.
Ijms 24 13317 g002

1.4. Other—Less Well Characterized—Members of the VEGF-Family

VEGF-B was first described in the 1990s. It is expressed mainly in muscle tissue of the heart and skeletal muscle. VEGF-B has two isoforms which can bind and activate VEGFR-1. It mainly maintains new blood vessels and prevents vascular leaking in the heart. Other physiological sites of expression include renal tubular epithelial cells [62]. VEGF-B, unlike other members of its family, is the least angiogenic factor [10,63]. However, its angiogenic potential plays a role in cardiac ischemia, where VEGF-B stimulates angiogenesis by displacing VEGF-A from VEGFR-1 [64,65]. Furthermore, a stronger expression of VEGF-B was observed in human and rodent tumor cells [64].
VEGF-E expressed in the Orf Parapox virus has a 25% amino acid homology to mammalian VEGF-A. The virus typically infects goats and sheep; however, it can occasionally infect humans and cause lesions [66]. In humans, VEGF-E has mitogenic effects on ECs and can enhance vascular permeability by activating VEGFR-2, similar to VEGF-A [67]. Its role in cancer is not characterized; however, its high angiogenic ability can be of therapeutic value in cardiovascular diseases [68].
In 1998, Komori isolated VEGF-F from the venom of Vipera aspis aspis (Aspic Viper), hence also termed snake venom VEGF [69]. Although not found in the human body, its potent ability to induce vascular permeability and proliferation of endothelial cells is being investigated for therapeutic interventions [69,70].
Placental Growth Factor (PIGF) expression in normal tissues is low or undetectable, and it does not affect angiogenesis directly. PIGF is important in facilitating VEGF-A binding to VEGFR-2 by binding VEGFR-1 and preventing the decoy action of VEGFR-1 [71]. However, PIGF expression is upregulated in cancer, correlating to progressive tumor grade and stage. PIGF promotes angiogenesis by activating the VEGFR1/VEGFR-2 heterodimer by dimerizing with VEGF-A itself [72].

2. Lymphangiogenesis and Angiogenesis, the Paths Cross?

Lymphangiogenesis is essentially a form of angiogenesis in which the vessels usually originate from large veins [73]. Much like blood vascular angiogenesis, lymphangiogenesis requires the activation of endothelial cells for proliferation. However, the triggers for this EC activation differ. As stated previously, blood vessel angiogenesis is usually triggered by a lack of oxygen supply, while lymph vessel growth is initiated in response to the interstitial pressure in normal development and inflammation in pathological cases [74]. Lymphatic Endothelial Cells (LECs) have an oak leaf-like morphology and are connected to each other with overlaps; however, LECs of the collecting lymphatic ducts have smooth and elongated morphology with tight junctions between cells [75].
Physiologically, lymphatic vessels differ from normal blood vessels as they lack tight junctions, and the LECs are connected by overlaps that can mechanically open to cope with increased interstitial pressure. Furthermore, lymph vessels lack a complete basement membrane [73,76]. Notably, this physiological profile is like the physiology of tumor vasculature [77,78,79,80,81].
In cancer, angiogenesis and lymphangiogenesis rely on a complex interplay of secreted growth factors and cytokines, a potential crosstalk between the two processes [82]. Canonically, LECs have a high expression of VEGFR-3, and their activation by VEGF-C or VEGF-D initiates and maintains lymphangiogenesis [83]. However, VEGF-A can also contribute to lymphangiogenesis by activating VEGFR2 and/or VEGFR2/VEGFR3 heterodimer and indirectly by recruiting VEGF-C and VEGF-D secreting macrophages and mast cells [84]. However, the contribution of VEGF-A is negligible compared to VEGF-C/D in inducing lymphangiogenesis [83].
It is possible that the pathways cross or mimic each other in cancer angiogenesis, although yet unidentified, exploring this relationship might open new directions to cancer metastasis and drug delivery research.

3. 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 [85]. Furthermore, the autocrine production of growth factors and cytokines by the active ECs promotes tumor progression [86]. 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) [24,87]. 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 [87,88] endothelial cell migration [89], and EC proliferation [90,91,92]. 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 [88]. The new ECs establish tight junctions, and hence, a luminal vessel is produced [93,94]. The newly formed vessels are then covered by tumor-recruited pericytes, which provide support to the vessels from outside (maturation step) (Figure 3) [95]. 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) [96,97]. 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 [98]. 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 [24,99].
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 [101]. 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 [102]. 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 [103,104].

3.1. VEGF-D and Blood Capillary Angiogenesis

Like many other types of anticancer treatments, tumor cells developed resistance to antiangiogenic therapies [105,106,107]. 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 [103,107,108,109]. 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 [110]. 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 [111]. 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 [112]. VEGF-D can, therefore, execute all steps of tumor angiogenesis in the absence of VEGF-A with comparable efficiency [91,100] (Figure 3).
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 [41]. Furthermore, patients with nonresectable hepatocellular carcinoma, already receiving bevacizumab, had a higher VEGF-D expression and promoted disease progression [113]. The ability of VEGF-D as an inducer of blood capillary angiogenesis was first characterized in rabbit retinal tissue [114] 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 [115]. 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 [115]. 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 [116]. A similar dose-dependent angiogenic response was observed in pig hearts [117]. VEGF-D also induced angiogenesis in the brain after blood–brain barrier breakdown by activating both VEGFR-2 and VEGFR-3 [118]. VEGF-D modulates angiogenesis by inducing EC proliferation and migration to branching points and facilitating tube formation (Figure 3) [116,119,120]. Based on these findings, VEGF-D expressed by tumor cells could be a way for cancer to circumvent VEGF-A targeting antiangiogenic therapy [121,122,123]. 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 [124]; 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.

3.2. 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 [104], essentially maintaining the vascular system in development and disease [23]. 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 [103], little is known about the signaling pathways and expression regulators involved. Here, we are mentioning data available to date about VEGF-D signaling in angiogenesis.
One of the main factors promoting VEGF-D expression in response to antiangiogenic therapy would be therapy-related hypoxia [125]. 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 [126]. 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 [55,127,128]. 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 [129] and invasive cervical carcinoma [130]. Furthermore, VEGF-D could induce proangiogenic phenotype in HUVECs by upregulating genes involved in matrix modulation and cell membrane alteration in an autocrine manner [131]. 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 [132]. 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 [133]. Based on these findings, we believe the autocrine action of VEGF-D might be stronger in executing angiogenic response and a potent target to manage antiangiogenic drug resistance.
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 [134]. 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 [135,136,137,138]. It would be interesting to investigate how VEGF-D/VEGFR-3 signaling is stimulating angiogenesis.

4. Clinical Significance of VEGF-D in Tumor Angiogenesis

Based on Cancer Protein Atlas data, VEGF-D expression is prevalent in malignant melanomas, urothelial, gastric, and pancreatic cancers [139]. VEGF-D expression is clinically detected by its serum and plasma levels by Enzyme-linked immune-sorbent assay (ELISA), multiplex assays such as Luminex technology [140], or by immunohistochemistry in resected tumor tissue. Although the mechanistic basis of VEGF-D signaling in cancer angiogenesis has not been fully elucidated, patient data analysis has indicated its significance in cancer progression and prognosis. In lung cancer, IL-6 promoted tube formation by enhancing VEGF-D expression [141]. Similarly, in invasive ductal breast carcinoma, VEGF-D but not VEGF-A or VEGF-C was upregulated in tumor tissue and promoted angiogenesis [142]. Cell lines established from Tumor-derived Endothelial Cells (TEC) from renal carcinoma patients expressed VEGF-D and VEGFR-2 but not VEGF-A compared to normal human mammary epithelial cells in vitro. Moreover, TEC implants in matrigel in SCID mice confirmed VEGF-D expressing TECs in vivo [143]. In pediatric hepatoblastoma and hepatocellular carcinoma, VEGF-D secreted by the reactive ductules (cells with a ductular phenotype that proliferate and accumulate in response to liver damage) was associated with a higher number of CD34+ (a marker of proliferating endothelial cells), blood vessels compared to low VEGF-D expressing hepatic tumors [144]. Therefore, the unique receptor expression of tumor vasculature and VEGF-D’s ability to activate more than one of them makes it a prominent regulator of blood vessel angiogenesis. Furthermore, these results are indicative of VEGF-D’s ability to circumvent antiangiogenic therapy and promote cancer survival and progression.
Clinical and pathological data established the role of VEGF-D in cancer, as high VEGF-D expression is linked to poor outcomes. Increased VEGF-D expression is an independent negative prognostic marker in ovarian carcinoma, colorectal cancer, breast cancer, and gastric carcinoma [145,146,147,148,149]. Furthermore, in different grades of endometrial cancer, a higher VEGF-D expression was observed in correlation to a higher tumor grade [150]. Similarly, a pilot study reported an increase in VEGF-D in early epithelial ovarian tumors, potentiating its role as an early-detection biomarker [151]. Clinical studies for metastatic colorectal cancer (mCRC) revealed that targeting VEGF-A with bevacizumab upregulated VEGF-D expression, explaining a poor response to bevacizumab [152,153]. However, mCRC patients with high VEGF-D expression, when treated with VEGFR-2 inhibitor ramucirumab, had a higher overall (OS) and progression-free survival (PFS) [154,155]. Higher VEGF-D expression also translated to poor prognostic significance and a 3-fold higher death risk in CRC patients [156,157,158]. In another clinical trial investigating mCRC in bevacizumab-treated patients, high plasma VEGF-D was associated with poor clinical outcomes [159]. Very recently, a phase II clinical study also showed similar results when a VEGFR inhibitor, Dovitinib, increased plasma VEGF-D levels [160].
Bioinformatics analysis of patient data from cancer databases of hepatocellular carcinoma (HCC) samples identified VEGF-D as a negative prognostic angiogenic marker [134,161]. VEGF-D was also upregulated in response to radiotherapy in lung cancer and was interpreted as an angiogenic biomarker [162]. Taniguchi and his colleagues’ clinical trial also reported that VEGF-D expression could be induced by Bevacizumab treatment; hence, its baseline expression can help choose a better antiangiogenic treatment regimen [163]. Similarly, a clinical trial investigated the efficacy of VEGF-D as a predictive biomarker prior to initiation and during second-line treatment with paclitaxel and ramucirumab in GC patients [164]. Furthermore, another clinical trial found a higher PFS in mCRC patients with lower VEGF-D compared to patients with high VEGF-D plasma levels when treated with ramucirumab and no previous bevacizumab therapy [140]. There is substantial evidence that VEGF-D has significant clinical relevance in antiangiogenic therapy. However, the mechanisms upregulating VEGF-D expression and downstream signaling activated by VEGF-D are still unclear. We believe a thorough insight into how VEGF-D steps up as an angiogenic mediator is an interesting research question with substantial translational potential.

5. Current VEGF-D Therapies

Despite its importance, there is currently no specific inhibitor of VEGF-D. Reasons include:
(1)
The structural similarity between VEGF-D, VEGF-C, and VEGF-A with similar receptor-binding domain (VHD);
(2)
The complex VEGF-D signaling through VEGFR-2, VEGFR-3, and NRP-1;
(3)
The multiplex proteolytic cleavage of VEGF-D by different proteases providing different isoforms with variable structures.
Furthermore, since the role of VEGF-D has been established in lymphangiogenesis and is deemed secondary in tumor blood neoangiogenesis, less attention has been devoted to the development of specific VEGF-D inhibitors.
VEGF-D can be directly targeted by monoclonal antibodies (mAb). Achen et al. generated antibodies against the VHD of various VEGF-D isoforms. VD-1 effectively reduced the interaction of human VEGF-D with both VEGFR-2 and VEGFR-3 and inhibited microvascular epithelial cells’ mitotic response to VEGF-D [165]. Further studies by Davydova et al. included protein mapping of the epitope binding site of VD-1 and reported that VD-1 binds to and inhibits a specific amino acid region ‘147NEESL151’ of VEGF-D to neutralize its action. However, a mutation in this particular amino acid sequence renders VD-1’s activity ineffective [166]. They also compared the effectivity of VD-1 with commercially available mAb-286, which binds the alpha helix structure of mature VEGF-D, and demonstrated that, like VD-1, mAb-286 is also ineffective in inhibiting receptor binding action of all VEGF-D variants [167]. It is, therefore, difficult to design an antibody that would neutralize all pre and post-translational variants of VEGF-D. Thus, further current treatment modalities include targeting VEGF-D inducers, such as c-fos, or inhibition of VEGF-D’s receptor interaction and/or receptor activation.
Ji and colleagues applied andrographolide, a plant-based naturally occurring diterpenoid lactone, which reduced c-fos expression and nuclear translocation, thereby resulting in reduced VEGF-D expression and angiogenesis in hepatoma cancer cells [168]. Inactivation of VEGF-D function by hindering receptor interaction and activation is another widely used approach that has yielded positive results. VGX-300: a soluble VEGFR-3 successfully reduced VEGF-D activity in age-related macular degeneration and diabetic macula edema of the eye in a rat model [169,170]. Similarly, a monoclonal antibody against VEGFR-3, mF4-3C1, could reduce lymphatic regeneration in mice [171], while a humanized antibody hF4-3C5 could reduce tube formation in response to VEGF-D by 60% in bovine aortic endothelial cells [172]. More recently, in a clinical trial, promising results were observed in age-related macular degeneration neovascularization by using VEGF-C/VEGF-D inhibitor OPT-302 along with VEGF-A inhibitor ranibizumab. OPT-302 is a trapping molecule composed of VEGFR-3 ligand binding domains that bind and reduce the availability of VEGF-C and VEGF-D for the endogenous VEGFR-3 receptor [173]. An ongoing clinical trial is aiming at evaluating the safety and pharmacological properties of OPT-302 in advanced neovascular age-related macular degeneration [174]. Similarly, monoclonal antibodies such as sorafenib, ramucirumab, etc., block the VEGFR-2 ligand interaction and can be used to target VEGF-A or VEGF-D mediated signaling. These VEGFR-2 mAbs reduce VEGF-D’s ability to activate VEGFR-2 and, thus, can inhibit the angiogenic role of VEGF-D. Tanibirumab, a humanized antibody against the VEGF binding domain of VEGFR-2, was very successful in blocking the receptor activation by VEGF-C and VEGF-D with fewer adverse effects [175]. Currently, two clinical trials are investigating the safety and tolerability of Tanibirumab in advanced metastatic cancer and recurrent glioblastoma [176,177]. These approaches, although promising, are nonspecific for VEGF-D and thus are not suitable to dissect the relative roles of VEGF-A or VEGF-D. The antibiotic salinomycin effectively reduced VEGF-D expression in endometrial cancer cells, as reported recently in 2021 [178]. Similarly, a naturally occurring small compound used in Chinese medicine, norcantharidin, also effectively reduced VEGF-D mRNA production [179].
The tumor vasculature development has a complex regulation and, thus, offers several points to interfere with tumor development, but requires a carefully crafted treatment regimen to halt the potential crosstalk between VEGF family members to have maximal effects. The most used treatment modalities at present to reduce VEGF-D activity are summarized in Table 1.

6. Conclusions

VEGF-A is well-established as a regulator of cancer angiogenesis, and substantial clinical evidence has accumulated already with approved inhibitors supporting anti-VEGF-A therapy in cancer treatment. There is accumulating evidence that VEGF-D, besides its role in regulating lymphangiogenesis, is also an important angiogenic factor, and can mediate angiogenesis in a manner comparable to VEGF-A. Although initially, VEGF-D stepping up as an angiogenic factor was detected in response to VEGF-A inhibition, later findings established VEGF-D as an angiogenesis biomarker in cancers independent of VEGF-A inhibition. Furthermore, VEGF-D may have direct effects on cancer cell proliferation, rendering it a primary anticancer therapeutic target. Substantial clinical evidence supports the therapeutic potential of VEGF-D inhibition; however, the lack of specific inhibitors at present prevents the dissection of the sole role of VEGF-D from the complex inhibition of the VEGF family by these inhibitors.

Author Contributions

Conceptualization, S.M.Z.B. and P.H.; writing—original draft preparation, S.M.Z.B.; review and editing, S.M.Z.B. and P.H.; supervision, P.H. All authors have read and agreed to the published version of the manuscript.

Funding

Semmelweis Science and Innovation Fund: Semmelweis NKFIH-OTKA Átsegítő Támogatás (STIA-OTKA-2022) to P.H.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Folkman, J. Patterns and Emerging Mechanisms of the Angiogenic Switch during Tumorigenesis. Cell 1996, 86, 353–364. [Google Scholar] [CrossRef]
  2. Ruoslahti, E. Specialization of tumour vasculature. Nat. Rev. Cancer 2002, 2, 83–90. [Google Scholar] [CrossRef]
  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. [Google Scholar]
  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. [Google Scholar] [CrossRef]
  5. Miron, L.; Gafton, B.; Marinca, M. Angiogeneza Tumorală-Implicaţii în Terapia Cancerelor. J. Chir. 2010, 6, 2. [Google Scholar]
  6. La Mendola, D.; Trincavell, M.L.; Martini, C. Angiogenesis in Disease. Int. J. Mol. Sci. 2022, 23, 10962. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  10. Li, X.; Lee, C.; Tang, Z.; Zhang, F.; Arjunan, P.; Li, Y.; Hou, X.; Kumar, A.; Dong, L. VEGF-B: A survival, or an angiogenic factor? Cell Adhes. Migr. 2009, 3, 322–327. [Google Scholar] [CrossRef]
  11. Künnapuu, J.; Bokharaie, H.; Jeltsch, M. Proteolytic Cleavages in the VEGF Family: Generating Diversity among Angiogenic VEGFs, Essential for the Activation of Lymphangiogenic VEGFs. Biology 2021, 10, 167. [Google Scholar] [CrossRef] [PubMed]
  12. Harris, N.C.; Paavonen, K.; Davydova, N.; Roufail, S.; Sato, T.; Karnezis, T.; Stacker, S.A.; Achen, M.G.; Zhang, Y.-F. Proteolytic processing of vascular endothelial growth factor-D is essential for its capacity to promote the growth and spread of cancer. FASEB J. 2011, 25, 2615–2625. [Google Scholar] [CrossRef]
  13. Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef]
  14. De Smet, F.; Segura, I.; De Bock, K.; Hohensinner, P.J.; Carmeliet, P. Mechanisms of vessel branching: Filopodia on endothelial tip cells lead the way. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 639–649. [Google Scholar] [CrossRef] [PubMed]
  15. Thurston, G.; Kitajewski, J. VEGF and Delta-Notch: Interacting signalling pathways in tumour angiogenesis. Br. J. Cancer 2008, 99, 1204–1209. [Google Scholar] [CrossRef]
  16. Majidpoor, J.; Mortezaee, K. Angiogenesis as a hallmark of solid tumors—Clinical perspectives. Cell. Oncol. 2021, 44, 715–737. [Google Scholar] [CrossRef]
  17. Uemura, A.; Fruttiger, M.; D’Amore, P.A.; De Falco, S.; Joussen, A.M.; Sennlaub, F.; Brunck, L.R.; Johnson, K.T.; Lambrou, G.N.; Rittenhouse, K.D.; et al. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog. Retin. Eye Res. 2021, 84, 100954. [Google Scholar] [CrossRef]
  18. Shibuya, M. Vascular endothelial growth factor receptor-1 (VEGFR-1/Flt-1): A dual regulator for angiogenesis. Angiogenesis 2006, 9, 225–230. [Google Scholar] [CrossRef]
  19. Lacal, P.M.; Graziani, G. Therapeutic implication of vascular endothelial growth factor receptor-1 (VEGFR-1) targeting in cancer cells and tumor microenvironment by competitive and non-competitive inhibitors. Pharmacol. Res. 2018, 136, 97–107. [Google Scholar] [CrossRef]
  20. Ceci, C.; Atzori, M.G.; Lacal, P.M.; Graziani, G. Role of VEGFs/VEGFR-1 Signaling and Its Inhibition in Modulating Tumor Invasion: Experimental Evidence in Different Metastatic Cancer Models. Int. J. Mol. Sci. 2020, 21, 1388. [Google Scholar] [CrossRef]
  21. Mabeta, P.; Steenkamp, V. The VEGF/VEGFR Axis Revisited: Implications for Cancer Therapy. Int. J. Mol. Sci. 2022, 23, 15585. [Google Scholar] [CrossRef] [PubMed]
  22. Siveen, K.S.; Prabhu, K.; Krishnankutty, R.; Kuttikrishnan, S.; Tsakou, M.; Alali, F.Q.; Dermime, S.; Mohammad, R.M.; Uddin, S. Vascular Endothelial Growth Factor (VEGF) Signaling in Tumour Vascularization: Potential and Challenges. Curr. Vasc. Pharmacol. 2017, 15, 339–351. [Google Scholar] [CrossRef]
  23. 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. [Google Scholar] [CrossRef] [PubMed]
  24. Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [PubMed]
  25. Kawasaki, T.; Kitsukawa, T.; Bekku, Y.; Matsuda, Y.; Sanbo, M.; Yagi, T.; Fujisawa, H. A requirement for neuropilin-1 in embryonic vessel formation. Development 1999, 126, 4895–4902. [Google Scholar] [CrossRef]
  26. Guo, H.-F.; Vander Kooi, C.W. Neuropilin Functions as an Essential Cell Surface Receptor. J. Biol. Chem. 2015, 290, 29120–29126. [Google Scholar] [CrossRef]
  27. Parker, M.W.; Guo, H.-F.; Li, X.; Linkugel, A.D.; Kooi, C.W.V. Function of Members of the Neuropilin Family as Essential Pleiotropic Cell Surface Receptors. Biochemistry 2012, 51, 9437–9446. [Google Scholar] [CrossRef]
  28. Peng, K.; Bai, Y.; Zhu, Q.; Hu, B.; Xu, Y. Targeting VEGF–neuropilin interactions: A promising antitumor strategy. Drug Discov. Today 2018, 24, 656–664. [Google Scholar] [CrossRef]
  29. Islam, R.; Mishra, J.; Bodas, S.; Bhattacharya, S.; Batra, S.K.; Dutta, S.; Datta, K. Role of Neuropilin-2-mediated signaling axis in cancer progression and therapy resistance. Cancer Metastasis Rev. 2022, 41, 771–787. [Google Scholar] [CrossRef]
  30. Tammela, T.; Zarkada, G.; Wallgard, E.; Murtomäki, A.; Suchting, S.; Wirzenius, M.; Waltari, M.; Hellström, M.; Schomber, T.; Peltonen, R.; et al. Blocking VEGFR-3 suppresses angiogenic sprouting and vascular network formation. Nature 2008, 454, 656–660. [Google Scholar] [CrossRef]
  31. Xu, Z.; Goel, H.L.; Burkart, C.; Burman, L.; Chong, Y.E.; Barber, A.G.; Geng, Y.; Zhai, L.; Wang, M.; Kumar, A.; et al. Inhibition of VEGF binding to neuropilin-2 enhances chemosensitivity and inhibits metastasis in triple-negative breast cancer. Sci. Transl. Med. 2023, 15, eadf1128. [Google Scholar] [CrossRef]
  32. Goel, H.L.; Mercurio, A.M. VEGF targets the tumour cell. Nat. Rev. Cancer 2013, 13, 871–882. [Google Scholar] [CrossRef] [PubMed]
  33. Yuan, L.; Moyon, D.; Pardanaud, L.; Bréant, C.; Karkkainen, M.J.; Alitalo, K.; Eichmann, A. Abnormal lymphatic vessel development in neuropilin 2 mutant mice. Development 2002, 129, 4797–4806. [Google Scholar] [CrossRef] [PubMed]
  34. Karpanen, T.; Heckman, C.A.; Keskitalo, S.; Jeltsch, M.; Ollila, H.; Neufeld, G.; Tamagnone, L.; Alitalo, K. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors. FASEB J. 2006, 20, 1462–1472. [Google Scholar] [CrossRef]
  35. Chen, J.-C.; Chang, Y.-W.; Hong, C.-C.; Yu, Y.-H.; Su, J.-L. The Role of the VEGF-C/VEGFRs Axis in Tumor Progression and Therapy. Int. J. Mol. Sci. 2012, 14, 88–107. [Google Scholar] [CrossRef]
  36. Scavelli, C.; Vacca, A.; Di Pietro, G.; Dammacco, F.; Ribatti, D. Crosstalk between angiogenesis and lymphangiogenesis in tumor progression. Leukemia 2004, 18, 1054–1058. [Google Scholar] [CrossRef]
  37. McColl, B.K.; Baldwin, M.E.; Roufail, S.; Freeman, C.; Moritz, R.L.; Simpson, R.J.; Alitalo, K.; Stacker, S.A.; Achen, M.G. Plasmin Activates the Lymphangiogenic Growth Factors VEGF-C and VEGF-D. J. Exp. Med. 2003, 198, 863–868. [Google Scholar] [CrossRef]
  38. McColl, B.K.; Paavonen, K.; Karnezis, T.; Harris, N.C.; Davydova, N.; Rothacker, J.; Nice, E.C.; Harder, K.W.; Roufail, S.; Hibbs, M.L.; et al. Proprotein convertases promote processing of VEGF-D, a critical step for binding the angiogenic receptor VEGFR. FASEB J. 2007, 21, 1088–1098. [Google Scholar] [CrossRef]
  39. Jha, S.K.; Rauniyar, K.; Chronowska, E.; Mattonet, K.; Maina, E.W.; Koistinen, H.; Stenman, U.H.; Alitalo, K.; Jeltsch, M. KLK3/PSA and cathepsin D activate VEGF-C and VEGF-D. eLife 2019, 8, e44478. [Google Scholar] [CrossRef]
  40. Baldwin, M.E.; Halford, M.M.; Roufail, S.; Williams, R.A.; Hibbs, M.L.; Grail, D.; Kubo, H.; Stacker, S.A.; Achen, M.G. Vascular Endothelial Growth Factor D Is Dispensable for Development of the Lymphatic System. Mol. Cell Biol. 2005, 25, 2441–2449. [Google Scholar] [CrossRef]
  41. 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. [Google Scholar] [CrossRef] [PubMed]
  42. Karkkainen, M.J.; Haiko, P.; Sainio, K.; Partanen, J.; Taipale, J.; Petrova, T.V.; Jeltsch, M.; Jackson, D.G.; Talikka, M.; Rauvala, H.; et al. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from em-bryonic veins. Nat. Immunol. 2004, 5, 74–80. [Google Scholar] [CrossRef]
  43. Wang, C.-A.; Tsai, S.-J. The non-canonical role of vascular endothelial growth factor-C axis in cancer progression. Exp. Biol. Med. 2015, 240, 718–724. [Google Scholar] [CrossRef]
  44. Mattila, M.M.-T.; Ruohola, J.K.; Karpanen, T.; Jackson, D.G.; Alitalo, K.; Härkönen, P.L. VEGF-C induced lymphangiogenesis is associated with lymph node metastasis in orthotopic MCF-7 tumors. Int. J. Cancer 2002, 98, 946–951. [Google Scholar] [CrossRef]
  45. Saharinen, P.; Tammela, T.; Karkkainen, M.J.; Alitalo, K. Lymphatic vasculature: Development, molecular regulation and role in tumor metastasis and inflammation. Trends Immunol. 2004, 25, 387–395. [Google Scholar] [CrossRef]
  46. Hirakawa, S.; Brown, L.F.; Kodama, S.; Paavonen, K.; Alitalo, K.; Detmar, M. VEGF-C–induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood 2006, 109, 1010–1017. [Google Scholar] [CrossRef] [PubMed]
  47. Achen, M.G.; Jeltsch, M.; Kukk, E.; Mäkinen, T.; Vitali, A.; Wilks, A.F.; Alitalo, K.; Stacker, S.A. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc. Natl. Acad. Sci. USA 1998, 95, 548–553. [Google Scholar] [CrossRef] [PubMed]
  48. Orlandini, M.; Marconcini, L.; Ferruzzi, R.; Oliviero, S. Identification of a c-fos-induced gene that is related to the platelet-derived growth factor/vascular endothelial growth factor family. Proc. Natl. Acad. Sci. USA 1996, 93, 11675–11680. [Google Scholar] [CrossRef]
  49. Avantaggiato, V.; Orlandini, M.; Acampora, D.; Oliviero, S.; Simeone, A. Embryonic expression pattern of the murine figf gene, a growth factor belonging to platelet-derived growth factor/vascular endothelial growth factor family. Mech. Dev. 1998, 73, 221–224. [Google Scholar] [CrossRef]
  50. Yamada, Y.; Nezu, J.-I.; Shimane, M.; Hirata, Y. Molecular Cloning of a Novel Vascular Endothelial Growth Factor, VEGF-D. Genomics 1997, 42, 483–488. [Google Scholar] [CrossRef]
  51. Baldwin, M.E.; Roufail, S.; Halford, M.M.; Alitalo, K.; Stacker, S.A.; Achen, M.G. Multiple forms of mouse vascular endothelial growth factor-D are generated by RNA splicing and proteolysis. J. Biol. Chem. 2001, 276, 44307–44314. [Google Scholar] [CrossRef]
  52. Alitalo, K.; Tammela, T.; Petrova, T.V. Lymphangiogenesis in development and human disease. Nature 2005, 438, 946–953. [Google Scholar] [CrossRef]
  53. Stacker, S.A.; Stenvers, K.; Caesar, C.; Vitali, A.; Domagala, T.; Nice, E.; Roufail, S.; Simpson, R.J.; Moritz, R.; Karpanen, T.; et al. Biosynthesis of Vascular Endothelial Growth Factor-D Involves Proteolytic Processing Which Generates Non-covalent Homodimers. J. Biol. Chem. 1999, 274, 32127–32136. [Google Scholar] [CrossRef]
  54. Byzova, T.V.; Goldman, C.K.; Jankau, J.; Chen, J.; Cabrera, G.; Achen, M.G.; Stacker, S.A.; Carnevale, K.A.; Siemionow, M.; Deitcher, S.R.; et al. Adenovirus encoding vascular endothelial growth factor–D induces tissue-specific vascular patterns in vivo. Blood 2002, 99, 4434–4442. [Google Scholar] [CrossRef]
  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. [Google Scholar] [CrossRef]
  56. Witmer, A.N.; van Blijswijk, B.C.; Dai, J.; Hofman, P.; Partanen, T.A.; Vrensen, G.F.J.M.; Schlingemann, R.O. VEGFR-3 in adult angiogenesis. J. Pathol. 2001, 195, 490–497. [Google Scholar] [CrossRef]
  57. Jia, H.; Bagherzadeh, A.; Bicknell, R.; Duchen, M.R.; Liu, D.; Zachary, I. Vascular Endothelial Growth Factor (VEGF)-D and VEGF-A Differentially Regulate KDR-mediated Signaling and Biological Function in Vascular Endothelial Cells. J. Biol. Chem. 2004, 279, 36148–36157. [Google Scholar] [CrossRef]
  58. Nilsson, I.; Bahram, F.; Li, X.; Gualandi, L.; Koch, S.; Jarvius, M.; Söderberg, O.; Anisimov, A.; Kholová, I.; Pytowski, B.; et al. VEGF receptor 2/-3 heterodimers detected in situ by proximity ligation on angiogenic sprouts. EMBO J. 2010, 29, 1377–1388. [Google Scholar] [CrossRef]
  59. Harris, N.C.; Davydova, N.; Roufail, S.; Paquet-Fifield, S.; Paavonen, K.; Karnezis, T.; Zhang, Y.-F.; Sato, T.; Rothacker, J.; Nice, E.C.; et al. The Propeptides of VEGF-D Determine Heparin Binding, Receptor Heterodimerization, and Effects on Tumor Biology. J. Biol. Chem. 2013, 288, 8176–8186. [Google Scholar] [CrossRef]
  60. Han, Z.; Jiang, G.; Zhang, Y.; Xu, J.; Chen, C.; Zhang, L.; Xu, Z.; DU, X. Effects of RNA interference-mediated NRP-1 silencing on the proliferation and apoptosis of breast cancer cells. Mol. Med. Rep. 2012, 12, 513–519. [Google Scholar] [CrossRef]
  61. Zhao, L.; Chen, H.; Lu, L.; Wang, L.; Zhang, X.; Guo, X. New insights into the role of co-receptor neuropilins in tumour angiogenesis and lymphangiogenesis and targeted therapy strategies. J. Drug Target. 2021, 29, 155–167. [Google Scholar] [CrossRef] [PubMed]
  62. Moessinger, C.; Nilsson, I.; Muhl, L.; Zeitelhofer, M.; Sahlgren, B.H.; Skogsberg, J.; Eriksson, U. VEGF-B signaling impairs endothelial glucose transcytosis by decreasing membrane cholesterol content. EMBO Rep. 2020, 21, e49343. [Google Scholar] [CrossRef] [PubMed]
  63. Ramjiawan, R.R.; Griffioen, A.W.; Duda, D.G. Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis 2017, 20, 185–204. [Google Scholar] [CrossRef] [PubMed]
  64. Chen, R.; Lee, C.; Lin, X.; Zhao, C.; Li, X. Novel function of VEGF-B as an antioxidant and therapeutic implications. Pharmacol. Res. 2019, 143, 33–39. [Google Scholar] [CrossRef]
  65. Robciuc, M.R.; Kivelä, R.; Williams, I.M.; de Boer, J.F.; van Dijk, T.H.; Elamaa, H.; Tigistu-Sahle, F.; Molotkov, D.; Leppänen, V.-M.; Käkelä, R.; et al. VEGFB/VEGFR1-Induced Expansion of Adipose Vasculature Counteracts Obesity and Related Metabolic Complications. Cell Metab. 2016, 23, 712–724. [Google Scholar] [CrossRef]
  66. Ogawa, S.; Oku, A.; Sawano, A.; Yamaguchi, S.; Yazaki, Y.; Shibuya, M. A Novel Type of Vascular Endothelial Growth Factor, VEGF-E (NZ-7 VEGF), Preferentially Utilizes KDR/Flk-1 Receptor and Carries a Potent Mitotic Activity without Heparin-binding Domain. J. Biol. Chem. 1998, 273, 31273–31282. [Google Scholar] [CrossRef]
  67. Brouillet, S.; Hoffmann, P.; Benharouga, M.; Salomon, A.; Schaal, J.-P.; Feige, J.-J.; Alfaidy, N.; Hellesøy, M.; Lorens, J.B.; Yap, M.E.A.; et al. Molecular Characterization of EG-VEGF-mediated Angiogenesis: Differential Effects on Microvascular and Macrovascular Endothelial Cells. Mol. Biol. Cell 2010, 21, 2832–2843. [Google Scholar] [CrossRef]
  68. Shibuya, M. Vascular endothelial growth factor receptor-2: Its unique signaling and specific ligand, VEGF-E. Cancer Sci. 2003, 94, 751–756. [Google Scholar] [CrossRef]
  69. Komori, Y.; Nikai, T.; Taniguchi, K.; Masuda, K.; Sugihara, H. Vascular Endothelial Growth Factor VEGF-like Heparin-Binding Protein from the Venom of Vipera aspis aspis (Aspic Viper). Biochemistry 1999, 38, 11796–11803. [Google Scholar] [CrossRef]
  70. Ferreira, I.G.; Pucca, M.B.; de Oliveira, I.S.; Cerni, F.A.; Jacob, B.d.C.d.S.; Arantes, E.C. Snake venom vascular endothelial growth factors (svVEGFs): Unravelling their molecular structure, functions, and research potential. Cytokine Growth Factor Rev. 2021, 60, 133–143. [Google Scholar] [CrossRef]
  71. De Falco, S. The discovery of placenta growth factor and its biological activity. Exp. Mol. Med. 2012, 44, 1–9. [Google Scholar] [CrossRef] [PubMed]
  72. Macarulla, T.; Montagut, C.; Sánchez-Martin, F.J.; Granja, M.; Verdaguer, H.; Sastre, J.; Tabernero, J. The role of PIGF blockade in the treatment of colorectal cancer: Overcoming the pitfalls. Expert Opin. Biol. Ther. 2019, 20, 15–22. [Google Scholar] [CrossRef]
  73. Krebs, R.; Jeltsch, M. The lymphangiogenic growth factors VEGF-C and VEGF-D. LymphForsch 2013, 17, 30–37. [Google Scholar]
  74. Carmeliet, P. Angiogenesis in life, disease and medicine. Nature 2005, 438, 932–936. [Google Scholar] [CrossRef]
  75. Oliver, G.; Kipnis, J.; Randolph, G.J.; Harvey, N.L. The Lymphatic Vasculature in the 21st Century: Novel Functional Roles in Homeostasis and Disease. Cell 2020, 182, 270–296. [Google Scholar] [CrossRef] [PubMed]
  76. Jeltsch, M.; Tammela, T.; Alitalo, K.; Wilting, J. Genesis and pathogenesis of lymphatic vessels. Cell Tissue Res. 2003, 314, 69–84. [Google Scholar] [CrossRef]
  77. Deng, H.; Zhang, J.; Wu, F.; Wei, F.; Han, W.; Xu, X.; Zhang, Y. Current Status of Lymphangiogenesis: Molecular Mechanism, Immune Tolerance, and Application Prospect. Cancers 2023, 15, 1169. [Google Scholar] [CrossRef]
  78. Suarez, A.C.; Hammel, J.H.; Munson, J.M. Modeling lymphangiogenesis: Pairing in vitro and in vivo metrics. Microcirculation 2023, 30, e12802. [Google Scholar] [CrossRef]
  79. Patnam, M.; Dommaraju, S.R.; Masood, F.; Herbst, P.; Chang, J.-H.; Hu, W.-Y.; Rosenblatt, M.I.; Azar, D.T. Lymphangiogenesis Guidance Mechanisms and Therapeutic Implications in Pathological States of the Cornea. Cells 2023, 12, 319. [Google Scholar] [CrossRef]
  80. Wang, C.; Chu, M. Advances in Drugs Targeting Lymphangiogenesis for Preventing Tumor Progression and Metastasis. Front. Oncol. 2022, 11, 783309. [Google Scholar] [CrossRef]
  81. Cadamuro, M.; Romanzi, A.; Guido, M.; Sarcognato, S.; Cillo, U.; Gringeri, E.; Zanus, G.; Strazzabosco, M.; Simioni, P.; Villa, E.; et al. Translational Value of Tumor-Associated Lymphangiogenesis in Cholangiocarcinoma. J. Pers. Med. 2022, 12, 1086. [Google Scholar] [CrossRef] [PubMed]
  82. Schito, L. Hypoxia-Dependent Angiogenesis and Lymphangiogenesis in Cancer, in Hypoxia and Cancer Metastasis; Gilkes, D.M., Ed.; Springer International Publishing: Cham, Switzerland, 2019; pp. 71–85. [Google Scholar]
  83. Tammela, T.; Alitalo, K. Lymphangiogenesis: Molecular Mechanisms and Future Promise. Cell 2010, 140, 460–476. [Google Scholar] [CrossRef] [PubMed]
  84. Simons, M.; Gordon, E.; Claesson-Welsh, L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 2016, 17, 611–625. [Google Scholar] [CrossRef] [PubMed]
  85. Suresh, R.; Diaz, R.J. The remodelling of actin composition as a hallmark of cancer. Transl. Oncol. 2021, 14, 101051. [Google Scholar] [CrossRef] [PubMed]
  86. Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of Tumor Endothelial Cells in Cancer Progression. Int. J. Mol. Sci. 2018, 19, 1272. [Google Scholar] [CrossRef] [PubMed]
  87. 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. [Google Scholar] [CrossRef] [PubMed]
  88. 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. [Google Scholar]
  89. 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. [Google Scholar] [CrossRef]
  90. 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. [Google Scholar] [CrossRef]
  91. 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. [Google Scholar] [CrossRef]
  92. Qi, S.; Deng, S.; Lian, Z.; Yu, K. Novel Drugs with High Efficacy against Tumor Angiogenesis. Int. J. Mol. Sci. 2022, 23, 6934. [Google Scholar] [CrossRef] [PubMed]
  93. Iruela-Arispe, M.L.; Davis, G.E. Cellular and Molecular Mechanisms of Vascular Lumen Formation. Dev. Cell 2009, 16, 222–231. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef]
  95. 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. [Google Scholar] [CrossRef] [PubMed]
  96. Maishi, N.; Annan, D.A.; Kikuchi, H.; Hida, Y.; Hida, K. Tumor Endothelial Heterogeneity in Cancer Progression. Cancers 2019, 11, 1511. [Google Scholar] [CrossRef]
  97. Taleb, M.; Mohammadkhani, N.; Bahreini, F.; Ovais, M.; Nie, G. Modulation of Tumor Vasculature Network: Key Strategies. Small Struct. 2022, 3, 2100164. [Google Scholar] [CrossRef]
  98. 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. [Google Scholar] [CrossRef]
  99. 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. [Google Scholar]
  100. Rajabi, M.; Mousa, S.A. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017, 5, 34. [Google Scholar] [CrossRef]
  101. 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. [Google Scholar] [CrossRef]
  102. 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. [Google Scholar] [CrossRef] [PubMed]
  103. 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. [Google Scholar] [CrossRef] [PubMed]
  104. 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. [Google Scholar] [CrossRef] [PubMed]
  105. 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. [Google Scholar] [CrossRef] [PubMed]
  106. Jain, R.K. Antiangiogenesis Strategies Revisited: From Starving Tumors to Alleviating Hypoxia. Cancer Cell 2014, 26, 605–622. [Google Scholar] [CrossRef] [PubMed]
  107. Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603. [Google Scholar] [CrossRef]
  108. 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. [Google Scholar] [CrossRef]
  109. 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. [Google Scholar] [CrossRef]
  110. 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. [Google Scholar] [CrossRef]
  111. 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. [Google Scholar] [CrossRef]
  112. Jain, R.K. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science 2005, 307, 58–62. [Google Scholar] [CrossRef] [PubMed]
  113. 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. [Google Scholar] [CrossRef]
  114. 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. [Google Scholar] [CrossRef] [PubMed]
  115. 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. [Google Scholar] [CrossRef] [PubMed]
  116. 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. [Google Scholar] [CrossRef]
  117. 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. [Google Scholar] [CrossRef]
  118. 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. [Google Scholar] [CrossRef]
  119. 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. [Google Scholar] [CrossRef]
  120. 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. [Google Scholar] [CrossRef]
  121. 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. [Google Scholar] [CrossRef]
  122. 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. [Google Scholar] [CrossRef]
  123. 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. [Google Scholar] [CrossRef]
  124. 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. [Google Scholar] [CrossRef]
  125. 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. [Google Scholar] [CrossRef]
  126. 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. [Google Scholar] [CrossRef]
  127. 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. [Google Scholar] [CrossRef]
  128. 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. [Google Scholar]
  129. 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. [Google Scholar]
  130. 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. [Google Scholar] [CrossRef]
  131. 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. [Google Scholar] [CrossRef]
  132. 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. [Google Scholar] [CrossRef] [PubMed]
  133. 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. [Google Scholar] [CrossRef]
  134. 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. [Google Scholar] [CrossRef] [PubMed]
  135. 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. [Google Scholar] [CrossRef]
  136. 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. [Google Scholar] [CrossRef]
  137. 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. [Google Scholar] [CrossRef]
  138. 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. [Google Scholar] [CrossRef]
  139. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Proteomics. Tissue-Based Map of the Human Proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  140. Hata, T.; Sugimoto, N.; Ohara, N.; Miyo, M.; Yoshioka, S.; Kagawa, Y.; Naito, A.; Tei, M.; Tamagawa, H.; Konishi, K.; et al. Clinical development and evaluation of plasma angiogenesis factors from phase II study of FOLFIRI plus ramucirumab with recurrent colorectal cancer refractory to adjuvant chemotherapy with oxaliplatin/fluoropyrimidine (RAINCLOUD): RAINCLOUD-TR. J. Clin. Oncol. 2023, 41, 173. [Google Scholar] [CrossRef]
  141. Nie, Z.; Zhang, K.; Li, Z.; Bing, X.; Jin, S.; Li, M. Human papillomavirus 16 E6 promotes angiogenesis of lung cancer via SNHG. Cell Biochem. Biophys. 2023, 81, 325–336. [Google Scholar] [CrossRef]
  142. Rusak, A.; Jablonska, K.; Piotrowska, A.; Grzegrzolka, J.; Nowak, A.; Wojnar, A.; Dziegiel, P. The Role of CHI3L1 Expression in Angiogenesis in Invasive Ductal Breast Carcinoma. Anticancer. Res. 2018, 38, 3357–3366. [Google Scholar] [CrossRef]
  143. Bussolati, B.; Deambrosis, I.; Russo, S.; Deregibus, M.C.; Camussi, G. Altered angiogenesis and survival in human tumor-derived endothelial cells. FASEB J. 2003, 17, 1159–1161. [Google Scholar] [CrossRef]
  144. Lee, S.; Zhou, P.; Gupta, A.; Shin, S. Reactive Ductules Are Associated with Angiogenesis and Tumor Cell Proliferation in Pediatric Liver Cancer. Hepatol. Commun. 2018, 2, 1199–1212. [Google Scholar] [CrossRef]
  145. Yokoyama, Y.; Charnock-Jones, S.; Licence, D.; Yanaihara, A.; Hastings, J.; Holland, C.; Emoto, M.; Umemoto, M.; Sakamoto, T.; Sato, S.; et al. Vascular endothelial growth factor-D is an independent prognostic factor in epithelial ovarian carcinoma. Br. J. Cancer 2003, 88, 237–244. [Google Scholar] [CrossRef]
  146. White, J.D.; Hewett, P.W.; Kosuge, D.; McCulloch, T.; Enholm, B.C.; Carmichael, J.; Murray, J.C. Vascular endothelial growth factor-D expression is an independent prognostic marker for survival in colo-rectal carcinoma. Cancer Res. 2002, 62, 1669–1675. [Google Scholar]
  147. Nakamura, Y.; Sato, S.; Futagami, M.; Fukushi, Y.; Sakamoto, T.; Umemoto, M.; Saito, Y. Prognostic significance of vascular endothelial growth factor D in breast carcinoma with long-term fol-low-up. Clin Cancer Res. 2003, 9, 716–721. [Google Scholar]
  148. Sopo, M.; Anttila, M.; Hämäläinen, K.; Kivelä, A.; Ylä-Herttuala, S.; Kosma, V.-M.; Keski-Nisula, L.; Sallinen, H. Expression profiles of VEGF-A, VEGF-D and VEGFR1 are higher in distant metastases than in matched primary high grade epithelial ovarian cancer. BMC Cancer 2019, 19, 584. [Google Scholar] [CrossRef]
  149. Nienhüser, H.; Crnovrsanin, N.; Nerz, D.; Heckler, M.; Sisic, L.; Lasitschka, F.; Schneider, M.; Schmidt, T. Expression of Angiogenic Proteins in Tumor and Stroma Affects Survival in Patients with Gastric Cancer. J. Surg. Res. 2020, 255, 172–180. [Google Scholar] [CrossRef]
  150. Oplawski, M.; Dziobek, K.; Zmarzły, N.; Grabarek, B.; Halski, T.; Januszyk, P.; Kuś-Kierach, A.; Adwent, I.; Dąbruś, D.; Kiełbasiński, K.; et al. Expression Profile of VEGF-C, VEGF-D, and VEGFR-3 in Different Grades of Endometrial Cancer. Curr. Pharm. Biotechnol. 2019, 20, 1004–1010. [Google Scholar] [CrossRef]
  151. Nalezinska, M. EP1288 The role of angiogenesis in early detection of epithelial ovarian carcinoma (EOC)—VEGF D (vascular endothelial growth factor D) as a potential novel biomarker. ePoster 2019, 29, A648. [Google Scholar] [CrossRef]
  152. Lieu, C.H.; Tran, H.; Jiang, Z.-Q.; Mao, M.; Overman, M.J.; Lin, E.; Eng, C.; Morris, J.; Ellis, L.; Heymach, J.V.; et al. The Association of Alternate VEGF Ligands with Resistance to Anti-VEGF Therapy in Metastatic Colorectal Cancer. PLoS ONE 2013, 8, e77117. [Google Scholar] [CrossRef]
  153. Weickhardt, A.J.; Williams, D.S.; Lee, C.K.; Chionh, F.; Simes, J.; Murone, C.; Wilson, K.; Parry, M.M.; Asadi, K.; Scott, A.M.; et al. Vascular endothelial growth factor D expression is a potential biomarker of bevacizumab benefit in colorectal cancer. Br. J. Cancer 2015, 113, 37–45. [Google Scholar] [CrossRef]
  154. Tabernero, J.; Hozak, R.; Yoshino, T.; Cohn, A.; Obermannova, R.; Bodoky, G.; Garcia-Carbonero, R.; Ciuleanu, T.-E.; Portnoy, D.; Prausová, J.; et al. Analysis of angiogenesis biomarkers for ramucirumab efficacy in patients with metastatic colorectal cancer from RAISE, a global, randomized, double-blind, phase III study. Ann. Oncol. 2017, 29, 602–609. [Google Scholar] [CrossRef]
  155. Carmeliet, P.; Li, X.; Treps, L.; Conradi, L.-C.; Loges, S. RAISEing VEGF-D’s importance as predictive biomarker for ramucirumab in metastatic colorectal cancer patients. Ann. Oncol. 2018, 29, 529–532. [Google Scholar] [CrossRef]
  156. Ose, J.; Gigic, B.; Hardikar, S.; Lin, T.; Himbert, C.; Warby, C.A.; Peoples, A.R.; Lindley, C.L.; Boehm, J.; Schrotz-King, P.; et al. Presurgery Adhesion Molecules and Angiogenesis Biomarkers Are Differently Associated with Outcomes in Colon and Rectal Cancer: Results from the ColoCare Study. Cancer Epidemiol. Biom. Prev. 2022, 31, 1650–1660. [Google Scholar] [CrossRef]
  157. Himbert, C.; Ose, J.; Lin, T.; Warby, C.A.; Gigic, B.; Steindorf, K.; Schrotz-King, P.; Abbenhardt-Martin, C.; Zielske, L.; Boehm, J.; et al. Inflammation- and angiogenesis-related biomarkers are correlated with cancer-related fatigue in colorectal cancer patients: Results from the ColoCare Study. Eur. J. Cancer Care 2019, 28, e13055. [Google Scholar] [CrossRef]
  158. Mazeda, I.; Martins, S.F.; Garcia, E.A.; Rodrigues, M.; Longatto, A. VEGF Expression in Colorectal Cancer Metastatic Lymph Nodes: Clinicopathological Correlation and Prognostic Significance. Gastrointest. Disord. 2020, 2, 267–280. [Google Scholar] [CrossRef]
  159. Izawa, N.; Shitara, K.; Masuishi, T.; Denda, T.; Yamazaki, K.; Moriwaki, T.; Okuda, H.; Kondoh, C.; Nishina, T.; Makiyama, A.; et al. Vascular endothelial growth factor (VEGF)-D and clinical outcomes in metastatic colorectal cancer (mCRC) patients (pts) treated with second-line FOLFIRI plus bevacizumab (Bev): A biomarker study of the WJOG 6210G trial. J. Clin. Oncol. 2020, 38, 226. [Google Scholar] [CrossRef]
  160. Woei-A-Jin, F.S.H.; Weijl, N.I.; Burgmans, M.C.; Fariña Sarasqueta, A.; Tom van Wezel, J.; Wasser, M.N.; Coenraad, M.J.; Burggraaf, J.; Osanto, S. Neoadjuvant Treatment with Angiogenesis-Inhibitor Dovitinib Prior to Local Therapy in Hepato-cellular Carcinoma: A Phase II Study. Oncologist 2021, 26, 854–864. [Google Scholar] [CrossRef]
  161. Miao, Y.-D.; Tang, X.-L.; Wang, J.-T.; Mi, D.-H. Prognostic role of expression of angiogenesis markers in hepatocellular carcinoma: A bioinformatics analysis. World J. Gastroenterol. 2022, 28, 4221–4226. [Google Scholar] [CrossRef]
  162. Gkika, E.; Adebahr, S.; Brenner, A.; Schimek-Jasch, T.; Radicioni, G.; Exner, J.-P.; Rühle, A.; Spohn, S.K.B.; Popp, I.; Zamboglou, C.; et al. Changes in Blood Biomarkers of Angiogenesis and Immune Modulation after Radiation Therapy and Their Association with Outcomes in Thoracic Malignancies. Cancers 2021, 13, 5725. [Google Scholar] [CrossRef]
  163. Taniguchi, H.; Yuki, S.; Shiozawa, M.; Masuishi, T.; Nishina, T.; Kagawa, Y.; Takahashi, N.; Yasui, H.; Denda, T.; Sunakawa, Y.; et al. Plasma VEGF-D and PlGF levels according to prior use of biologics among metastatic colorectal cancer: Preliminary results from GI-SCREEN CRC-Ukit study. J. Clin. Oncol. 2020, 38, 178. [Google Scholar] [CrossRef]
  164. Pisa, U.O. Predictive Role of Circulating Angiogenic Factors for Second-line Paclitaxel and Ramucirumab. 2021. Available online: https://ClinicalTrials.gov/show/NCT05301465 (accessed on 24 July 2023).
  165. Achen, M.G.; Roufail, S.; Domagala, T.; Catimel, B.; Nice, E.C.; Geleick, D.M.; Murphy, R.; Scott, A.M.; Caesar, C.; Makinen, T.; et al. Monoclonal antibodies to vascular endothelial growth factor-D block its interactions with both VEGF receptor-2 and VEGF receptor-3. JBIC J. Biol. Inorg. Chem. 2000, 267, 2505–2515. [Google Scholar] [CrossRef]
  166. Davydova, N.; Roufail, S.; Streltsov, V.A.; Stacker, S.A.; Achen, M.G. The VD1 Neutralizing Antibody to Vascular Endothelial Growth Factor-D: Binding Epitope and Rela-tionship to Receptor Binding. J. Mol. Biol. 2011, 407, 581–593. [Google Scholar] [CrossRef]
  167. Davydova, N.; Harris, N.C.; Roufail, S.; Paquet-Fifield, S.; Ishaq, M.; Streltsov, V.A.; Williams, S.P.; Karnezis, T.; Stacker, S.A.; Achen, M.G. Differential receptor binding and regulatory mechanisms for the lymphangiogenic growth factors vas-cular endothelial growth factor (VEGF)-C and-D. J. Biol. Chem. 2016, 291, 27265–27278. [Google Scholar] [CrossRef] [PubMed]
  168. Ji, L.; Zheng, Z.; Shi, L.; Huang, Y.; Lu, B.; Wang, Z. Andrographolide decreased VEGFD expression in hepatoma cancer cells by inducing ubiquitin/proteasome-mediated cFos protein degradation. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2015, 1850, 750–758. [Google Scholar] [CrossRef]
  169. Turunen, T.; Hua, A.; Shatos, M.; Teague, G.C.; Baldwin, M.; Lashkari, K. VEGF-C and VEGF-D Inhibition by VGX-300 Effectively Reduces Leukocyte Adhesion and Vascular Leakage in the STZ-Rat Model of Diabetic Retinal Edema. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3667. [Google Scholar]
  170. Lashkari, K.; Ma, J.; Sun, Y.; Teague, G.C.; Baldwin, M.E. VGX-300, a ‘Trap’for VEGF-C and VEGF-D, Inhibits Choroidal Neovascularization and Vascular Leakage in a Mouse Model of Wet AMD. Investig. Ophthalmol. Vis. Sci. 2015, 56, 4802. [Google Scholar]
  171. Pytowski, B.; Goldman, J.; Persaud, K.; Wu, Y.; Witte, L.; Hicklin, D.J.; Skobe, M.; Boardman, K.C.; Swartz, M.A. Complete and Specific Inhibition of Adult Lymphatic Regeneration by a Novel VEGFR-3 Neutralizing Antibody. Gynecol. Oncol. 2005, 97, 14–21. [Google Scholar] [CrossRef]
  172. Persaud, K.; Tille, J.-C.; Liu, M.; Zhu, Z.; Jimenez, X.; Pereira, D.S.; Miao, H.-Q.; Brennan, L.A.; Witte, L.; Pepper, M.S.; et al. Involvement of the VEGF receptor 3 in tubular morphogenesis demonstrated with a human anti-human VEGFR-3 monoclonal antibody that antagonizes receptor activation by VEGF-C. J. Cell Sci. 2004, 117, 2745–2756. [Google Scholar] [CrossRef]
  173. Jackson, T.L.; Slakter, J.; Buyse, M.; Wang, K.; Dugel, P.U.; Wykoff, C.C.; Boyer, D.S.; Gerometta, M.; Baldwin, M.E.; Price, C.F.; et al. A Randomized Controlled Trial of OPT-302, a VEGF-C/D Inhibitor for Neovascular Age-Related Macular Degeneration. Ophthalmology 2023, 130, 588–597. [Google Scholar] [CrossRef]
  174. Limited, O. Study Evaluating the Safety, Pharmacokinetics and Pharmacodynamics of OPT-302 with or Without Lucentis™ in Patients with Wet AMD. 2015. Available online: https://ClinicalTrials.gov/show/NCT02543229 (accessed on 24 July 2023).
  175. Lee, W.S.; Pyun, B.-J.; Kim, S.-W.; Shim, S.R.; Nam, J.R.; Yoo, J.Y.; Jin, Y.; Jin, J.; Kwon, Y.-G.; Yun, C.-O.; et al. TTAC-0001, a human monoclonal antibody targeting VEGFR-2/KDR, blocks tumor angiogenesis. mAbs 2015, 7, 957–968. [Google Scholar] [CrossRef]
  176. PharmAbcine. Phase I Trial of Tanibirumab in Advanced or Metastatic Cancer. 2011. Available online: https://ClinicalTrials.gov/show/NCT01660360 (accessed on 24 July 2023).
  177. PharmAbcine. Trial to Evaluate the Safety of TTAC-0001(Tanibirumab) in Recurrent Glioblastoma. 2016. Available online: https://ClinicalTrials.gov/show/NCT03033524 (accessed on 24 July 2023).
  178. Kras, P.; Talkowski, K.; Grabarek, B.O.; Skalska-Dziobek, N.; Boroń, D.; Oplawski, M. Evaluation of Variances in VEGF-AD and VEGFR-1-3 Expression in the Ishikawa Endometrial Cancer Cell Line Treated with Salinomycin and Anti-Angiogenic/Lymphangiogenic Effect. Curr. Pharm. Biotechnol. 2021, 22, 697–705. [Google Scholar] [CrossRef]
  179. Pan, M.-S.; Cao, J.; Fan, Y.-Z. Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities. Chin. Med. 2020, 15, 55. [Google Scholar] [CrossRef]
  180. Laakkonen, P.; Waltari, M.; Holopainen, T.; Takahashi, T.; Pytowski, B.; Steiner, P.; Hicklin, D.; Persaud, K.; Tonra, J.R.; Witte, L.; et al. Vascular Endothelial Growth Factor Receptor 3 Is Involved in Tumor Angiogenesis and Growth. Cancer Res. 2007, 67, 593–599. [Google Scholar] [CrossRef]
  181. Hwang, S.D.; Song, J.H.; Kim, Y.; Lim, J.H.; Kim, M.Y.; Kim, E.N.; Hong, Y.A.; Chung, S.; Choi, B.S.; Kim, Y.-S.; et al. Inhibition of lymphatic proliferation by the selective VEGFR-3 inhibitor SAR131675 ameliorates diabetic nephropathy in db/db mice. Cell Death Dis. 2019, 10, 219. [Google Scholar] [CrossRef]
  182. Tempfer, H.; Spitzer, G.; Lehner, C.; Wagner, A.; Gehwolf, R.; Fierlbeck, J.; Weissenbacher, N.; Jessen, M.; Heindl, L.M.; Traweger, A. VEGF-D-mediated signaling in tendon cells is involved in degenerative processes. FASEB J. 2022, 36, e22126. [Google Scholar] [CrossRef]
  183. Oholendt, A.L.; Zadlo, J.L. Ramucirumab: A New Therapy for Advanced Gastric Cancer. J. Adv. Pract. Oncol. 2015, 6, 71–75. [Google Scholar] [CrossRef]
  184. Lee, S.H. Tanibirumab (TTAC-0001): A fully human monoclonal antibody targets vascular endothelial growth factor receptor 2 (VEGFR-2). Arch. Pharmacal Res. 2011, 34, 1223–1226. [Google Scholar] [CrossRef]
  185. Hao, Z.; Sadek, I. Sunitinib: The antiangiogenic effects and beyond. OncoTargets Ther. 2016, 9, 5495–5505. [Google Scholar] [CrossRef]
  186. Taghour, M.S.; Mahdy, H.A.; Gomaa, M.H.; Aglan, A.; Eldeib, M.G.; Elwan, A.; Dahab, M.A.; Elkaeed, E.B.; Alsfouk, A.A.; Khalifa, M.M.; et al. Benzoxazole derivatives as new VEGFR-2 inhibitors and apoptosis inducers: Design, synthesis, in silico studies, and antiproliferative evaluation. J. Enzym. Inhib. Med. Chem. 2022, 37, 2063–2077. [Google Scholar] [CrossRef] [PubMed]
  187. Jia, Y.; Liu, M.; Huang, W.; Wang, Z.; He, Y.; Wu, J.; Ren, S.; Ju, Y.; Geng, R.; Li, Z. Recombinant Human Endostatin Endostar Inhibits Tumor Growth and Metastasis in a Mouse Xenograft Model of Colon Cancer. Pathol. Oncol. Res. 2011, 18, 315–323. [Google Scholar] [CrossRef] [PubMed]
Figure 1. VEGF-receptors and their ligands. VEGF-A binds VEGFR-1 and 2. VEGF-B and PIGF both bind VEGFR-1 only. VEGF-C and -D can bind both VEGFR-2 and 3. VEGF-E binds only VEGFR-2. VEGFR-R1 and 2 regulate blood vessel angiogenesis, whereas VEGFR-3’s main function is the regulation of lymphangiogenesis. VEGFR-1 also acts as a decoy receptor, preventing its ligands from binding VEGFR-2 [7,8,10,11,12]. Created with BioRender.com accessed on 27 July 2023.
Figure 1. VEGF-receptors and their ligands. VEGF-A binds VEGFR-1 and 2. VEGF-B and PIGF both bind VEGFR-1 only. VEGF-C and -D can bind both VEGFR-2 and 3. VEGF-E binds only VEGFR-2. VEGFR-R1 and 2 regulate blood vessel angiogenesis, whereas VEGFR-3’s main function is the regulation of lymphangiogenesis. VEGFR-1 also acts as a decoy receptor, preventing its ligands from binding VEGFR-2 [7,8,10,11,12]. Created with BioRender.com accessed on 27 July 2023.
Ijms 24 13317 g001
Figure 3. Steps of tumor angiogenesis. Created with BioRender.com accessed on 27 July 2023 based on: [91,100]. Step 1 (hypoxia): the hypoxic or inflamed tumor microenvironment (TME) induces the production of vasculogenetic growth factors. Step 2 (proteolytic degradation): VEGF-A and -D binding to VEGFR-2 induce the production of matrix metalloproteinases (MMPs), degrading the extracellular matrix (ECM). Step 3 (tip cell migration): VEGFR-2 activation induces the transformation of ECs to tip cells (brown). Step 4 (tube formation): tip cells migrate into the TME with coordinated proliferation of stalk cells, forming a new tube. Step 5 (tumor vascularization): the new vessels mature by establishing tight junctions and by the coverage by pericytes (lila).
Figure 3. Steps of tumor angiogenesis. Created with BioRender.com accessed on 27 July 2023 based on: [91,100]. Step 1 (hypoxia): the hypoxic or inflamed tumor microenvironment (TME) induces the production of vasculogenetic growth factors. Step 2 (proteolytic degradation): VEGF-A and -D binding to VEGFR-2 induce the production of matrix metalloproteinases (MMPs), degrading the extracellular matrix (ECM). Step 3 (tip cell migration): VEGFR-2 activation induces the transformation of ECs to tip cells (brown). Step 4 (tube formation): tip cells migrate into the TME with coordinated proliferation of stalk cells, forming a new tube. Step 5 (tumor vascularization): the new vessels mature by establishing tight junctions and by the coverage by pericytes (lila).
Ijms 24 13317 g003
Table 1. Antiangiogenic drugs targeting VEGF-D.
Table 1. Antiangiogenic drugs targeting VEGF-D.
DrugMechanism of ActionSelectivity, Mechanism of ActionReference
AndrographolideTargets the VEGF-D inducer: c-fosNon-selective[168]
VGX-300Soluble VEGFR-3Bind and inhibit the activation of VEGFR-3 by all of its ligands (VEGF-D/C/A)[169,170,171,172,173,180,181,182]
SAR131675VEGFR-3 tyrosine kinase inhibitor (it does not say about its property. Inhibits phosphorylation and hence activation of the receptor)
mF4-31C1, hF4-3C5mABs against VEGFR-3
OPT-302VEGFR-3 decoy
mAB 286mAbSelective: Binds mature VEGF-D, prevents its binding to VEGFR-2 and VEGFR-3[165,167]
VD1mAb Competes with VEGF-D in activation of VEGFR-2 and VEGFR-3[166]
Tanibitumab, Ramucirumab,
SU5614,
Sunitinib,
Benzoxazole and its derivatives
VEGFR-2 inhibitorsVEGFR-2 inhibition prevents blood angiogenic signaling from (VEGF-A/C/D)[57,183,184,185,186]
EndostatinVEGF-D (and VEGF-A, VEGF-C) inhibitorNon-selective[187]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bokhari, S.M.Z.; Hamar, P. Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors. Int. J. Mol. Sci. 2023, 24, 13317. https://doi.org/10.3390/ijms241713317

AMA Style

Bokhari SMZ, Hamar P. Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors. International Journal of Molecular Sciences. 2023; 24(17):13317. https://doi.org/10.3390/ijms241713317

Chicago/Turabian Style

Bokhari, Syeda Mahak Zahra, and Peter Hamar. 2023. "Vascular Endothelial Growth Factor-D (VEGF-D): An Angiogenesis Bypass in Malignant Tumors" International Journal of Molecular Sciences 24, no. 17: 13317. https://doi.org/10.3390/ijms241713317

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop