Next Article in Journal
Evaluation of Bi-Lateral Co-Infections and Antibiotic Resistance Rates among COVID-19 Patients in Lahore, Pakistan
Previous Article in Journal
Darier Disease with Psoriasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview

by
Cătălina Ionescu
1,*,
Bogdan Oprea
2,*,
Georgeta Ciobanu
1,
Milena Georgescu
3,
Ramona Bică
4,
Garofiţa-Olivia Mateescu
2,
Fidan Huseynova
5,6,7 and
Veronique Barragan-Montero
5
1
Department of Chemistry, Faculty of Sciences, University of Craiova, 107i Calea București, 200144 Craiova, Romania
2
Histology Department, University of Medicine and Pharmacy, 2-4 Petru Rares, 200349 Craiova, Romania
3
Clinic for Plastic Surgery and Burns, County Emergency Hospital Craiova, 200642 Craiova, Romania
4
General Hospital—“Victor Babes”, 281 Mihai Bravu St., Sector III, 030303 Bucharest, Romania
5
LBN, University of Montpellier, 34193 Montpellier, France
6
Institute of Molecular Biology and Biotechnologies, Azerbaïjan National Academy of Sciences (ANAS), AZ1073 Baku, Azerbaijan
7
Department of Histology, Cytology and Embryology, Azerbaijan Medical University, AZ1078 Baku, Azerbaijan
*
Authors to whom correspondence should be addressed.
Medicina 2022, 58(7), 903; https://doi.org/10.3390/medicina58070903
Submission received: 15 June 2022 / Revised: 4 July 2022 / Accepted: 5 July 2022 / Published: 6 July 2022
(This article belongs to the Section Cardiology)

Abstract

:
Angiogenesis is the process of developing new blood vessels from pre-existing ones. This review summarizes the main features of physiological and pathological angiogenesis and those of angiogenesis activation and inhibition. In healthy adults, angiogenesis is absent apart from its involvement in female reproductive functions and tissue regeneration. Angiogenesis is a complex process regulated by the action of specific activators and inhibitors. In certain diseases, modulating the angiogenic balance can be a therapeutic route, either by inhibiting angiogenesis (for example in the case of tumor angiogenesis), or by trying to activate the process of new blood vessels formation, which is the goal in case of cardiac or peripheral ischemia.

1. Introduction

According to the American Heart Association, in 2013, one out of three persons died because of cardiovascular diseases in the United States, while stroke and heart attacks were considered to be the primordial causes of worldwide death [1].
Cancer is another disease affecting many lives, being one of the major and most frequently observed death causes in the European Union, based on statistical reports. What could bring together these diseases that have such different causes and specific molecular mechanisms?
One of the principles that could somehow “bridge” them is angiogenesis regulation. By inhibiting tumor angiogenesis, anticancer treatments become more effective whereas, on the contrary, by activating angiogenesis, the formation of new blood vessels would lead to a better vascularization of ischemic heart regions.
Due to its implications of the utmost importance, a great deal of research has been devoted to angiogenesis and its underlying mechanisms. Physiological angiogenesis occurs as a result of a fine-tuned balance of endogenous regulators, having, mostly, a protein structure. The actions of activators, such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), “platelet-derived endothelial cell growth factor” (PD-ECGF), angiopoietin, and angiogenin [2,3,4,5,6], are balanced by processes determined by inhibitors, such as endostatin and angiostatin. The vascular endothelial growth factor (VEGF) action is closely related to the stimulation of angiogenesis [5,6]. The VEGF proteins family consists of six members [5,7,8], with VEGF-A being the most studied, included in certain anti-cancer therapies [7,9]. Exogenous compounds such as polypeptides [10] or small synthetic molecules, antibiotics, saccharides and steroids have been shown to induce angiogenesis regulation [11,12].
Disequilibrium of the balance of angiogenesis regulators leads to insufficient angiogenesis, linked to ischemia, ulcers, infertility or hair loss. Excessive angiogenesis has been identified as being related to tumor growth, metastasis appearance or to a series of other diseases, among which is age-related macular degeneration, associated with elevated VEGF levels. Therefore, the therapeutic approach of anti-angiogenic therapy involves inhibitors that are able to recognize and block either VEGF itself or its receptors. On the other hand, therapeutic angiogenesis may be induced through protein, gene or cell therapy. Careful clinical studies are being conducted, as systemic effects must be avoided.
Based on the above, our mini-review is organized in sections, as follows: formation and remodeling of blood vessels; the angiogenic balance—synthetic and endogenous regulators; physiological and pathological angiogenesis; angiogenesis inhibition and current implications in cancer treatment; angiogenesis activation and current implications in cardiovascular diseases.

2. Formation and Remodeling of Blood Vessels

Blood vessels that supply the body with oxygen and nutrients, are formed through three different mechanisms (vasculo-genesis, angiogenesis, arterio-genesis) united under the name of neovascularization.
Vasculo-genesis. Blood vessels are formed in the early stages of embryo development through vasculo-genesis when the embryonic mesenchymal cells differentiate into endothelial cells and a “primary capillary plexus” is formed. At present, it is known that, besides its role in embryonic development, vasculo-genesis is also induced after birth. In this case, bone marrow-derived endothelial progenitor cells mediate the cases of physiological and pathological neovascularization, for example, in the cases of wound healing or cardiac ischemia [13,14,15].
Angiogenesis is the main process in post-natal neovascularization and represents the process of new vessel formation from pre-existing ones [16], through two mechanisms: intussusception [17,18] and sprouting [19]. Intussusception represents the “splitting” of an existing vessel into two new vessels, with interior reconstruction of novel blood vessel walls. Leading to a rise in the number of vessels without augmentation of the number of endothelial cells, intussusception angiogenesis is involved in the remodeling of existing vessels, for example, in capillary formation starting from the primary plexus in embryo development, but it can also be seen in post-natal development. Sprouting angiogenesis is a more complicated process. It is regulated by different angiogenic factors and involves several steps: degradation of the basement membrane of the vessel under enzymatic conditions, endothelial cells activation, proliferation and migration, formation of a new lumen and pericyte stabilization [19,20,21,22]. There is much evidence in different studies that a tumor’s vascularization is associated with the metastatic risk and negatively influences the survival rate. The micro-density of a tumor’s vascularization is especially used for the follow-up of anti-angiogenetic therapy [23].
Arterio-genesis represents the formation of new blood vessels from co-existing, co-lateral vessels, with the participation of smooth muscle cells, as an adaptive process to an arterial occlusion [24,25], being rather a re-modeling process. In the proximity of an arterial stenosis, the substitution network is architectured through both arterio-genesis (in near regions, unleashed by mechanical constraints and cytokines) and angiogenesis (in distal sites, where hypoxia would generate new vessel sproutings) [26,27].

3. The Angiogenic Balance: Synthetic and Endogenous Regulators

3.1. Synthetic Modulators of Angiogenesis

Most of the endogenous molecules playing a role in angiogenesis modulation have protein structure, but their usage in therapy is delicate, because of the high cost of their production in large quantities and because of the difficulty in penetrating tissues.
For this reason, more attention has been shown to the preparation and investigation of certain compounds impacting the angiogenesis, such as the polypeptides with therapeutic effect [10]. In the same terms, relating to angiogenesis modeling, some antibiotics, polysaccharides, steroids, and other synthetic small-molecular compounds have been reported [11,12].
As our research area is represented by carbohydrates, we review in detail this type of compounds used as angiogenesis modulators. Many carbohydrate-binding proteins are involved in angiogenesis; therefore, carbohydrates and their analogues may be important factors for angiogenesis regulation [28].
One example is represented by galectin-3 and MCP (modified citrus pectin). Galectin-3 is a β-galactoside-binding lectin, which mediates endothelial cell morphogenesis in vitro and angiogenesis in vivo [29].
Nangia-Makker et al. proved that it is able to tightly bind to galectin-3, via recognition of its carbohydrate recognition domain, and to inhibit angiogenesis and tumor growth [30]. Johnston at al. synthesized and studied heparan sulfate mimetics, represented by a series of poly-sulfated penta- and tetra-saccharide glycosides containing alpha(1→3)/alpha(1→2)-linked mannose residues. They found that the investigated mimetics bound tightly to angiogenic growth factors and exhibited potent activity indicative of angiogenesis; they strongly inhibited heparanase activity and also showed good antitumor activity [31].
Our research group has proved that synthetic mannose-6-phosphate analogues can act as angiogenesis activators or inhibitors, depending on the structure of the chemical group functionalizing the C6 position of mannose [32,33]. In another study, we prepared gold nanoparticles decorated with various mannose derivatives functionalized in the C6 position and they proved to be effective over angiogenesis [34]. The cation-independent mannose-6-phosphate receptor was previously indicated as inducing angiogenesis through several possible mechanisms [35], but this was the first time that mono-carbohydrates have directly been indicated as an agent possessing angiogenic activities.
Since then, it has been proven that 1,2,3,4,6-penta-O-galloyl-β-d-glucopyranose (PGG) has antiangiogenic activity in vitro and in vivo. Derivatives of PGG with different sugar cores and phenolic substituents have been tested and they are also angiogenesis inhibitors [36,37,38].

3.2. Endogenous Regulators of Angiogenesis

Besides mechanical (shear stress and blood flow augmentation) and chemical (hypoxia and nitric oxide increase) influences, angiogenesis is regulated by molecular influences, among which the most important are the angiogenic growth factors: fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), “platelet-derived endothelial cell growth factor” (PD-ECGF), angiopoietin; angiogenin, etc. [2,3,4]. The vascular endothelial growth factor (VEGF) action is closely related to the stimulation of angiogenesis [5,6]. Angiogenesis is, on the other hand, inhibited by anti-angiogenic factors, such as angiostatin, endostatin, thrombospondin-1 (TSP-1), heparinases, etc. [39]. When the balance between angiogenesis activators and inhibitors loses equilibrium, abnormal (either insufficient or excessive) angiogenesis occurs and various diseases appear or degenerate [40].
The main endogenous angiogenesis activators are summarized in Table 1. The most commonly studied endogenous angiogenesis activators are the FGF and VEGF families. bFGF is the first angiogenic factor that has been purified, in 1975 [41], and FGFs are the first angiogenic factors that were sequenced, in 1985 [42]. VEGF has been identified in 1983 as a vascular permeability factor [43], and only later, in 1989, has it been shown to possess angiogenic action [44]. Table 2 summarizes the main endogenous angiogenesis inhibitors. Thrombospondin-1 is the first protein observed to possess naturally occurring antiangiogenic properties [45,46]. Since then, the number of angiogenesis regulators have grown and, besides endogenous regulators, synthetic molecules with effect on angiogenesis have been obtained and tested, some of them already on the market and available for treatment.
There are studies that prove the binding of growth factors to the cell surface, serving as target, receptors, or even as a storage mechanism. This seems to be valid for the inductor FGF which induces the activation of VEGF.
The class of VEGF proteins consists of several derivatives such as VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E (encoded-virus) and VEGF-F (derived from snake venom) and placental growth factor (PlGF) [5,7,8]. Vascular permeability and inflammation, angiogenesis and apoptosis, lymphangio-genesis and fibrogenesis can be adjusted by the VEGF family [8]. From all the VEGF class components, VEGF-A is the most individualized, representing a substantial angiogenesis promotor and consequently designed as an objective for the study of certain anti-cancer therapies [7,9]. According to the chain length, different VEGF-A isoforms have resulted after the splicing of alternative VEGF mRNA [8], and have further been referred to as VEGFXXX, where “XXX” indicates the number of amino acids from the final protein chain [7]. The most well-known subtypes are VEGF111, VEGF121, VEGF145, VEGF165, VEGF189 and VEGF206 [5].
Human VEGF-A contains eight exons separated by seven introns, all their subtypes presenting similar regions, namely exons 1–5 and 8 [8]. The longer VEGF isoforms containing both exons 6a and 7, such as VEGF145, VEGF189 and VEGF206 have high affinity for heparin sulphate glycoproteins [5,7,47,48]. VEGF162 is a VEGF isoform, whose protein sequence has exons 1–5, 6a, 6b and 8. proliferating the angiogenesis in vivo, while VEGF165 is the most potent endothelial cells proliferation agent. The VEGF shorter isoforms, such as VEGF111 and VEGF121, do not have exons 6 and 7, are highly diffusible and. therefore, cannot connect to the extracellular matrix [5].
The main endogenous activators and inhibitors of angiogenesis are summarized in Table 1 and Table 2.
Certain studies have demonstrated that the thickness of the capillary’s basal lamina may lead to insufficient oxygen diffusion, limiting the elimination of some metabolites, eventually leading to the increase of different diseases’ severity [80,81]. Minchenko et al. [82] suggested that hypoxia is both inductor and stimulator of VEGF expression in vivo, along with the increase of glycemia [83].
VEGF also increases the microvascular permeability that precedes and accompanies angiogenesis, playing a central role in its process, acting as an anti-apoptotic factor for endothelial cells in newly formed blood vessels.
Many studies have highlighted the histological structural changes in the blood vessels of periodontopathic diabetic patients, but less data were reported on the number of abnormally evolution vessels (MVD) and/or their tissue distribution process, which can lead to tumor growth and the development of metastases (neo-angiogenesis).
Thus, in addition to the structural changes visible with light microscopy for the gingival blood capillaries, the number of blood vessels from the gingiva of the periodontal patients with diabetes mellitus (DM) can be displayed by quantitative immunohistochemical method and vascular markers.
We believe that the best immunohistochemical marker to highlight the newly formed blood vessels is the CD31+CD34 antibody cocktail which may show the entire micro-vascular network, as shown in Figure 1.

4. Physiological and Pathological Angiogenesis

The balance of activators and inhibitors is shown in Figure 2. Besides its role in embryonic development, angiogenesis appears as a normal process in adults, in female reproductive functions [84,85,86,87,88] and in tissue regeneration (ex. wounds healing) [89] (Figure 2B). Under certain conditions, it can appear as a disequilibrium in the synthesis of the endogenous factors that control angiogenesis, and an abnormal angiogenesis can occur. It can be either insufficient, leading to the impossibility of the body healing wounds or participating in normal organ regeneration, or it can lead to the ischemia of a part of the body, such as myocardial, peripheral of intestinal ischemia. Other complications include ulcers, infertility and hair loss (Figure 2A).
As mentioned above, excessive angiogenesis can favor tumor growth and metastasis appearance or it can induce the evolution of diseases such as rheumatoid arthritis, psoriasis, etc. (Figure 2C) [3,16].
Excessive angiogenesis is also linked to a series of eye diseases, which can lead to blindness. It is the cause of visual loss in the case of age-related macular degeneration. Patients who suffer from proliferative diabetic retinopathy are known to have higher VEGF levels when compared to healthy persons [90]. Increased VEGF levels cause uncontrolled angiogenesis in these patients.
Normal angiogenesis is the balanced action of angiogenesis activators and inhibitors. In the case of preponderant action of angiogenesis inhibitors, insufficient angiogenesis appears, leading to diseases such as chronic wounds, cardiovascular diseases, neuropathies, ulcers, hair loss, and infertility (Figure 2A). Physiological angiogenesis appears as a normal process in female reproductive functions and in tissue regeneration (Figure 2B). On the contrary, if the action of angiogenesis activators prevails, complications in diseases such as cancer, diabetic retinopathy, rheumatoid arthritis, AIDS, psoriasis, osteomyelitis or uterine bleeding may appear (Figure 2C).

5. Angiogenesis Inhibition (Anti-Angiogenic Therapy) and Current Implications in Cancer Treatment

The study of angiogenesis inhibitors gained much attention after the discovery by Judah Folkman showing that tumors do not grow bigger than 2–3 mm3 because of lack of nutrients, without supply of new blood vessels [91]. Indeed, under oxygen deprivation, tumors produce specific pro-angiogenic factors, which lead to the development of a new bold vasculature feeding the tumor and producing its growth, a phenomenon known as tumor angiogenesis [91,92].
Inhibiting the development of new blood vessels in tumors seemed to be an interesting path in fighting cancer. The anti-angiogenic therapy seemed a very promising target, but clinical medicine showed that, after years of practice, “the miracle drug” still did not show up. This might be due to a series of factors linked to the fact that tumors have unique properties that distinguish them from all other tissues.
Tumor vasculature is heterogeneous while tumor blood vessels are larger in size and more permeable than those of normal tissues [93]. The blood pressure is also different inside tumors: it is higher at the periphery, where arteries and arterioles are preponderant, and smaller in the core, where most blood vessels are veins and venules. Therefore, the access of both nutrients and medication through the blood stream is restrained to the margins of the tumor, while the interior, fed through diffusion, becomes necrotic.
A direct consequence of this heterogeneous structure inside tumors is that the liquids tend to accumulate in the interstitium of tumors, creating an increased interstitial fluid pressure (IFP) in the center of the tumor compared to the periphery [94,95]. This is why the liquid flow is oriented from the center to the margins of the tumor and the access of drugs to the tumor core is difficult [96,97]. This affects nanoparticles to a less extent than small drugs. Nanoparticles’ access to the tumor site is possible because blood vessels in tumors are permeable, a phenomenon known as the enhanced permeability and retention (EPR) effect, according to which macromolecules and nanoparticles tend to accumulate more in a tumoral tissue than in a healthy one [98]. The EPR effect represents the basis of the whole generation of nanoparticles intending to reach tumor sites through passive targeting.
All of these factors have made anti-angiogenic therapy an extremely complicated procedure. Two classes of angiogenesis inhibitors may be distinguished: direct inhibitors, that target endothelial cells (ECs), and indirect inhibitors, that target cancer cells or tumor-related stromal cells [99]. Indirect inhibitors act by disrupting the proangiogenic contact between tumor cells and ECs. Even though several mechanisms are known, most of these inhibitors act as anti-VEGF therapies, by blocking either VEGF itself or VEGF receptors (VEGFRs) [100]. These molecules are able to recognize and inhibit VEGF or VEGFRs. In Table 3 are presented the approved angiogenesis inhibitors used to treat cancer in humans, according to the National Cancer Institute [101].
Treatments with anti-VEGF drugs are known to have side-effects (hypertension, proteinuria, thromboembolisms, bleeding, etc.) [117,118] and treatment discontinuity causes vascular reformation in tumors, so new anti-angiogenesis targets are being investigated [119]. In patients treated with Avastin (the trade name for bevacizumab, a monoclonal antibody that acts as an anti-VEGF agent), temporary remission has been observed, but the overall survival time has not been effectively increased [120]. These patients developed resistance to treatment [121] and the investigation of the molecular mechanisms of resistance to anti-angiogenesis treatment has recently attracted great attention [122,123]. Anti-angiogenesis therapy showed better results combined with other treatments, the angiogenesis inhibitors acting in this case as agents that “normalize” the structure and functions of tumor vasculature, facilitating medication (usually chemotherapy) access inside tumors [124,125]. Avastin has been approved by the FDA as treatment for metastatic colorectal cancer combined with chemotherapy [126] and is also used in the treatment of other types of cancer (lung, ovarian, cervix, kidney, brain).
Besides the acquired resistance to chemotherapy and to anti-angiogenic treatments of cancer cells, another intriguing aspect regarding the difficulty of winning the fight against tumors is their incredible ways of finding previously unthinkable resources for opposing the healthy body through surprising “conquering strategies”. Indeed, when tumors exceed a size of several mm3, along with the formation of a new blood vasculature that brings oxygen and nutrients to cancer cells, an opposite phenomenon also takes place: tumor cells fight in order to escape the hypoxic area, and, through mechanisms involving Hepatocyte Growth Factor (HGF), a protein that links to the Met tyrosine kinase receptor, they leave the tumor and fix to distal sites, leading to metastasis [127]. Thus, new directions in cancer therapy tend to combine the administration of anti-angiogenic drugs with the anti-Met treatment [128,129]. The right amount and combination of medication and its well-scheduled administration could hopefully alleviate hypoxia and its non-desired side-effects [124].
The dual inhibition strategy using brivanib (dual FGF/VEGF inhibitor) increased overall survival (OS), and dovitinib (VEGFR, FGFR and PDGR inhibitor) retarded growth of tumors in mice cancer types. On the other hand, VEGF/ANG2 restricting/blocking leads to revascularization suppression and, consequently, the tumor evolution can be stopped, thus increasing OS [6,130].
Although some preclinical prototypes have been shown to be effective in addressing tumor angiogenesis, the complexity of tumor vascularization processes has led to less convenient clinical outcomes [6]. In order to prevent the recurrence of tumors, their surgical resection followed by chemotherapy and/or radiotherapy represents the standard procedure constituting the pathway to an eventual cure [6,131].

6. Angiogenesis Activation (Therapeutic Angiogenesis) and Current Implications in Cardiovascular Diseases

Cardiovascular diseases can be divided into cerebrovascular disease, coronary artery disease, peripheral arterial disease and aortic (thoracic or abdominal) atherosclerosis [132]. Usually, the problem is that of an occlusion of a vessel, followed by the ischemization of the surrounding tissue. The required medical intervention aims to restore the normal level of oxygen and nutrients in the affected area, either pharmacologically, or mechanically using surgical interventions (through vascular bypass and angioplasty). Some patients cannot be submitted to these treatments. Others do not respond to treatment as expected and cannot regain a high percentage of revascularization. In all of these cases, methods that could restore natural processes of blood vessel formation are highly welcomed.
Pro-angiogenic therapy would be, in this context, a very interesting opportunity [133]. However, excessive angiogenesis has to be avoided, as it can lead to systemic side-effects, which include the acceleration of diseases such as proliferative retinopathy or atherosclerosis [134].
Therapeutic angiogenesis can be achieved through three routes, as shown below.
  • Protein therapy: consists of the repeated administration of angiogenic factors in order to promote angiogenesis. This strategy has the advantage that angiogenic protein production and purification is known, and proteins may now be stored after lyophilization and reconditioned in a buffer upon need. Most of the angiogenic proteins are now commercialized and available for research. Besides VEGF and FGF (the most extensively studied proteins in therapeutic angiogenesis), other factors such as the PDGF family and Angiopoietin-1 have also been investigated. Systemic delivery of proteins has as a drawback the low concentration of the angiogenic factor in the desired tissue, due to both low targeting and rapid protein clearance by the mononuclear phagocyte system. Because of the rapid clearance of proteins in blood, their local delivery, either directly, or using adequate biomaterials with slow-delivery of active principles, is better suited in this case. Local administration (intracoronary, intramyocardial or intracerebral) is possible, but involves the usage of special devices or invasive surgery. Biomaterials of natural or synthetic origin such as hydrogels (e.g., alginate hydrogel and peptide nanofibers); micro- and nano-particles (e.g., poly (lactic acid-co-glycolic acid) microspheres and liposomes); porous scaffolds (e.g., poly (ε-caprolactone) scaffolds); coacervate (e.g., (polycation–heparin) coacervates), have been studied as delivery vehicles of angiogenic proteins [135].
  • Gene therapy consists of administrating genes whose expression would lead to proteins that will induce angiogenesis activation [136]. Gene therapy has as an advantage the fact that the protein continues to be secreted a long time after drug administration, as well as the fact that genes might be targeted to specific tissues [137]. Employed vector systems are plasmids and viral vectors. Adeno-associated viruses have been investigated as promising new vectors for gene therapy. The obstacles that have to be overcome are related to the low concentration of gene product at the target site as well as the activation of inflammatory and immune responses. Additional aspects, such as the incomplete understanding of angiogenesis mechanisms at molecular level and the difference between animal models and humans, represent the main obstacles encountered when angiogenesis gene therapy has been applied to humans. Although not yet implemented in clinical practice, data gathered in more than 20 years of preclinical and clinical studies has brought great insights and advancement in this field [138,139]. Several clinical trials investigating the effect of different gene therapies on cardiac regeneration are currently ongoing [140].
  • Cellular therapy [141,142] induces angiogenesis using cells known to produce angiogenic factors, such as monocytes and endothelial progenitor cells. Recent research indicates that stem cells-derived extracellular vesicles promote angiogenesis in cellular experiments and animal models. Extracellular vesicles transport informational molecules, including proteins, mRNA, microRNAs, DNA fragments, and lipids [143,144,145,146].
The ability of VEGF and FGF factors to induce therapeutic angiogenesis has been widely studied in preclinical and clinical trials [147]. Through vascular permeability increase and incorporation of endothelial cells, VEGF promotes angiogenesis [8]. The stimulation of endothelial cell layer growth covering the inner lumen of blood vessels, having an effect on the blood supply regularization according to local requirements, leads to the damaged vascular network reconstruction [148,149,150]. Thus, a good strategy for therapeutic angiogenesis can be designed in this direction [148,149,150]. It is considered that the “truthful” endothelial precursor is represented by endothelial colony forming cells (ECFCs) [148,151], which play an important role in conserving endothelial homeostasis and ameliorating ischemia impact on human health by regulating the local blood flow [148].
Studies indicate that cells with vasculo-reparative properties may represent an interesting target for potential therapies in vascular injury and impaired regenerative responses [152].
As shown in a recent review [148], clinical trials have accurately shown that cell therapy based on transplantation of myeloid EPCs fails to provide a significant improvement in capillary density and local blood flow in the case of patients with Cardio Vascular Diseases (CVD) [148].
Cardiac myocytes, known as cardiomyocytes (CM), are the muscle cells which compose the heart muscle and constitute a VGEF-A source that carries out certain functions of the heart [48]. In the case of rat CM, the mechanical stress controls VEGF-A expression and stretching contributes to secretion improvement, [48], involving hypoxia-inducible factor 1-α [48,153]. The absence of transcription factor, GATA-4 protein, that directly links VEGF-A. highlighted a low capillary density of mice hearts, while GATA4 overexpression led to vascularization regulation and cardiac function improvement after myocardial infarction [48].
Clinical trials show that therapeutic angiogenesis is generally safe and does not produce side-effects [154], but accidents may occur [155]. Initial results looked very promising, but most of the clinical trials were considered to be inconclusive. One drawback of reported clinical trials has been the lack of large enough control groups [156], while other clinical trials did not provide good enough positive results, as previously reviewed [157,158]. In future clinical trials, larger randomized groups are expected to be enrolled in order to obtain significant results and long-term studies still need to be finalized in order to have solid conclusions [159].

7. Conclusions

Normal angiogenesis is the exact balance of angiogenesis activators and inhibitors and its disequilibrium leads to life-threatening diseases or to diseases seriously affecting patients’ life-quality. In the beginning, a huge amount of attention was dedicated to the inhibition of angiogenesis, mainly in order to inhibit tumor angiogenesis. More recently, therapeutic angiogenesis has attracted great attention, due to its applications mainly in the field of ischemic tissue recovery.
The results of clinical trials prove that anti-angiogenic therapy alone will not be able to effectively treat cancer. Further insights into the molecular mechanisms of cancer resistance to anti-angiogenic therapy will provide important knowledge in this field. Future treatments for cancer can be successful only by combining different types of medication, through well scheduled therapeutic schemes that should try to overcome current limitations.
Therapeutic angiogenesis is under study, the most promising strategy being protein therapy or a combination of treatments involving protein therapy. The implementation of new biomaterials with specific local-delivery properties will probably bring important advancements in this field.

Author Contributions

C.I. and V.B.-M.; methodology, C.I., B.O. and V.B.-M.; software, C.I. and B.O.; validation, M.G., R.B., G.-O.M., F.H. and V.B.-M.; formal analysis, G.C.; investigation, C.I. and B.O.; resources, C.I. and B.O.; data curation, G.C., M.G., R.B., G.-O.M. and F.H; writing-original draft preparation, C.I. and B.O.; writing-review and editing, C.I., B.O., G.-O.M. and F.H.; visualization, G.C. and V.B.-M.; supervision, M.G., R.B., G.-O.M., F.H. and V.B.-M.; project administration, C.I. and B.O.; funding acquisition, C.I., B.O., G.C., M.G., R.B., G.-O.M., F.H. and V.B.-M. All authors had equal contribution to the paper to that of the first author. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data included in this study are identified within the text, tables, and figures.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. American Heart Association. New Statistics Show One of Every Three US Deaths Caused by Cardiovascular Disease. ScienceDaily. 16 December 2015. Available online: www.sciencedaily.com/releases/2015/12/151216144511.htm (accessed on 2 March 2021).
  2. Al Sabti, H. Therapeutic angiogenesis in cardiovascular disease. J. Cardiothorac. Surg. 2007, 2, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pandya, N.M.; Dhalla, N.S.; Santani, D.D. Angiogenesis—A new target for future therapy. Vascul. Pharmacol. 2006, 44, 265–274. [Google Scholar] [CrossRef] [PubMed]
  4. Bouıs, D.; Kusumanto, Y.; Meijer, C.; Mulder, N.H.; Hospers, G.A. A review on pro- and anti-angiogenic factors as targets of clinical intervention. Pharmacol. Res. 2006, 53, 89–103. [Google Scholar] [CrossRef] [PubMed]
  5. Al Kawas, H.; Saaid, I.; Jank, P.; Westhoff, C.C.; Denkert, C.; Pross, T.; Weiler, K.B.S.; Karsten, M.M. How VEGF-A and its splice variants affect breast cancer development—clinical implications. Cell Oncol. 2022, 45, 227–239. [Google Scholar] [CrossRef]
  6. Lopes-Coelho, F.; Martins, F.; Pereira, S.A.; Jacinta Serpa, J. Anti-Angiogenic Therapy: Current Challenges and Future Perspectives. Int. J. Mol. Sci. 2021, 22, 3765. [Google Scholar] [CrossRef]
  7. Peach, C.J.; Mignone, V.W.; Arruda, M.A.; Diana, C.; Alcobia, D.C.; Stephen, J.; Hill, S.J.; Kilpatrick, L.E.; Woolard, J. Molecular Pharmacology of VEGF-A Isoforms: Binding and Signalling at VEGFR2. Int. J. Mol. Sci. 2018, 19, 1264. [Google Scholar] [CrossRef] [Green Version]
  8. Zhou, Y.; Zhu, X.; Cui, H.; Shi, J.; Yuan, G.; Shi, S.; Hu, Y. The Role of the VEGF Family in Coronary Heart Disease. Front. Cardiovasc. Med. 2021, 8, 738325. [Google Scholar] [CrossRef]
  9. Ferrara, N.; Adamis, A.P. Ten years of anti-vascular endothelial growth factor therapy. Nat. Rev. Drug Discov. 2016, 15, 385–403. [Google Scholar] [CrossRef] [Green Version]
  10. Rosca, E.V.; Koskimaki, J.E.; Rivera, C.G.; Pandey, N.B.; Tamiz, A.P.; Popel, A.S. Anti-angiogenic peptides for cancer therapeutics. Curr. Pharm. Biotechnol. 2011, 12, 1101–1116. [Google Scholar] [CrossRef]
  11. Fan, T.P.; Jaggar, R.; Bicknell, R. Controlling the vasculature: Angiogenesis, anti-angiogenesis and vascular targeting of gene therapy. Trends Pharmacol. Sci. 1995, 16, 57–66. [Google Scholar] [CrossRef]
  12. Quesada, A.R.; Munoz-Chapuli, R.; Medina, M.A. Anti-Angiogenic Drugs: From Bench to Clinical Trials. Med. Res. Rev. 2006, 26, 483–530. [Google Scholar] [CrossRef] [PubMed]
  13. Eguchi, M.; Masuda, H.; Asahara, T. Endothelial progenitor cells for postnatal vasculogenesis. Clin. Exp. Nephrol. 2007, 11, 18–25. [Google Scholar] [CrossRef] [PubMed]
  14. Murasawa, S.; Asahara, T. Endothelial Progenitor Cells for Vasculogenesis. Physiology 2005, 20, 36–42. [Google Scholar] [CrossRef] [PubMed]
  15. Asahara, T.; Masuda, H.; Takahashi, T.; Kalka, C.; Pastore, C.; Silver, M.; Kearne, M.; Magner, M.; Isner, J.M. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ. Res. 1999, 85, 221–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Carmeliet, P. Angiogenesis in health and disease. Nat. Med. 2003, 9, 653–660. [Google Scholar] [CrossRef]
  17. Djonov, V.; Baum, O.; Burri, P.H. Vascular remodeling by intussusceptive angiogenesis. Cell Tissue Res. 2003, 314, 107–117. [Google Scholar] [CrossRef]
  18. Djonov, V.; Schmid, M.; Tschanz, S.A.; Burri, P.H. Intussusceptive Angiogenesis: Its Role in Embryonic Vascular Network Formation. Circ. Res. 2000, 86, 286–292. [Google Scholar] [CrossRef]
  19. Tomanek, R.J.; Schatteman, G.C. Angiogenesis: New insights and therapeutic potential. Anat. Rec. 2000, 261, 126–135. [Google Scholar] [CrossRef]
  20. Couffinhal, T.; Dufourcq, P.; Daret, D.; Duplaa, C. The mechanisms of angiogenesis. Medical and therapeutic applications. Rev. Méd. Interne 2001, 22, 1064–1082. [Google Scholar] [CrossRef]
  21. Bussolino, F.; Mantovani, A.; Persico, G. Molecular mechanisms of blood vessel formation. Trends Biochem. Sci. 1997, 22, 251–256. [Google Scholar] [CrossRef]
  22. Ribatti, D.; Crivellato, E. “Sprouting angiogenesis”, a reappraisal. Dev. Biol. 2012, 372, 157–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tanigawa, N.; Amaya, H.; Matsumura, M.; Shimomatsuya, T.; Horiuchi, T.; Muraoka, R.; Iki, M. Extent of tumor vascularization correlates with prognosis and hematogenous metastasis in gastric carcinomas. Cancer Res. 1996, 56, 2671–2676. [Google Scholar] [PubMed]
  24. Scholz, D.; Cai, W.J.; Schaper, W. Arteriogenesis, a new concept of vascular adaptation in occlusive disease. Angiogenesis 2001, 4, 247–257. [Google Scholar] [CrossRef]
  25. Heil, M.; Wagner, S.; Schaper, W. Arterial regeneration by collateral artery growth (arteriogenesis). Drug Discov. Today Dis. Models 2004, 1, 265–271. [Google Scholar] [CrossRef]
  26. Stephan, D.; Weltin, D.; Zaric, V.; Chapelon, D.; Da Silva, A.; Lugnier, C. Angiogenèse: De la physiologie à la thérapeutique. Réanim. Urgences 2000, 9, 534–544. [Google Scholar] [CrossRef]
  27. Levy, A.P.; Levy, N.S.; Goldberg, N.A. Post-transcriptional Regulation of Vascular Endothelial Growth Factor by Hypoxia. J. Biol. Chem. 1996, 271, 2746–2753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Nangia-Makker, P.; Baccarini, S.; Raz, A. Carbohydrate-recognition and angiogenesis. Cancer Metastasis Rev. 2000, 19, 51–57. [Google Scholar] [CrossRef]
  29. Funasaka, T.; Raz, A.; Nangia-Makker, P. Galectin-3 in angiogenesis and metastasis. Glycobiology 2014, 24, 886–891. [Google Scholar] [CrossRef] [Green Version]
  30. Nangia-Makker, P.; Hogan, V.; Honjo, Y.; Baccarini, S.; Tait, L.; Bresalier, R.; Raz, A. Inhibition of human cancer cell growth and metastasis in nude mice by oral intake of modified citrus pectin. J. Natl. Cancer. Inst. 2002, 94, 1854–1862. [Google Scholar] [CrossRef] [Green Version]
  31. Johnstone, K.D.; Karoli, T.; Liu, L.; Dredge, K.; Copeman, E.; Li, C.P.; Davis, K.; Hammond, E.; Bytheway, I.; Kostewicz, E.; et al. Synthesis and biological evaluation of polysulfated oligosaccharide glycosides as inhibitors of angiogenesis and tumor growth. J. Med. Chem. 2010, 53, 1686–1699. [Google Scholar] [CrossRef] [Green Version]
  32. Barragan-Montero, V.; Awwad, A.; Combemale, S.; de Santa Barbara, P.; Jover, B.; Molès, J.P.; Montero, J.L. Synthesis of Mannose-6-Phosphate Analogues and their Utility as Angiogenesis Regulators. ChemMedChem 2011, 6, 1771–1774. [Google Scholar] [CrossRef] [PubMed]
  33. Ionescu, C.; Sippelli, S.; Toupet, L.; Barragan-Montero, V. New mannose derivatives: The tetrazole analogue of mannose-6-phosphate as angiogenesis inhibitor. Bioorg. Med. Chem. Lett. 2016, 26, 636–639. [Google Scholar] [CrossRef] [PubMed]
  34. Combemale, S.; Assam-Evoung, J.N.; Houaidji, S.; Bibi, R.; Barragan-Montero, V. Gold Nanoparticles Decorated with Mannose-6-phosphate Analogues. Molecules 2014, 19, 1120–1149. [Google Scholar] [CrossRef] [Green Version]
  35. Volpert, O.; Jackson, D.; Bouck, N.; Linzer, D.I. The insulin-like growth factor II/Mannose 6 -phosphate receptor is required for proliferin-induced angiogenesis. Endocrinology 1996, 137, 3871–3876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Doyagüez, G.E.; Carrero, P.; Madrona, A.; Rodriguez-Salamanca, P.; Martínez-Gualda, B.; Camarasa, M.J.; Jimeno, M.L.; Bennallack, P.R.; Finnell, J.G.; Tsang, T.M.; et al. Galloyl Carbohydrates with Antiangiogenic Activity Mediated by Capillary Morphogenesis Gene 2 (CMG2) Protein Binding. J. Med. Chem. 2019, 62, 3958–3970. [Google Scholar] [CrossRef] [PubMed]
  37. Cryan, L.M.; Bazinet, L.; Habeshian, K.A.; Cao, S.; Clardy, J.; Christensen, K.A.; Rogers, M.S. 1,2,3,4,6-Penta-O-galloyl-β-D-glucopyranose inhibits angiogenesis via inhibition of capillary morphogenesis gene 2. J. Med. Chem. 2013, 56, 1940–1945. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Huh, J.E.; Lee, E.O.; Kim, M.S.; Kang, K.S.; Kim, C.H.; Cha, B.C.; Surh, Y.J.; Kim, S.H. Penta-O-galloyl-beta-D-glucose suppresses tumor growth via inhibition of angiogenesis and stimulation of apoptosis: Roles of cyclooxygenase-2 and mitogen-activated protein kinase pathways. Carcinogenesis 2005, 26, 1436–1445. [Google Scholar] [CrossRef]
  39. Nyberg, P.; Xie, L.; Kalluri, R. Endogenous Inhibitors of Angiogenesis. Cancer Res. 2005, 65, 3967–3979. [Google Scholar] [CrossRef] [Green Version]
  40. Iruela-Arispe, M.L.; Dvorak, H.F. Angiogenesis: A dynamic balance of stimulators and inhibitors. Thromb. Haemost. 1997, 78, 672–677. [Google Scholar] [CrossRef]
  41. Gospodarowicz, D. Purification of a fibroblast growth factor from bovine pituitary. J. Biol. Chem. 1975, 250, 2515–2520. [Google Scholar] [CrossRef]
  42. Esch, F.; Baird, A.; Ling, N.; Ueno, N.; Hill, F.; Denoroy, L.; Klepper, R.; Gospodarowicz, D.; Böhlen, P.; Guillemin, R. Primary structure of bovine pituitary basic fibroblast growth factor (FGF) and comparison with the amino-terminal sequence of bovine brain acidic FGF. Proc. Natl. Acad. Sci. USA 1985, 82, 6507–6511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Senger, D.R.; Galli, S.J.; Dvorak, A.M.; Perruzzi, C.A.; Harvey, V.S.; Dvorak, H.F. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983, 219, 983–9855. [Google Scholar] [CrossRef]
  44. Leung, D.W.; Cachianes, G.; Kuang, W.J.; Goeddel, D.V.; Ferrara, N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989, 246, 1306–1309. [Google Scholar] [CrossRef]
  45. Guo, N.; Krutzsch, H.C.; Inman, J.K.; Roberts, D.D. Thrombospondin 1 and type I repeat peptides of thrombospondin 1 specifically induce apoptosis of endothelial cells. Cancer Res. 1997, 57, 1735–1742. [Google Scholar]
  46. Good, D.J.; Polverini, P.J.; Rastinejad, F.; Le Beau, M.M.; Lemons, R.S.; Frazier, W.A.; Bouck, N.P. A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proc. Natl. Acad. Sci. USA 1990, 87, 6624–6628. [Google Scholar] [CrossRef] [Green Version]
  47. Mateescu, G.O.; Comanescu, M.; Mehedinţi, R.; Niculescu, Z.; Bold, A.; Panduru, L.; Cernea, D. Immunohistochemical expression of growth factors in the exocrine pancreas of patients with chronic liver diseases. Rom. J. Morphol. Embryol. 2010, 51, 303–307. [Google Scholar]
  48. Braile, M.; Marcella, S.; Cristinziano, L.; Galdiero, M.R.; Luca Modestino, L.; Ferrara, A.L.; Varricchi, G.; Marone, G.; Loffredo, S. VEGF-A in Cardiomyocytes and Heart Diseases. Int. J. Mol. Sci. 2020, 21, 5294. [Google Scholar] [CrossRef]
  49. Manetti, F.; Corelli, F.; Botta, M. Fibroblast growth factors and their inhibitors. Curr. Pharm. Des. 2000, 6, 1897–1924. [Google Scholar] [CrossRef] [PubMed]
  50. Zakrzewska, M.; Marcinkowska, E.; Wiedlocha, A. FGF-1: From biology through engineering to potential medical applications. Crit. Rev. Clin. Lab. Sci. 2008, 45, 91–135. [Google Scholar] [CrossRef] [PubMed]
  51. Yayon, A.; Klagsbrun, M.; Esko, J.D.; Leder, P.; Omitz, D.M. Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor. Cell 1991, 64, 841–848. [Google Scholar] [CrossRef]
  52. Roghani, M.; Mansukhani, A.; Dell’Era, P.; Bellosta, P.; Basilico, C.; Rifkin, D.B.; Moscatelli, D. Heparin increases the affinity of basic fibroblast growth factor for its receptor but is not required for binding. J. Biol. Chem. 1994, 269, 3976–3984. [Google Scholar] [CrossRef]
  53. Friesel, R.E.; Maciag, T. Molecular mechanisms of angiogenesis: Fibroblast growth factor signal transduction. FASEB J. 1995, 9, 919–925. [Google Scholar] [CrossRef] [PubMed]
  54. Hagedorn, M.; Bikfalvi, A. Target molecules for anti-angiogenic therapy: From basic research to clinical trials. Crit. Rev. Oncol. Hematol. 2000, 34, 89–110. [Google Scholar] [CrossRef]
  55. Meyer, M.; Clauss, M.; Lepple-Wienhues, A.; Waltenberger, J.; Augustin, H.G.; Ziche, M.; Lanz, C.; Büttner, M.; Rziha, H.J.; Dehio, C. A novel vascular endothelial growth factor encoded by Orf virus, VEGF-E, mediates angiogenesis via signalling through VEGFR-2 (KDR) but not VEGFR-1 (Flt-1) receptor tyrosine kinases. EMBO J. 1999, 18, 363–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Veikkola, T.; Alitalo, K. VEGFs, receptors and angiogenesis. Semin. Cancer Bio. 1999, 9, 211–220. [Google Scholar] [CrossRef]
  57. Koblizek, T.I.; Weiss, C.; Yancopoulos, G.D.; Deutsch, U.; Risau, W. Angiopoietin-1 induces sprouting angiogenesis in vitro. Curr. Biol. 1998, 8, 529–532. [Google Scholar] [CrossRef] [Green Version]
  58. Maisonpierre, P.C.; Suri, C.; Jones, P.F.; Bartunkova, S.; Wiegand, S.J.; Radziejewski, C.; Compton, D.; McClain, J.; Aldrich, T.H.; Papadopoulos, N.; et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997, 277, 55–60. [Google Scholar] [CrossRef]
  59. Yu, X.; Seegar, T.M.C.; Dalton, A.C.; Tzvetkova-Robev, D.; Goldgur, Y.; Rajashankar, K.R.; Nikolov, D.B.; Barton, W.A. Structural basis for angiopoietin-1–mediated signaling initiation. Proc. Natl. Acad. Sci. USA 2013, 110, 7205–7210. [Google Scholar] [CrossRef] [Green Version]
  60. Fagiani, E.; Christofori, G. Angiopoietins in angiogenesis. Cancer Lett. 2013, 328, 18–26. [Google Scholar] [CrossRef]
  61. Cheng, N.; Brantley, D.M.; Chen, J. The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev. 2002, 13, 75–85. [Google Scholar] [CrossRef]
  62. Salvucci, O.; Tosato, G. Essential roles of EphB receptors and EphrinB ligands in endothelial cell function and angiogenesis. Adv. Cancer Res. 2012, 114, 21–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kullander, K.; Klein, R. Mechanisms and functions of Eph and ephrin signalling. Nat. Rev. Mol. Cell Biol. 2002, 3, 475–486. [Google Scholar] [CrossRef] [PubMed]
  64. Zitka, O.; Kukacka, J.; Krizkova, S.; Huska, D.; Adam, V.; Masarik, M.; Prusa, R.; Kizek, R. Matrix metalloproteinases. Curr. Med. Chem. 2010, 17, 3751–3768. [Google Scholar] [CrossRef] [PubMed]
  65. Johnson, L.L.; Dyer, R.; Hupe, D.J. Matrix metalloproteinases. Curr. Opin. Chem. Biol. 1998, 2, 466–471. [Google Scholar] [CrossRef]
  66. Boire, A.; Covic, L.; Agarwal, A.; Jacques, S.; Sherifi, S.; Kuliopulos, A. PAR1 is a matrix metalloprotease-1 receptor that promotes invasion and tumorigenesis of breast cancer cells. Cell 2005, 120, 303–313. [Google Scholar] [CrossRef] [Green Version]
  67. Sasaki, T.; Larsson, H.; Kreuger, J.; Salmivirta, M.; Claesson-Welsh, L.; Lindahl, U.; Hohenester, E.; Timpl, R. Structural basis and potential role of heparin/heparan sulfate binding to the angiogenesis inhibitor endostatin. EMBO J. 1999, 18, 6240–6248. [Google Scholar] [CrossRef] [Green Version]
  68. O’Reilly, M.S.; Boehm, T.; Shing, Y.; Fukai, N.; Vasios, G.; Lane, W.S.; Flynn, E.; Birkhead, J.R.; Olsen, B.R.; Folkman, J. Endostatin: An endogenous inhibitor of angiogenesis and tumor growth. Cell 1997, 88, 277–285. [Google Scholar] [CrossRef] [Green Version]
  69. Kim, Y.M.; Jang, J.W.; Lee, O.H.; Yeon, J.; Choi, E.Y.; Kim, K.W.; Lee, S.-T.; Kwon, Y.G. Endostatin inhibits endothelial and tumor cellular invasion by blocking the activation and catalytic activity of matrix metalloproteinase. Cancer Res. 2000, 60, 5410–5413. [Google Scholar]
  70. Sudhakar, A.; Sugimoto, H.; Yang, C.; Lively, J.; Zeisberg, M.; Kalluri, R. Human tumstatin and human endostatin exhibit distinct antiangiogenic activities mediated by αvβ3 and α5β1 integrins. Proc. Natl. Acad. Sci. USA 2003, 100, 4766–4771. [Google Scholar] [CrossRef] [Green Version]
  71. O’Reilly, M.S.; Holmgren, L.; Shing, Y.; Chen, C.; Rosenthal, R.A.; Moses, M.; Lane, W.S.; Cao, Y.; Sage, E.; Folkman, J. Angiostatin: A novel angiogenesis inhibitor that mediates the suppression of metastases by a Lewis lung carcinoma. Cell 1994, 79, 315–328. [Google Scholar] [CrossRef]
  72. Geiger, J.H.; Cnudde, S.E. What the structure of angiostatin may tell us about its mechanism of action. J. Thromb. Haemost. 2004, 2, 23–34. [Google Scholar] [CrossRef] [PubMed]
  73. Troyanovsky, B.; Levchenko, T.; Mansson, G.; Matvijenko, O.; Holmgren, L. Angiomotin: An angiostatin binding protein that regulates endothelial cell migration and tube formation. J. Cell Biol. 2001, 152, 1247–1254. [Google Scholar] [CrossRef] [PubMed]
  74. Iruela-Arispe, M.L.; Lombardo, M.; Krutzsch, H.C.; Lawler, J.; Roberts, D.D. Inhibition of angiogenesis by thrombospondin-1 is mediated by 2 independent regions within the type 1 repeats. Circulation 1999, 100, 1423–1431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Adams, J.; Lawler, J. The thrombospondin family. Curr. Biol. 1993, 3, 188–190. [Google Scholar] [CrossRef]
  76. Bornstein, P. Thrombospondins function as regulators of angiogenesis. J. Cell Commun. Signal 2009, 3, 189–200. [Google Scholar] [CrossRef] [Green Version]
  77. Mirochnik, Y.; Kwiatek, A.; Volpert, O.V. Thrombospondin and apoptosis: Molecular mechanisms and use for design of complementation treatments. Curr. Drug Targets 2008, 9, 851–862. [Google Scholar] [CrossRef] [Green Version]
  78. Pribluda, V.S.; Gubish, E.R., Jr.; Lavallee, T.M.; Treston, A.; Swartz, G.M.; Green, S.J. 2-Methoxyestradiol: An endogenous antiangiogenic and antiproliferative drug candidate. Cancer Metastasis Rev. 2000, 19, 173–179. [Google Scholar] [CrossRef]
  79. Yue, T.L.; Wang, X.; Louden, C.S.; Gupta, S.; Pillarisetti, K.; Gu, J.L.; Hart, T.K.; Lysko, P.G.; Feuerstein, G.Z. 2-Methoxyestradiol, an endogenous estrogen metabolite, induces apoptosis in endothelial cells and inhibits angiogenesis: Possible role for stress-activated protein kinase signaling pathway and Fas expression. Mol. Pharmacol. 1997, 51, 951–962. [Google Scholar] [CrossRef]
  80. Oliver, R.C.; Tervonen, T. Diabetes-a risk factor for periodontitis in adults? J. Periodontol. 1994, 65, 530–538. [Google Scholar] [CrossRef]
  81. Yalda, B.; Offenbacher, S.; Collins, J.G. Diabetes as a modifier of periodontal disease expression. Periodontology 2000 1994, 6, 37–49. [Google Scholar] [CrossRef]
  82. Minchenko, A.; Bauer, T.; Salceda, S.; Caro, J. Hypoxic stimulation of vascular endothelial growth factor expression in vitro and in vivo. Lab. Investig. 1994, 71, 374–379. Available online: https://pubmed.ncbi.nlm.nih.gov/7933988/ (accessed on 4 July 2022). [PubMed]
  83. Teshima-Kondo, S.; Kondo, K.; Prado-Lourenco, L.; Gonzalez-Herrera, I.; Rokutan, K.; Bayard, F.; Arnal, J.F.; Prats, A.C. Hyperglycemia up-regulates translation of the fibroblast growth factor 2 mRNA in mouse aorta via internal ribosome entry site. FASEB J. 2004, 18, 1583–1585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Groothius, P.G. Angiogenesis and vascular remodelling in female reproductive organs. Angiogensis 2005, 8, 87–88. [Google Scholar] [CrossRef] [PubMed]
  85. Gargett, C.E.; Rogers, P.A.W. Human endometrial angiogenesis. Reproduction 2001, 121, 181–186. [Google Scholar] [CrossRef] [PubMed]
  86. Okada, H.; Tsuzuki, T.; Shindoh, H.; Nishigaki, A.; Yasuda, K.; Kanzaki, H. Regulation of decidualization and angiogenesis in the human endometrium: Mini review. J. Obstet. Gynaecol. Res. 2014, 40, 1180–1187. [Google Scholar] [CrossRef]
  87. Chung, A.S.; Lee, J.; Ferrara, N. Targeting the tumour vasculature: Insights from physiological angiogenesis. Nat. Rev. Cancer 2010, 10, 505–514. [Google Scholar] [CrossRef] [PubMed]
  88. Boldeanu, L.; Dijmărescu, A.L.; Radu, M.; Siloşi, C.A.; Popescu-Drigă, M.V.; Poenariu, I.S.; Siloşi, I.; Boldeanu, M.V.; Novac, M.B.; Novac, L.V. The role of mediating factors involved in angiogenesis during implantation. Rom. J. Morphol. Embryol. 2020, 61, 665–672. [Google Scholar] [CrossRef]
  89. Singer, A.J.; Clark, R.A. Cutaneous Wound Healing. N. Engl. J. Med. 1999, 341, 738–746. [Google Scholar] [CrossRef]
  90. Adamis, A.P.; Miller, J.W.; Bernal, M.T. Increased vascular endothelial growth factor levels in the vitreous of eyes with proliferative diabetic retinopathy. Am. J. Ophthalmol. 1994, 118, 445–450. [Google Scholar] [CrossRef]
  91. Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef]
  92. Folkman, J.; Shing, Y. Angiogenesis. J. Biol. Chem. 1992, 267, 10931–10934. [Google Scholar] [CrossRef]
  93. Zhang, J.; Lan, C.Q.; Post, M.; Simard, B.; Deslandes, Y.; Hseih, T.H. Design of Nanoparticles as Drug Carriers for Cancer Therapy. Cancer Genom. Proteom. 2006, 3, 147–158. [Google Scholar]
  94. Heldin, C.H.; Rubin, K.; Pietras, K.; Ostman, A. High interstitial fluid pressure—An obstacle in cancer therapy. Nat. Rev. Cancer 2004, 4, 806–813. [Google Scholar] [CrossRef]
  95. Danhier, F.; Feron, O.; Préat, V. To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. J. Control Release 2010, 148, 135–146. [Google Scholar] [CrossRef] [PubMed]
  96. Jain, R.K. Transport of molecules across tumor vasculature. Cancer Metastasis Rev. 1987, 6, 559–593. [Google Scholar] [CrossRef]
  97. Jain, R.K. Transport of molecules in the tumor interstitium: A review. Cancer Res. 1987, 47, 3039–3051. [Google Scholar]
  98. Maeda, H. The enhanced permeability and retention (EPR) effect in tumor vasculature: The key role of tumor-selective macromolecular drug targeting. Adv. Enzyme Regul. 2001, 41, 189–207. [Google Scholar] [CrossRef]
  99. Al-Ostoot, F.H.; Salah, S.; Khamees, H.A.; Khanum, S.A. Tumor angiogenesis: Current challenges and therapeutic opportunities, Cancer Treat. Res. Commun. 2021, 28, 100422. [Google Scholar] [CrossRef]
  100. 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]
  101. National Cancer Institute. Angiogenesis Inhibitors. Available online: https://www.cancer.gov/about-cancer/treatment/types/immunotherapy/angiogenesis-inhibitors-fact-sheet (accessed on 4 July 2022).
  102. Drugbank Online, General Site. Available online: https://go.drugbank.com/drugs (accessed on 4 July 2022).
  103. Keating, G.M. Axitinib: A review in advanced renal cell carcinoma. Drugs 2015, 75, 1903–1913. [Google Scholar] [CrossRef]
  104. Tewari, K.S.; Burger, R.A.; Enserro, D.; Norquist, B.M.; Swisher, E.M.; Brady, M.F.; Bookman, M.A.; Fleming, G.F.; Huang, H.; Homesley, H.D.; et al. Final Overall Survival of a Randomized Trial of Bevacizumab for Primary Treatment of Ovarian Cancer. J. Clin. Oncol. 2019, 37, 2317–2328. [Google Scholar] [CrossRef]
  105. Pal, S.K.; Tangen, C.; Thompson, I.M., Jr.; Balzer-Haas, N.; George, D.J.; Heng, D.Y.C.; Shuch, B.; Stein, M.; Tretiakova, M.; Humphrey, P.; et al. A comparison of sunitinib with cabozantinib, crizotinib, and savolitinib for treatment of advanced papillary renal cell carcinoma: A randomised, open-label, phase 2 trial. Lancet 2021, 397, 695–703. [Google Scholar] [CrossRef]
  106. Adib, E.; Klonowska, K.; Giannikou, K.; Do, K.T.; Pruitt-Thompson, S.; Bhushan, K.; Milstein, M.I.; Hedglin, J.; Kargus, K.E.; Sholl, L.M.; et al. Phase II Clinical Trial of Everolimus in a Pan-Cancer Cohort of Patients with mTOR Pathway Alterations. Clin. Cancer Res. 2021, 27, 3845–3853. [Google Scholar] [CrossRef] [PubMed]
  107. Mateos, M.-V.; Hernández, M.-T.; Giraldo, P.; De La Rubia, J.; De Arriba, F. Lenalidomide plus Dexamethasone for High-Risk Smoldering Multiple Myeloma. N. Engl. J. Med. 2013, 369, 438–447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Schlumberger, M.; Tahara, M.; Wirth, L.J.; Robinson, B.; Brose, M.S. Lenvatinib versus Placebo in Radioiodine-Refractory Thyroid Cancer. N. Engl. J. Med. 2015, 372, 621–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Ward, J.E.; Stadler, W.M. Pazopanib in renal cell carcinoma. Clin. Cancer. Res. 2010, 16, 5923–5927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Verdaguer, H.; Tabernero, J.; Macarulla, T. Ramucirumab in metastatic colorectal cancer: Evidence to date and place in therapy. Ther. Adv. Med. Oncol. 2016, 8, 230–242. [Google Scholar] [CrossRef] [Green Version]
  111. Aljubran, A.; Elshenawy, M.A.; Kandil, M.; Zahir, M.N.; Shaheen, A.; Gad, A.; Alshaer, O.; Alzahrani, A.; Eldali, A.; Bazarbashi, S. Efficacy of Regorafenib in Metastatic Colorectal Cancer: A Multi-institutional Retrospective Study. Clin. Med. Insights Oncol. 2019, 13, 1179554918825447. [Google Scholar] [CrossRef]
  112. Keating, G.M.; Santoro, A. Sorafenib: A review of its use in advanced hepatocellular carcinoma. Drugs 2009, 69, 223–240. [Google Scholar] [CrossRef]
  113. Mulet-Margalef, N.; Garcia-Del-Muro, X. Sunitinib in the treatment of gastrointestinal stromal tumor: Patient selection and perspectives. Onco. Targets Ther. 2016, 9, 7573–7582. [Google Scholar] [CrossRef] [Green Version]
  114. Breitkreutz, I.; Anderson, K.C. Thalidomide in multiple myeloma-clinical trials and aspects of drug metabolism and toxicity. Expert Opin. Drug. Metab. Toxicol. 2008, 4, 973–985. [Google Scholar] [CrossRef] [PubMed]
  115. Kim, M.; Yoon, J.H.; Ahn, J.; Jeon, M.J.; Kim, H.K.; Lim, D.J.; Kang, H.C.; Kim, I.J.; Shong, Y.K.; Kim, T.Y.; et al. Vandetanib for the Management of Advanced Medullary Thyroid Cancer: A Real-World Multicenter Experience. Endocrinol. Metab. 2020, 35, 587–594. [Google Scholar] [CrossRef] [PubMed]
  116. Perkins, S.L.; Cole, S.W. Ziv-aflibercept (Zaltrap) for the treatment of metastatic colorectal cancer. Ann. Pharmacother. 2014, 48, 93–98. [Google Scholar] [CrossRef]
  117. Kamba, T.; McDonald, D.M. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br. J. Cancer 2007, 96, 1788–1795. [Google Scholar] [CrossRef]
  118. Yu, I.; Chen, L.; Ruan, J.Y.; Chang, J.T.; Cheung, W.Y. Risk and management of venous thromboembolisms in bevacizumab-treated metastatic colorectal cancer patients. Support Care Cancer 2016, 24, 1199–1208. [Google Scholar] [CrossRef]
  119. Yamamizu, K.; Hamada, Y.; Narita, M. Opioid receptor ligands regulate angiogenesis in development and in tumours. Br. J. Pharmaco. 2015, 172, 268–276. [Google Scholar] [CrossRef] [Green Version]
  120. Norden, A.D.; Drappatz, J.; Muzikansky, A.; David, K.; Gerard, M.; McNamara, M.B.; Phan, P.; Ross, A.; Kesari, S.; Wen, P.Y. An exploratory survival analysis of anti-angiogenic therapy for recurrent malignant glioma. J. Neurooncol. 2009, 92, 149–155. [Google Scholar] [CrossRef]
  121. Lu, K.V.; Bergers, G. Mechanisms of evasive resistance to anti-VEGF therapy in glioblastoma. CNS Oncol. 2013, 2, 49–65. [Google Scholar] [CrossRef] [PubMed]
  122. Bergers, G.; Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 2008, 8, 592–603. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, M.; Ye, G.; Li, J.; Wang, Y. Recent advance in molecular angiogenesis in glioblastoma: The challenge and hope for anti-angiogenic therapy. Brain Tumor Pathol. 2015, 32, 229–236. [Google Scholar] [CrossRef]
  124. Jain, R.K. Normalization of tumor vasculature: An emerging concept in antiangiogenic therapy. Science 2005, 307, 58–62. [Google Scholar] [CrossRef] [PubMed]
  125. Moserle, L.; Jiménez-Valerio, G.; Casanovas, O. Antiangiogenic Therapies: Going beyond Their Limits. Cancer Discov. 2014, 4, 31–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. McCarthy, M. Antiangiogenesis drug promising for metastatic colorectal cancer. Lancet 2003, 361, 1959. [Google Scholar] [CrossRef]
  127. Michieli, P. Hypoxia, angiogenesis and cancer therapy: To breathe or not to breathe? Cell Cycle 2009, 8, 3291–3296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Hayden, E.C. Cutting off cancer’s supply lines. Nature 2009, 458, 686–687. [Google Scholar] [CrossRef]
  129. You, W.K.; McDonald, D.M. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008, 41, 833–839. [Google Scholar] [CrossRef] [Green Version]
  130. Hidalgo, M.; Martinez-Garcia, M.; Le Tourneau, C.; Massard, C.; Garralda, E.; Boni, V.; Taus, A.; Albanell, J.; Sablin, M.P.; Alt, M.; et al. First-in-Human Phase i Study of Single-Agent Vanucizumab, a First-in-Class Bispecific Anti-Angiopoietin-2/Anti-Vegf-a Antibody, in Adult Patients with Advanced Solid Tumors. Clin. Cancer Res. 2018, 24, 1536–1545. [Google Scholar] [CrossRef] [Green Version]
  131. Kajiyama, H.; Suzuki, S.; Yoshihara, M.; Nishino, K.; Yoshikawa, N.; Utsumi, F.; Niimi, K.; Mizuno, M.; Kawai, M.; Oguchi, H.; et al. The Possible Existence of Occult Metastasis in Patients with Ovarian Clear-Cell Carcinoma Who Underwent Complete Resection without Any Residual Tumours. Oncotarget 2018, 9, 6298–6630. [Google Scholar] [CrossRef] [Green Version]
  132. Deveza, L.; Choi, J.; Yang, F. Therapeutic Angiogenesis for Treating Cardiovascular Diseases. Theranostics 2012, 2, 801–814. [Google Scholar] [CrossRef]
  133. Kawasuji, M. Therapeutic Angiogenesis for Ischemic Heart Disease. Ann. Thorac. Cardiovasc. Surg. 2002, 8, 59–61. [Google Scholar]
  134. Tabibiazar, R.; Rockson, S.G. Angiogenesis and the ischaemic heart. Eur. Heart J. 2001, 22, 903–918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chu, H.; Wang, Y. Therapeutic angiogenesis: Controlled delivery of angiogenic factors. Ther. Deliv. 2012, 3, 693–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Nordlie, M.A.; Wold, L.E.; Simkhovich, B.Z.; Sesti, C.; Kloner, R.A. Molecular Aspects of Ischemic Heart Disease: Ischemia/Reperfusion–Induced Genetic Changes and Potential Applications of Gene and RNA Interference Therapy. J. Cardiovasc. Pharmacol. Therapeut. 2006, 11, 17–30. [Google Scholar] [CrossRef] [PubMed]
  137. Lewis, B.S.; Flugelman, M.Y.; Weisz, A.; Keren-Tal, I.; Schaper, W. Angiogenesis by gene therapy: A new horizon for myocardial revascularization? Cardiovasc. Res. 1997, 35, 490–497. [Google Scholar] [CrossRef] [Green Version]
  138. Sanada, F.; Taniyama, Y.; Muratsu, J.; Otsu, R.; Shimizu, H.; Rakugi, H.; Morishita, R. Gene-Therapeutic Strategies Targeting Angiogenesis in Peripheral Artery Disease. Medicines 2018, 5, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ylä-Herttuala, S.; Bridges, C.; Katz, M.G.; Korpisalo, P. Angiogenic gene therapy in cardiovascular diseases: Dream or vision? Eur. Heart. J. 2017, 38, 1365–1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Kim, Y.; Zharkinbekov, Z.; Sarsenova, M.; Yeltay, G.; Saparov, A. Recent Advances in Gene Therapy for Cardiac Tissue Regeneration. Int. J. Mol. Sci. 2021, 22, 9206. [Google Scholar] [CrossRef] [PubMed]
  141. Dai, W.; Kay, G.L.; Kloner, R.A. The Therapeutic Effect of Cell Transplantation Versus Noncellular Biomaterial Implantation on Cardiac Structure and Function Following Myocardial Infarction. J. Cardiovasc. Pharmacol. Ther. 2014, 19, 350–357. [Google Scholar] [CrossRef]
  142. Tse, H.F.; Lau, C.P. Therapeutic Angiogenesis With Bone Marrow—Derived Stem Cells. J. Cardiovasc. Pharmacol. Ther. 2007, 12, 89–97. [Google Scholar] [CrossRef]
  143. Bian, X.; Ma, K.; Zhang, C.; Fu, X. Therapeutic angiogenesis using stem cell-derived extracellular vesicles: An emerging approach for treatment of ischemic diseases. Stem Cell Res. Ther. 2019, 10, 158. [Google Scholar] [CrossRef]
  144. Merino-González, C.; Zuñiga, F.A.; Escudero, C.; Ormazabal, V.; Reyes, C.; Nova-Lamperti, E.; Salomón, C.; Aguayo, C. Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Angiogenesis: Potencial Clinical Application. Front. Physiol. 2016, 7, 24. [Google Scholar] [CrossRef] [Green Version]
  145. Kim, J.Y.; Rhim, W.K.; Yoo, Y.I.; Kim, D.S.; Ko, K.W.; Heo, Y.; Park, C.G.; Han, D.K. Defined MSC exosome with high yield and purity to improve regenerative activity. J. Tissue Eng. 2021, 12, 20417314211008626. [Google Scholar] [CrossRef] [PubMed]
  146. Hade, M.D.; Suire, C.N.; Suo, Z. Mesenchymal Stem Cell-Derived Exosomes: Applications in Regenerative Medicine. Cells 2021, 10, 1959. [Google Scholar] [CrossRef]
  147. Jo, D.H.; Kim, J.H. Toward the clinical application of therapeutic angiogenesis against pediatric ischemic retinopathy. J. Lipid Atheroscler. 2020, 9, 268–282. [Google Scholar] [CrossRef] [PubMed]
  148. Moccia, F.; Antognazza, M.R.; Lodola, F. Towards Novel Geneless Approaches for Therapeutic Angiogenesis. Front. Physiol. 2021, 11, 616189. [Google Scholar] [CrossRef] [PubMed]
  149. Qadura, M.; Terenzi, D.C.; Verma, S.; Al-Omran, M.; Hess, D.A. Concise review: Cell therapy for critical limb ischemia: An integrated review of preclinical and clinical studies: Stem cell therapy for critical limb ischemia. Stem Cells 2018, 36, 161–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Prasad, M.; Corban, M.T.; Henry, T.D.; Dietz, A.B.; Lerman, L.O.; Lerman, A. Promise of autologous CD34+ stem/progenitor cell therapy for treatment of cardiovascular disease. Cardiovasc. Res. 2020, 116, 1424–1433. [Google Scholar] [CrossRef]
  151. Medina, R.J.; Barber, C.L.; Sabatier, F.; Dignat-George, F.; Melero-Martin, J.M.; Khosrotehrani, K.; Ohneda, O.; Randi, A.M.; Chan, J.K.; Yamaguchi, T.; et al. Endothelial progenitors: A consensus statement on nomenclature: Endothelial progenitors nomenclature. Stem Cells Transl. Med. 2017, 6, 1316–1320. [Google Scholar] [CrossRef]
  152. Basile, D.P.; Yoder, M.C. Chapter 9—Regeneration and replacement of endothelial cells and renal vascular repair. In Regenerative Nephrology, 2nd ed.; Goligorsky, M.S., Ed.; Academic Press: Cambridge, MA, USA, 2022; pp. 129–144. ISBN 9780128233184. [Google Scholar] [CrossRef]
  153. Shen, J.; Xie, Y.; Liu, Z.; Zhang, S.; Wang, Y.; Jia, L.; Wang, Y.; Cai, Z.; Ma, H.; Xiang, M. Increased myocardial stiffness activates cardiac microvascular endothelial cell via VEGF paracrine signaling in cardiac hypertrophy. J. Mol. Cell. Cardiol. 2018, 122, 140–151. [Google Scholar] [CrossRef]
  154. Eibel, B.; Rodrigues, C.G.; Giusti, I.I.; Nesralla, I.A.; Prates, P.R.; Sant’Anna, R.T.; Nardi, N.B.; Kalil, R.A. Gene therapy for ischemic heart disease: Review of clinical trials. Rev. Bras. Cir. Cardiovasc. 2011, 26, 635–646. [Google Scholar] [CrossRef] [Green Version]
  155. Hollon, T. Researchers and regulators reflect on first gene therapy death. Nat. Med. 2000, 6, 6. [Google Scholar] [CrossRef]
  156. Annex, B.H.; Cooke, J.P. New Directions in Therapeutic Angiogenesis and Arteriogenesis in Peripheral Arterial Disease. Circ Res. 2021, 128, 1944–1957. [Google Scholar] [CrossRef]
  157. Iyer, S.R.; Annex, B.H. Therapeutic angiogenesis for peripheral artery disease: Lessons learned in translational science. JACC Basic Transl. Sci. 2017, 2, 503–512. [Google Scholar] [CrossRef] [PubMed]
  158. Cooke, J.P.; Meng, S. Vascular regeneration in peripheral artery disease. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1627–1634. [Google Scholar] [CrossRef] [PubMed]
  159. Staudacher, D.L.; Flugelman, M.Y. Cell and gene therapies in cardiovascular disease with special focus on the no Option patient. Curr. Gene Ther. 2006, 6, 609–623. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Small caliper blood vessels immunostained for CD34 antibody (DAB ×200).
Figure 1. Small caliper blood vessels immunostained for CD34 antibody (DAB ×200).
Medicina 58 00903 g001
Figure 2. The balance of activators and inhibitors in: (A)—insufficient angiogenesis; (B)—normal angiogenesis; (C)—excessive angiogenesis. Abbreviations: FGF: fibroblast growth factor, VEGF: vascular endothelial growth factor, PD-ECGF: platelet-derived endothelial cell growth factor, TSP-1: thrombospondin-1.
Figure 2. The balance of activators and inhibitors in: (A)—insufficient angiogenesis; (B)—normal angiogenesis; (C)—excessive angiogenesis. Abbreviations: FGF: fibroblast growth factor, VEGF: vascular endothelial growth factor, PD-ECGF: platelet-derived endothelial cell growth factor, TSP-1: thrombospondin-1.
Medicina 58 00903 g002
Table 1. Endogenous angiogenesis activators.
Table 1. Endogenous angiogenesis activators.
ActivatorDescription/StructureReceptor(s)/Cellular TargetsMechanism
of Action
References
FGF family
-
group of over 20 members;
-
the most studied members are: aFGF (FGF1) and bFGF (FGF2);
-
small polypeptides of 155–268 amino acids.
-
group of receptors, four of them (FGFR1 to FGFR4) being the most studied;
-
belong to the tyrosine kinase superfamily and are known to dimerize;
-
FGFRs are composed of an extracellular domain that binds the ligand, having three immunoglobulin-like domains (D1-D3), a single transmembrane helix domain and an intracellular tyrosine-kinase domain.
-
FGFs binding to FGFRs is influenced by FGFs interaction with HSPG associated to cell surface.
-
FGFs are strong mitogens of different cell types
[4,49,50,51,52,53]
VEGF family
-
group of six members: VEGF-A (commonly termed as VEGF); VEGF-B; VEGF-C; VEGF-D; VEGF-E; PlGF.
-
VEGF-A isoforms:
-
VEGF121, VEGF145, VEGF162, VEGF165, VEGF165B, VEGF183, VEGF189 and VEGF206.
-
group of 3 receptors (VEGFR1 to VEGFR3)
-
like FGFRs, they belong to the tyrosine kinase superfamily and are known to dimerize;
-
VEGFRs are composed of an extracellular domain having seven immunoglobulin-like domains, a transmembrane domain and an intracellular tyrosine-kinase domain.
-
mitogenic action restricted to endothelial cells;
-
VEGF-A is a vasodilator and was initially referred to as VPF because of its capacity of increasing microvascular permeability.
[3,4,54,55,56]
Ang 1
-
belongs to the angiopoietins’ group, along to Ang2, plus other several angiopoietin-related factors;
-
Ang 2 is an antagonist of Ang1;
-
Ang 1 is a protein formed of 498 amino-acid residues (57 kDa);
-
Ang1 binds to TIE2 receptor;
-
Ang1 multimerizes (the tetramer level of four or higher must be archived) prior to receptor binding;
-
TIE2 heterodimerizes with TIE1 in order to exhibit its biological activity
-
mediates endothelial cells’ migration, adhesion and survival and plays an essential role in vessel maturation
[4,57,58,59,60]
Ephrins
-
group of 8 proteins linked to cell membrane;
-
ephrins are divided into two subgroups:
-
group A (5 ephrin ligands: ephrin A1–A5) and group B (3 ephrin ligands: ephrin B1–B3)
-
group of receptors (Ephs) divided into two subclasses, depending mainly on the type of ligand: EphsA and EphsB;
-
Ephs are the largest subfamily of RTKs and dimerize upon ligand binding.
-
Eph recognition by Eph receptors mediates cell adhesion to extracellular matrix, juxta-crine cell–cell contacts, and cell migration.
[4,61,62,63]
MMPs
-
zinc-dependent endopeptidases (protein structure) also known as matrixins;
-
MMPs can be divided into secreted MMPs (gelatinases, strome-lysins, collagenases, other secreted MMPs) and membrane-bound MMPs.
-
Secreted Mms interact with membrane–bound MMps, activated, at their turn, intracellularly.
-
PAR1 (a G protein-coupled receptor involved in various types of cancer) is known as receptor of MMP-1.
-
MMPs are involved in tissue remodeling, by degradation of the extracellular matrix.
[4,64,65,66]
Abbreviations: FGF—fibroblast growth factor; aFGF—acidic FGF; bFGF—basic FGF; FGFR—FGF receptor; HSPG—heparan sulfate proteoglycans; VEGF—vascular endothelial growth factor; VEGFR—vascular endothelial growth factor receptor; PlGF—placental growth factor; Ang 1—Angiopoietin 1; Ang 2—Angiopoietin 2; TIE2—tyrosine kinase with immunoglobulin and epidermal growth factor homology domain 2; Ephs—ephrin receptors; RTKs—receptor protein tyrosine-kinase; MMPs—Matrix metalloproteinases; PAR—Protease-activated receptor.
Table 2. Endogenous Angiogenesis inhibitors.
Table 2. Endogenous Angiogenesis inhibitors.
InhibitorDescription/StructureReceptor(s)/Cellular TargetsMechanism of ActionReferences
Endostatin
-
Fragment of collagen XVIII;
-
It can exist as a monomer or as a trimer, in soluble or insoluble form;
-
Endostatin binds with low affinity to all surface heparan sulfate proteoglycans involved in growth factor signaling and it also binds to heparin;
-
it inhibits certain MMPs;
-
it reduces the invasion of tumor cells and of ECs, with no effect on proliferation.
[39,67,68,69]
Tumstatin
-
Fragment of type IV collagen (28kDa);
-
Tumstatin binds to integrins (ex. αvβ3)
-
it inhibits the proliferation of ECs and promotes apoptosis with no effect on migration of ECs.
[39,70]
Angiostatin
-
Plasminogen fragment (it is known that plasminogen itself doesn’t possess antiangiogenic properties);
-
Angiostatin and plasminogen bind to αvβ3 integrin;
-
it binds to ATP synthase at the surface of endothelial cells;
-
Angiomotin is a protein known to bind and to internalize angiostatin.
-
Angiostatin inhibits ECs’ proliferation and migration.
[39,71,72,73]
TSPs
-
TSPs are divided into two subgroups: subgroup A (including TSP-1, the most studied member, and TSP-2), and subgroup B (including TSP-3, TSP-4 and COMP);
-
TSP-1 is a multifunctional glycoprotein stored in the extracellular matrix;
-
TSP-1 is a homotrimer of three glycopeptides linked through disulfide bonds. The amino-terminal domain of TPS-1 has heparin-binding activity; the carboxyl terminal domain has cell-binding and calcium ion binding activity and between them there are domains with procollagen, properdin and EGF amino-acid homology.
-
TPS-1 is recognized by at least 12 adhesion receptor (CD36, αv integrins, β1 integrins, etc.) and proteases involved in angiogenesis (plasminogen, urokinase, matrix metalloproteinase, etc.)
-
TPS-1 induces ECs’ apoptosis;
[39,45,46,74,75,76,77]
2-ME
-
estradiol metabolite with no estrogenic activity
-
2-ME binds poorly to estrogen receptors
-
inhibits ECs’ proliferation and induces apoptosis in ECs.
[39,78,79]
Abbreviations: MMPs—Matrix metalloproteinases; EC—endothelial cell; ATP—adenosine triphosphate; TSPs—Thrombospondins; EGF—Epidermal growth factor; COMP—cartilage oligomeric matrix protein; 2-ME—2-Methoxyestradiol.
Table 3. Angiogenesis inhibitors approved to treat cancer in humans.
Table 3. Angiogenesis inhibitors approved to treat cancer in humans.
Angiogenesis Inhibitor
Generic Name
(Trade Name)
Description/Chemical TaxonomyMechanism of Action
[102]
Approved to Treat
(Alone or with Other Drugs)
[101]
Axitinib (Inlyta®)
https://go.drugbank.com/drugs/DB06626,
accessed on 4 July 2022
Medicina 58 00903 i001Axitinib selectively blocks the tyrosine kinase receptors VEGFR-1, VEGFR-2, and VEGFR-3.Renal cell carcinoma [103]
Bevacizumab (Avastin®, Mvasi®, Zirabev®)
https://go.drugbank.com/drugs/DB00112
accessed on 4 July 2022
Recombinant humanized monoclonal antibodyVEGF-A inhibitorCervical and colorectal cancer, glioblastoma, hepatocellular carcinoma, Non-squamous non-small cell lung cancer, Ovarian epithelial, fallopian tube or primary peritoneal cancer, Renal cell carcinoma [104]
Cabozantinib (Cometriq®)
https://go.drugbank.com/drugs/DB08875
accessed on 4 July 2022
Medicina 58 00903 i002Non-specific receptor tyrosine kinase inhibitorHepatocellular carcinoma, Medullary thyroid cancer, Renal cell carcinoma [105]
Everolimus (Afinitor®)
https://go.drugbank.com/drugs/DB01590
accessed on 4 July 2022
Medicina 58 00903 i003Everolimus works similarly to Rapamycin, being a derivative of Rapamycin (sirolimus). After binding to FKBP-12, Everolimus inhibits the activation of mTOR, a key regulatory kinase. Breast, pancreatic, gastrointestinal and lung cancer, renal cell carcinoma, subependymal giant cell astrocytoma [106]
Lenalidomide (Revlimid®)
https://go.drugbank.com/drugs/DB00480
accessed on 4 July 2022
Medicina 58 00903 i004Being an analogue of thalidomide, Lenalidomide works through various mechanisms of action, promoting malignant cell death and enhancing host immunity.Anemia, Follicular lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma [107]
Lenvatinib mesylate (Lenvima®)
https://go.drugbank.com/drugs/DB09078
accessed on 4 July 2022
Medicina 58 00903 i005Receptor tyrosine kinase inhibitorEndometrial carcinoma, hepatocellular carcinoma, renal cell carcinoma, thyroid cancer [108]
Pazopanib (Votrient®)
https://go.drugbank.com/drugs/DB06589
accessed on 4 July 2022
Medicina 58 00903 i006Second-generation multitargeted tyrosine kinase inhibitorRenal cell carcinoma; Soft tissue sarcoma [109]
Ramucirumab (Cyramza®)
https://go.drugbank.com/drugs/DB05578
accessed on 4 July 2022
Human monoclonal antibody (IgG1) against vascular endothelial growth factor receptor 2 (VEGFR2)Ramucirumab is a direct VEGFR-2 antagonist, that blocks the binding of natural VEGF ligands.Colorectal cancer, Hepatocellular carcinoma, Non-small cell lung cancer, Stomach adenocarcinoma or gastroesophageal junction adenocarcinoma [110]
Regorafenib (Stivarga®)
https://go.drugbank.com/drugs/DB08896
accessed on 4 July 2022
Medicina 58 00903 i007Multiple kinases inhibitorColo-rectal cancer, Gastrointestinal stromal tumor, Hepato-cellular carcinoma [111]
Sorafenib (Nexavar®)
https://go.drugbank.com/drugs/DB00398
accessed on 4 July 2022
Medicina 58 00903 i008Kinase inhibitorHepatocellular carcinoma, Renal cell carcinoma, Thyroid cancer [112]
Sunitinib (Sutent®)
https://go.drugbank.com/drugs/DB01268
accessed on 4 July 2022
Medicina 58 00903 i009Receptor tyrosine kinase inhibitorGastrointestinal stromal tumor; Pancreatic cancer; Renal cell carcinoma [113]
Thalidomide (Synovir, Thalomid®)
https://go.drugbank.com/drugs/DB01041
accessed on 4 July 2022
Medicina 58 00903 i010As a cancer treatment, thalidomide may act as a VEGF inhibitor.Multiple myeloma [114]
Vandetanib (Caprelsa®)
https://go.drugbank.com/drugs/DB05294
accessed on 4 July 2022
Medicina 58 00903 i011Tyrosine kinases inhibitorMedullary thyroid cancer [115]
Ziv-aflibercept (Zaltrap®)
https://go.drugbank.com/drugs/DB08885
accessed on 4 July 2022
Recombinant protein composed of the binding domains of two human VEGFRs fused with the Fc region of human IgG1.VEGF inhibitorMetastasized colorectal cancer [116]
Abbreviations: VEGF: vascular endothelial growth factor; VEGFR: Vascular endothelial growth factor receptor; FKBP-12: FK Binding Protein-12; mTOR: mammalian target of Rapamycin; IgG1: immunoglobulin gamma 1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ionescu, C.; Oprea, B.; Ciobanu, G.; Georgescu, M.; Bică, R.; Mateescu, G.-O.; Huseynova, F.; Barragan-Montero, V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina 2022, 58, 903. https://doi.org/10.3390/medicina58070903

AMA Style

Ionescu C, Oprea B, Ciobanu G, Georgescu M, Bică R, Mateescu G-O, Huseynova F, Barragan-Montero V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina. 2022; 58(7):903. https://doi.org/10.3390/medicina58070903

Chicago/Turabian Style

Ionescu, Cătălina, Bogdan Oprea, Georgeta Ciobanu, Milena Georgescu, Ramona Bică, Garofiţa-Olivia Mateescu, Fidan Huseynova, and Veronique Barragan-Montero. 2022. "The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview" Medicina 58, no. 7: 903. https://doi.org/10.3390/medicina58070903

Article Metrics

Back to TopTop