J Lipid Atheroscler. 2020 May;9(2):268-282. English.
Published online May 18, 2020.
Copyright © 2020 The Korean Society of Lipid and Atherosclerosis.
Review

Toward the Clinical Application of Therapeutic Angiogenesis Against Pediatric Ischemic Retinopathy

Dong Hyun Jo,1 and Jeong Hun Kim2,3,4
    • 1Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea.
    • 2Fight against Angiogenesis-Related Blindness, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea.
    • 3Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
    • 4Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea.
Received March 29, 2020; Revised April 29, 2020; Accepted May 13, 2020.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Therapeutic angiogenesis refers to strategies of inducing angiogenesis to treat diseases involving ischemic conditions. Historically, most attempts and achievements have been related to coronary and peripheral artery diseases. In this review, we propose the clinical application of therapeutic angiogenesis for the treatment of pediatric ischemic retinopathy, including retinopathy of prematurity, familial exudative retinopathy, and NDP-related retinopathy. These diseases are all characterized by the reduction of physiological angiogenesis and the following induction of pathological angiogenesis. Therapeutic angiogenesis, which supplements insufficient physiological angiogenesis, may be a therapeutic approach for ischemic conditions. Various molecules and modalities can be utilized to apply therapeutic angiogenesis for the treatment of ischemic retinopathy, as in coronary and peripheral artery diseases. Experiences with cardiovascular diseases provide a useful reference for the further clinical application of therapeutic angiogenesis in pediatric ischemic retinopathy. Recombinant proteins and gene therapy are powerful tools to deliver angiogenic factors to retinal tissues directly. Furthermore, endothelial progenitor or bone marrow-derived cells can be injected into the vitreous cavity of the eye for therapeutic angiogenesis. Intraocular injections are highly promising for the delivery of therapeutics for therapeutic angiogenesis. We expect that therapeutic angiogenesis will be a breakthrough in the treatment of pediatric ischemic retinopathy.

Keywords
Retinal diseases; Ischemia; Physiologic neovascularization; Angiogenesis modulating agents; Therapeutics

INTRODUCTION

Therapeutic angiogenesis was proposed as a therapeutic approach targeting clinical problems due to the local rarefaction of blood vessels, insufficient neovascularization, or both.1, 2 After the first suggestion in 1993, ongoing active attempts have been made to apply therapeutic angiogenesis to the treatment of cardiovascular diseases.3, 4, 5, 6 Therapeutic angiogenesis employs various angiogenic factors, including vascular endothelial growth factors (VEGFs), fibroblast growth factors (FGFs), and hepatocyte growth factor (HGF), to induce neovascularization in ischemic tissues. Although preclinical studies of protein, gene, and cell therapies were promising, therapeutic angiogenesis is still not a mainstay treatment option for coronary and peripheral artery diseases after the modest success of clinical trials. In this review, we instead suggest that pediatric ischemic retinopathy might be a promising target of therapeutic angiogenesis. Based on the lessons from clinical studies on cardiovascular diseases and recent preclinical studies on ischemic retinopathy, the clinical application of therapeutic angiogenesis to treat patients with pediatric ischemic retinopathy should be investigated through a mechanism-based approach. The eye is an easily accessible organ for the local delivery of therapeutic materials in protein, gene, and cell therapies. This characteristic may help apply modalities of therapeutic angiogenesis in a more feasible way for the treatment of ischemic retinopathy.

PEDIATRIC ISCHEMIC RETINOPATHY

Pediatric ischemic retinopathy refers to a spectrum of retinal diseases characterized by hypovascularization-related ischemia of the retinal tissue (Fig. 1). In patients with retinopathy of prematurity (ROP), familial exudative vitreoretinopathy (FEVR), and NDP-related retinopathy, there are areas without retinal vessels (known as avascular retina) in the peripheral retina due to the insufficient development of retinal vasculature. The presence of insufficient retinal vessels results in an ischemic retinal microenvironment.

Fig. 1
Representative clinical photographs of patients with pediatric ischemic retinopathy. (A) A fundus photograph of a patient with retinopathy of prematurity. (B) A fluorescein angiography photograph of a patient with familial exudative vitreoretinopathy. (C) A fluorescein angiography photograph of a patient with NDP-related retinopathy. The margins between vascularized and avascular retinas are indicated with white arrows.

ROP

ROP is a vision-threatening retinopathy involving abnormal retinal vascular development in premature infants, as its name implies (Fig. 1A).7 The incidence and severity are related to low birth weight and gestational age.8 Because the retinal vasculature is not complete in premature infants, the peripheral retina is without retinal vessels. Hypoxia in the peripheral retina activates hypoxia-mediated signaling pathways, increasing the secretion of angiogenic factors.7 This, in turn, induces a fibrovascular reaction in the retinal tissues. The stages of ROP reflect the degree of fibrovascular proliferation along the borders of the avascular and vascularized retina. The severity increases from the demarcation line (stage 1) to ridge with volume (stage 2), extraretinal fibrovascular proliferation (stage 3), and tractional retinal detachment (stage 4, partial; stage 5, total).7

Animal models that mimic the pathogenesis and consequences of ROP have been developed.2, 9, 10 These models are valuable tools to investigate the potential of therapeutic approaches for ROP. The most widely utilized model is an oxygen-induced retinopathy (OIR) model in mice.9,10 In this model, neonatal pups are exposed to 75% oxygen for 5 days from postnatal day 7 (P7) to P12. This procedure leads to the regression of central vessels. Then, the mice in normal room air (20% oxygen) experience relative hypoxia (due to the rarefaction of the retinal vessels) in the central retina, which peaks at P14. As in patients with ROP, neovascular tufts develop along the area between the avascular and vascularized retina. Usually, pathological neovascularization peaks at P17. If therapeutic approaches are developed to recover from rarefaction of the retinal vasculature, it will be possible to prevent the following pathological neovascularization.

FEVR

FEVR is another vision-threatening retinopathy involving retinal hypovascularization in infants and children (Fig. 1B).11 The principal feature of the disease is an avascular peripheral retina, as in ROP.12 This leads to pathological retinal neovascularization in the peripheral retina, with or without exudates and further retinal detachment.13 It is noteworthy that patients with FEVR possess variants in genes encoding molecules of the norrin/frizzled class receptor-4 pathway, one of the Wnt/β-catenin signaling pathways, including NDP, FZD4, LRP5, and TSPAN12.14 Norrin (encoded by the NDP gene) is a Wnt ligand, frizzled class receptor-4 (coded by the FZD4 gene) is a receptor, and low-density lipoprotein receptor-related protein 5 (from the LRP5 gene) and transpanin 12 (from the TSPAN12 gene) are coreceptors with frizzled class receptor-4.

As in patients with FEVR, Ndp or Fzd4 knockout mice demonstrated insufficient retinal vasculature compared to wild-type mice.15, 16 Angiogenesis takes place through multiple steps: 1) the activation of endothelial cells by angiogenic factors, 2) the invasion and protrusion of new sprouts, 3) the proliferation of endothelial cells to support sprout elongation, 4) lumen formation to build vessel loops, and 5) the initiation of blood flow, the establishment of a basement membrane, and the recruitment of mural cells to stabilized new vessels.17 Primary retinal endothelial cells from Fzd4 knockout mice demonstrated a lower potential to migrate and form tubules in in vitro angiogenesis assays.15 In addition, Ndp knockout mice showed less proliferation of endothelial cells in the retinal vasculature in vivo. Because insufficient frizzled signaling leads to retinal hypovascularization in mutant mice and possibly in patients with FEVR, therapeutic strategies to restore aberrant frizzled signaling might be of therapeutic potential.

1. NDP-related retinopathies

Mutations in the NDP gene have been reported in patients with a spectrum of retinal diseases, including FEVR and ROP.18, 19, 20, 21, 22, 23 In this context, NDP-related retinopathies have been proposed to include X-linked FEVR, ROP, persistent hyperplastic primary vitreous, Norrie disease, and Coats disease.24 Although the manifestations of NDP-related retinopathies vary, incomplete retinal vascular vasculature is a common feature of these conditions (Fig. 1C). Other phenotypes include fibrous stalk in persistent hyperplastic primary vitreous, exudates in Coats disease, and fibrovascular membranes in FEVR, ROP, and Norrie disease, which are provoked and exacerbated by ischemia due to incomplete retinal vasculature. NDP-related retinopathies are mainly induced by mutations in the NDP gene and resultant insufficiency of the norrin protein. In this context, NDP gene therapy and norrin protein supplementation may be useful therapeutic approaches.

2. Common pathological mechanisms and current treatment options of pediatric ischemic retinopathy

It is noteworthy that insufficient physiological angiogenesis is a crucial feature of ischemic retinopathy. This leads to hypoxia-mediated pathological neovascularization, which results in bleeding, exudate formation, and fibrovascular proliferation.25, 26 Nevertheless, current treatment options for ischemic retinopathy only target pathological neovascularization or its complications, such as vitreous hemorrhage and retinal detachment. In patients with pediatric ischemic retinopathy, laser photocoagulation destroys the retinal tissues in the peripheral vascular retina, suppressing the metabolic demand and the secretion of angiogenic and inflammatory factors.7, 12 In addition, to minimize VEGF-mediated pathological neovascularization, anti-VEGF antibody (bevacizumab; Genentech, South San Francisco, CA, USA) is administered to patients with ischemic retinopathies such as ROP, FEVR, and diabetic retinopathy.27, 28, 29 Vitreous hemorrhage and retinal detachment are managed by surgery, such as vitrectomy and encircling. Unfortunately, there are no clinically proven approaches directly targeting retinal hypovascularization for the treatment of ischemic retinopathy.

THERAPEUTIC ANGIOGENESIS

Therapeutic angiogenesis is a direct therapeutic approach to supplement physiological angiogenesis in ischemic areas for the treatment of diseases involving ischemic conditions. Historically, attempts have been made to apply therapeutic angiogenesis for the treatment of cardiovascular diseases characterized by hypovascularization. These experiences might help to develop therapeutic approaches based on therapeutic angiogenesis for the treatment of pediatric ischemic retinopathy. Coronary artery disease occurs when atheromatous processes prevent blood flow through the coronary artery.30 In contrast, peripheral artery disease is caused by atherosclerotic occlusion of the arteries to the legs.31 The ability of various angiogenic factors to induce therapeutic angiogenesis has been tested in preclinical studies and patients with coronary and peripheral artery diseases. The list of these factors includes angiogenin, angiopoietin, FGF, granulocyte colony-stimulating factor, HGF, insulin-like growth factor-1, nitric oxide, platelet-derived growth factor, transforming growth factor, tumor necrosis factor alpha, and VEGF.1, 32, 33, 34 Among them, VEGF, FGF, and HGF have been the most widely studied in clinical trials.

1. Coronary artery disease

Selected clinical studies of therapeutic angiogenesis on coronary artery disease are summarized in Table 1, which includes the first trials of different therapeutic materials, phases, and administration methods. These studies exemplify the primary concerns regarding the clinical application of therapeutic angiogenesis for the treatment of human diseases.

Table 1
Selected clinical studies on therapeutic angiogenesis for coronary artery diseases

Protein therapy

The first clinical trial using a recombinant protein for therapeutic angiogenesis in patients with coronary artery disease tested the therapeutic potential of FGF-1 injected into the myocardium.35 During an elective bypass operation for multivessel coronary artery disease, FGF-1 was applied to the myocardium. In this study, at 12 weeks after the injection, intra-arterial digital subtraction angiography demonstrated that a dense capillary network appeared around the injection site.35 Laham et al.36 showed that there was a trend toward a reduction in the target ischemic area on magnetic resonance assessment in patients who received an epicardial injection of FGF-2 while undergoing coronary bypass surgery. The first clinical trial using intracoronary recombinant VEGF reported that there was an improvement in myocardial perfusion on single-photon emission computed tomography (SPECT) in patients with severe coronary artery disease who were not optimal candidates for angioplasty or bypass surgery.37 The results of these phase 1 trials were promising. However, the following phase 2 trials failed to provide clear-cut evidence of therapeutic effectiveness for more extensive clinical utilization of therapeutic angiogenesis to treat coronary artery disease.3, 38, 39, 40 A single intracoronary infusion of FGF-2 did not improve exercise tolerance or myocardial perfusion in patients with coronary artery disease who were considered suboptimal candidates for standard surgical or catheter-based revascularization in the FIRST trial.38 In addition, an intracoronary infusion of VEGF did not offer any improvement in exercise treadmill test time by day 60 in patients with stable angina who were judged unsuitable for revascularization based on coronary angiography.39

Gene therapy

In the treatment of coronary artery disease, naked plasmid DNA encoding the VEGF165 gene was injected directly into the ischemic myocardium, improving myocardial perfusion on SPECT imaging.41 Additionally, intramyocardial delivery of an adenoviral vector encoding the VEGF121 gene improved symptoms, treadmill exercise assessment, and angiographic assessment in patients with reversible left ventricular ischemia by dobutamine stress echocardiography.42 Similarly, intramyocardially administered naked plasmid DNA encoding the VEGF-2 gene also reduced ischemia on electromechanical mapping and improved myocardial perfusion on SPECT scanning.43 A phase 1/2 trial showed that the myocardial transfer of plasmid DNA encoding the VEGF-2 gene through catheter delivery significantly reduced the anginal class in patients with Canadian Cardiovascular Society (CCS) class III or IV angina refractory to maximum medical therapy, multivessel coronary artery disease not suitable for bypass surgery or angioplasty, and reversible ischemia on stress SPECT imaging.44 However, another phase 1/2 study (the AGENT trial) on the intracoronary administration of an adenoviral vector encoding the FGF-4 gene only demonstrated insignificant trends of improvement in exercise time in patients with CCS class 2 or 3 angina.45

Progenitor cells

Progenitor cells can home to local injured and ischemic tissues and participate in damage repairing and wound healing by secreting growth factors and stimulating neovascularization.46, 47 In this context, bone marrow-derived or circulating blood-derived cells were administered to patients with coronary artery disease in several clinical trials. In the TOPCARE-AMI trial, an intracoronary infusion of bone marrow or circulating blood-derived progenitor cells was associated with functional recovery at a 4-month follow-up in patients with acute myocardial infarction.48 Strauer et al.49 attributed the therapeutic effects of intracoronary transplantation of bone marrow-derived mononuclear cells to myocardial regeneration and neovascularization in patients with acute myocardial infarction after mechanical angioplasty and subsequent stent implantation. Transendocardial and intramyocardial implantation of bone marrow-derived mononuclear cells also demonstrated beneficial effects on myocardial blood flow and ventricular function.50, 51

2. Peripheral artery disease

Selected clinical studies of therapeutic angiogenesis for peripheral artery disease are summarized in Table 2, which includes the first trials of various therapeutic materials, phases, and administration methods.

Table 2
Selected clinical studies of therapeutic angiogenesis for peripheral artery diseases

Protein therapy

In patients with peripheral artery disease and intermittent claudication, FGF-2 was infused into the femoral artery of the ischemic leg in a phase 1 trial by Lazarous et al.52 Intraarterial FGF-2 was well-tolerated and increased the blood flow of the calf.52 In a phase 2 clinical trial of 190 patients with intermittent claudication, intraarterial FGF-2 resulted in a significant increase in peak walking time at 90 days.53

Gene therapy

The potential of gene therapy in the treatment of peripheral artery disease has been more extensively investigated in several clinical trials. The first clinical trial using a plasmid encoding the VEGF165 gene was done in a female patient with 40% stenosis of the proximal popliteal artery on arteriography.54 Arterial gene transfer with a hydrogel-coated balloon-angioplasty catheter resulted in an increase in collateral vessels at the knee, mid-tibial, and ankle levels at 4 weeks after treatment.54 Intramuscular administration of naked plasmid DNA encoding the VEGF165 gene transiently increased serum levels of VEGF, induced newly visible collateral blood vessels on contrast angiography, and improved distal flow on magnetic resonance angiography in patients with nonhealing ischemic ulcers and/or rest pain due to peripheral artery disease.55 In a phase 1/2 trial using a plasmid encoding the HGF gene, intramuscular injection induced a dose-dependent increase in transcutaneous oxygen tension in patients with critical limb ischemia.56 In a randomized, placebo-controlled, double-blinded phase 2 study, intraarterial gene transfer via an adenoviral vector or a plasmid encoding the VEGF165 gene increased vascularity distal to the gene transfer site on digital subtraction angiography in patients with claudication or critical lower-limb ischemia.57 Unfortunately, intramuscular delivery of an adenoviral vector encoding the VEGF121 gene did not increase the peak walking time, ankle-brachial index, or quality-of-time measures in another phase 2 randomized, double-blind clinical trial (the RAVE trial) in patients with disabling intermittent claudication.58

Cell therapy

As preclinical studies have demonstrated that bone marrow-derived mononuclear cells increased collateral vessel formation in ischemic limbs, intramuscular injection of these cells into the gastrocnemius of patients with unilateral ischemia of the leg improved the ankle-brachial index, transcutaneous oxygen pressure, rest pain, and pain-free walking time.59

3. Issues in the application of therapeutic angiogenesis for cardiovascular diseases

Several concerns have blocked the widespread clinical application of therapeutic angiogenesis for cardiovascular diseases. The first concern is whether new capillaries (formed through angiogenesis) without the simultaneous formation of larger arteries (through arteriogenesis) for supplying the capillaries are of less importance and patency.60, 61 Second, there was no clear consensus on the concentration, the timing, and the area of locally administered angiogenic factors.3, 33, 60 Prolonged tissue exposure to growth factors might be required for the development of robust and sustained neovascularization to secure the survival of the newly formed vasculature.62, 63 Third, another major concern is excessive vessel growth in the target tissue in the form of hemangioma or a glomeruloid body.64 Fourth, concerns have been raised regarding the development of abnormal vessels in other organs after systemic administration of angiogenic factors.60, 65 In addition, a combination of 2 or more angiogenic factors might be required for functionally mature neovascularization and consistent clinical benefits.66, 67, 68 In this context, using different factors acting through complementary mechanisms has been proposed as a solution to the clinical failure of single agents in cardiovascular diseases.69 For therapeutic angiogenesis to be applied to the treatment of human diseases, including ischemic retinopathy, these issues should be appropriately addressed.

POTENTIAL OPTIONS OF THERAPEUTIC ANGIOGENESIS AGAINST ISCHEMIC RETINOPATHY

The eye is easily accessible for the local delivery of therapeutic materials using various administration routes, including intravitreal and suprachoroidal injections, which are currently utilized for the treatment of retinal diseases (Fig. 2). Antibodies (bevacizumab), antibody fragments (ranibizumab; Genentech), and an antibody-mimicking fusion protein (aflibercept; Regeneron, Tarrytown, NY, USA) are widely administered via intravitreal injections to treat age-related macular degeneration and diabetic retinopathy. In addition, retinal pigment epithelial cells from induced pluripotent stem cells and adeno-associated viral vectors containing therapeutic genes are injected into the subretinal space of patients with age-related macular degeneration and retinal dystrophies, respectively.70, 71 These administration routes can also be used for therapeutic approaches inducing therapeutic angiogenesis for ischemic retinopathy. Potential options and examples of therapeutic angiogenesis against ischemic retinopathy are summarized in Table 3.

Fig. 2
Various administration routes for delivery of therapeutic materials to the eye. The subretinal and intravitreal routes are currently utilized for the treatment of retinal diseases.

Table 3
Potential options and examples of therapeutic angiogenesis against ischemic retinopathy

For protein therapy, recombinant proteins can be injected into the vitreous cavity. However, VEGF and FGF are unlikely to be suitable for the treatment of ischemic retinopathy via intravitreal injections, because VEGF and FGF levels are usually elevated in the vitreous in patients with ischemic retinopathy.72, 73, 74, 75 Instead, a still-unidentified ‘X’ protein might be associated with an increase in therapeutic angiogenesis, as opposed to pathological angiogenesis.

One of the candidates for this ‘X’ protein is norrin, which is encoded by the NDP gene. As mentioned, Ndp-deficient mice demonstrate a distinct failure in the development of retinal vasculature.15, 76 It is remarkable that the transgenic expression of norrin by a lens-specific promoter restores the formation of a normal retinal vasculature in Ndp-deficient mice.76 This implies that intravitreal injection of norrin or ectopic expression of the NDP gene might restore physiological angiogenesis in ischemic retinopathy. Transgenic expression of norrin in the lens or retinal pigment epithelium successfully suppressed the pathological phenotypes of OIR in mice, the most widely utilized animal model of ischemic retinopathy.77 In that study, there was no increase in pathological neovascularization. Norrin also decreased the avascular area and inhibited the formation of neovascular tuft in a murine model of OIR.78 This effect might be linked with the induction of insulin-like growth factor-1.79 Direct injection of the norrin protein or gene delivery through adeno-associated viral vectors of the NDP gene might have potential for the treatment of ischemic retinopathy. In another study using COMP-Ang1, a soluble and stable variant of angiopoietin-1,80 intravitreal injection of COMP-Ang1 promoted the formation of a vascular network in the central avascular area in OIR mice.81

Studies on animal models of ischemia provide evidence that endothelial progenitor cells (EPCs) migrate to the ischemic tissue.46, 47 Circulating progenitor cells expressing the surface marker CD34, which are capable of differentiating into endothelial cells, are recruited to ischemic sites and committed to forming capillaries by hypoxia-regulated factors, such as stromal-derived factor-1, insulin-like growth factor binding protein-3, and VEGF.82, 83 This tendency can also be utilized in the treatment of ischemic retinopathy. Intravitreally administered CD34+ EPCs incorporate into the damaged retinal vasculature in mice with OIR, after ischemia/reperfusion injury, or streptozotocin-induced diabetic retinopathy, as well as in BBZDR/WOR rats in a rat model of diabetic retinopathy.84 Several groups have also reported the therapeutic potential of EPCs, although the protocols and the levels of commitment vary.85, 86, 87, 88, 89 Medina et al. reported that outgrowth endothelial cells, also called endothelial colony-forming cells,90 with higher expression of CD105 or CD146 from peripheral blood mononuclear cells contributed to vascular repair and reduced the stimuli for pathological angiogenesis.85 Prasain et al.86 demonstrated that outgrowth endothelial cells from human induced pluripotent stem cells reduced the avascular area and preretinal neovascular tufts. In another study, cord blood-derived EPCs were effective in restoring pathological changes in mice with OIR.87 A combination of bone marrow-derived CD34+ cells and vascular wall-derived endothelial colony-forming cells88 or co-administration of a peptide based on the helix-B domain of erythropoietin91 was suggested to enhance the therapeutic effects of EPCs in OIR mice.

CONCLUSION

It is essential to restore the processes of physiological angiogenesis for the direct treatment of patients with ischemic retinopathy. Current treatment options, including laser photocoagulation and surgery, only target the resultant pathological angiogenesis and complications. In the clinical application of therapeutic angiogenesis for human diseases, the most important aspect is the tight regulation of angiogenic processes, from the initiation of angiogenesis to the remodeling of newly formed vessels. The angiogenic process must be controlled in order to obtain a functional vascular network.92 Avascular retinal tissues should be exposed to angiogenic factors or progenitor cells for a prolonged time to promote the sustained development of vessels. Preclinical studies using presumptive therapeutic approaches have shown potential, but more studies should be performed for clinical applications to be viable. However, we expect that the clinical application of therapeutic angiogenesis will be more promising in the context of ischemic retinopathy than for diseases in other organs because of the easy accessibility of the eye for local delivery methods. In addition, as in cardiovascular diseases, there is a tremendous unmet clinical need for the development of therapeutic approaches based on therapeutic angiogenesis in the treatment of ischemic retinopathy, as there is no effective pharmacological treatment.3, 32 We suggest that robust preclinical and clinical studies should investigate the use of therapeutic angiogenesis for the treatment of ischemic retinopathy.

Notes

Funding:This research was supported by the Bio and Medical Technology Development Program of the National Research Foundation funded by the Korean government, MSIP (NRF-2015M3A9E6028949 to JH Kim), the Creative Materials Discovery Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (2018M3D1A1058826 to JH Kim), the Development of Platform Technology for Innovative Medical Measurements funded by the Korea Research Institute of Standards and Science (KRISS – 2020 – GP2020-0004 to JH Kim), and the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (2017R1A6A3A04004741 to D.H.J.).

Conflict of Interest:The authors have no conflicts of interest to declare.

Author Contributions:

  • Conceptualization: Kim JH.

  • Funding acquisition: Kim JH.

  • Project administration: Kim JH.

  • Supervision: Kim JH.

  • Writing - original draft: Jo DH, Kim JH.

References

    1. Höckel M, Schlenger K, Doctrow S, Kissel T, Vaupel P. Therapeutic angiogenesis. Arch Surg 1993;128:423–429.
    1. Penn JS, Thum LA. Oxygen-induced retinopathy in the rat. Basic Life Sci 1988;49:1025–1028.
    1. Simons M, Ware JA. Therapeutic angiogenesis in cardiovascular disease. Nat Rev Drug Discov 2003;2:863–871.
    1. Losordo DW, Dimmeler S. Therapeutic angiogenesis and vasculogenesis for ischemic disease: part II: cell-based therapies. Circulation 2004;109:2692–2697.
    1. Losordo DW, Dimmeler S. Therapeutic angiogenesis and vasculogenesis for ischemic disease. Part I: angiogenic cytokines. Circulation 2004;109:2487–2491.
    1. Johnson T, Zhao L, Manuel G, Taylor H, Liu D. Approaches to therapeutic angiogenesis for ischemic heart disease. J Mol Med (Berl) 2019;97:141–151.
    1. Hartnett ME, Penn JS. Mechanisms and management of retinopathy of prematurity. N Engl J Med 2012;367:2515–2526.
    1. Palmer EA, Flynn JT, Hardy RJ, Phelps DL, Phillips CL, Schaffer DB, et al. Incidence and early course of retinopathy of prematurity. Ophthalmology 1991;98:1628–1640.
    1. Smith LE, Wesolowski E, McLellan A, Kostyk SK, D'Amato R, Sullivan R, et al. Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci 1994;35:101–111.
    1. Connor KM, Krah NM, Dennison RJ, Aderman CM, Chen J, Guerin KI, et al. Quantification of oxygen-induced retinopathy in the mouse: a model of vessel loss, vessel regrowth and pathological angiogenesis. Nat Protoc 2009;4:1565–1573.
    1. Gilmour DF. Familial exudative vitreoretinopathy and related retinopathies. Eye (Lond) 2015;29:1–14.
    1. Tauqeer Z, Yonekawa Y. Familial exudative vitreoretinopathy: pathophysiology, diagnosis, and management. Asia Pac J Ophthalmol (Phila) 2018;7:176–182.
    1. Pendergast SD, Trese MT. Familial exudative vitreoretinopathy. Results of surgical management. Ophthalmology 1998;105:1015–1023.
    1. Warden SM, Andreoli CM, Mukai S. The Wnt signaling pathway in familial exudative vitreoretinopathy and Norrie disease. Semin Ophthalmol 2007;22:211–217.
    1. Ye X, Wang Y, Cahill H, Yu M, Badea TC, Smallwood PM, et al. Norrin, frizzled-4, and Lrp5 signaling in endothelial cells controls a genetic program for retinal vascularization. Cell 2009;139:285–298.
    1. Wang Y, Rattner A, Zhou Y, Williams J, Smallwood PM, Nathans J. Norrin/Frizzled4 signaling in retinal vascular development and blood brain barrier plasticity. Cell 2012;151:1332–1344.
    1. Potente M, Gerhardt H, Carmeliet P. Basic and therapeutic aspects of angiogenesis. Cell 2011;146:873–887.
    1. Chen ZY, Battinelli EM, Fielder A, Bundey S, Sims K, Breakefield XO, et al. A mutation in the Norrie disease gene (NDP) associated with X-linked familial exudative vitreoretinopathy. Nat Genet 1993;5:180–183.
    1. Kim JH, Yu YS, Kim J, Park SS. Mutations of the Norrie gene in Korean ROP infants. Korean J Ophthalmol 2002;16:93–96.
    1. Dickinson JL, Sale MM, Passmore A, FitzGerald LM, Wheatley CM, Burdon KP, et al. Mutations in the NDP gene: contribution to Norrie disease, familial exudative vitreoretinopathy and retinopathy of prematurity. Clin Exp Ophthalmol 2006;34:682–688.
    1. Rathi S, Jalali S, Musada GR, Patnaik S, Balakrishnan D, Hussain A, et al. Mutation spectrum of NDP, FZD4 and TSPAN12 genes in Indian patients with retinopathy of prematurity. Br J Ophthalmol 2018;102:276–281.
    1. Tang M, Sun L, Hu A, Yuan M, Yang Y, Peng X, et al. Mutation spectrum of the LRP5, NDP, and TSPAN12 genes in Chinese patients with familial exudative vitreoretinopathy. Invest Ophthalmol Vis Sci 2017;58:5949–5957.
    1. Li JK, Li Y, Zhang X, Chen CL, Rao YQ, Fei P, et al. Spectrum of variants in 389 Chinese probands with familial exudative vitreoretinopathy. Invest Ophthalmol Vis Sci 2018;59:5368–5381.
    1. Sims KB. NDP-related retinopathies. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, Amemiya A, editors. GeneReviews(R). Seattle (WA): University of Washington, Seattle; 1993.
    1. Jo DH, Kim JH, Kim JH. How to overcome retinal neuropathy: the fight against angiogenesis-related blindness. Arch Pharm Res 2010;33:1557–1565.
    1. Gariano RF, Gardner TW. Retinal angiogenesis in development and disease. Nature 2005;438:960–966.
    1. Mintz-Hittner HA, Kennedy KA, Chuang AZ. BEAT-ROP Cooperative Group. Efficacy of intravitreal bevacizumab for stage 3+ retinopathy of prematurity. N Engl J Med 2011;364:603–615.
    1. Tagami M, Kusuhara S, Honda S, Tsukahara Y, Negi A. Rapid regression of retinal hemorrhage and neovascularization in a case of familial exudative vitreoretinopathy treated with intravitreal bevacizumab. Graefes Arch Clin Exp Ophthalmol 2008;246:1787–1789.
    1. Mansour SE, Browning DJ, Wong K, Flynn HW Jr, Bhavsar AR. The evolving treatment of diabetic retinopathy. Clin Ophthalmol 2020;14:653–678.
    1. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005;352:1685–1695.
    1. Hiatt WR. Medical treatment of peripheral arterial disease and claudication. N Engl J Med 2001;344:1608–1621.
    1. Webster KA. Therapeutic angiogenesis: a complex problem requiring a sophisticated approach. Cardiovasc Toxicol 2003;3:283–298.
    1. Henry TD. Therapeutic angiogenesis. BMJ 1999;318:1536–1539.
    1. Bobek V, Taltynov O, Pinterova D, Kolostova K. Gene therapy of the ischemic lower limb--Therapeutic angiogenesis. Vascul Pharmacol 2006;44:395–405.
    1. Schumacher B, Pecher P, von Specht BU, Stegmann T. Induction of neoangiogenesis in ischemic myocardium by human growth factors: first clinical results of a new treatment of coronary heart disease. Circulation 1998;97:645–650.
    1. Laham RJ, Sellke FW, Edelman ER, Pearlman JD, Ware JA, Brown DL, et al. Local perivascular delivery of basic fibroblast growth factor in patients undergoing coronary bypass surgery: results of a phase I randomized, double-blind, placebo-controlled trial. Circulation 1999;100:1865–1871.
    1. Hendel RC, Henry TD, Rocha-Singh K, Isner JM, Kereiakes DJ, Giordano FJ, et al. Effect of intracoronary recombinant human vascular endothelial growth factor on myocardial perfusion: evidence for a dose-dependent effect. Circulation 2000;101:118–121.
    1. Simons M, Annex BH, Laham RJ, Kleiman N, Henry T, Dauerman H, et al. Pharmacological treatment of coronary artery disease with recombinant fibroblast growth factor-2: double-blind, randomized, controlled clinical trial. Circulation 2002;105:788–793.
    1. Henry TD, Annex BH, McKendall GR, Azrin MA, Lopez JJ, Giordano FJ, et al. The VIVA trial: vascular endothelial growth factor in Ischemia for vascular angiogenesis. Circulation 2003;107:1359–1365.
    1. Giacca M. Virus-mediated gene transfer to induce therapeutic angiogenesis: where do we stand? Int J Nanomedicine 2007;2:527–540.
    1. Losordo DW, Vale PR, Symes JF, Dunnington CH, Esakof DD, Maysky M, et al. Gene therapy for myocardial angiogenesis: initial clinical results with direct myocardial injection of phVEGF165 as sole therapy for myocardial ischemia. Circulation 1998;98:2800–2804.
    1. Rosengart TK, Lee LY, Patel SR, Sanborn TA, Parikh M, Bergman GW, et al. Angiogenesis gene therapy: phase I assessment of direct intramyocardial administration of an adenovirus vector expressing VEGF121 cDNA to individuals with clinically significant severe coronary artery disease. Circulation 1999;100:468–474.
    1. Vale PR, Losordo DW, Milliken CE, McDonald MC, Gravelin LM, Curry CM, et al. Randomized, single-blind, placebo-controlled pilot study of catheter-based myocardial gene transfer for therapeutic angiogenesis using left ventricular electromechanical mapping in patients with chronic myocardial ischemia. Circulation 2001;103:2138–2143.
    1. Losordo DW, Vale PR, Hendel RC, Milliken CE, Fortuin FD, Cummings N, et al. Phase 1/2 placebo-controlled, double-blind, dose-escalating trial of myocardial vascular endothelial growth factor 2 gene transfer by catheter delivery in patients with chronic myocardial ischemia. Circulation 2002;105:2012–2018.
    1. Grines CL, Watkins MW, Helmer G, Penny W, Brinker J, Marmur JD, et al. Angiogenic gene therapy (AGENT) trial in patients with stable angina pectoris. Circulation 2002;105:1291–1297.
    1. Fan Y, Yang GY. Therapeutic angiogenesis for brain ischemia: a brief review. J Neuroimmune Pharmacol 2007;2:284–289.
    1. Goldberg JL, Laughlin MJ. UC blood hematopoietic stem cells and therapeutic angiogenesis. Cytotherapy 2007;9:4–13.
    1. Assmus B, Schächinger V, Teupe C, Britten M, Lehmann R, Döbert N, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 2002;106:3009–3017.
    1. Strauer BE, Brehm M, Zeus T, Köstering M, Hernandez A, Sorg RV, et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 2002;106:1913–1918.
    1. Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107:2294–2302.
    1. Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47–49.
    1. Lazarous DF, Unger EF, Epstein SE, Stine A, Arevalo JL, Chew EY, et al. Basic fibroblast growth factor in patients with intermittent claudication: results of a phase I trial. J Am Coll Cardiol 2000;36:1239–1244.
    1. Lederman RJ, Mendelsohn FO, Anderson RD, Saucedo JF, Tenaglia AN, Hermiller JB, et al. Therapeutic angiogenesis with recombinant fibroblast growth factor-2 for intermittent claudication (the TRAFFIC study): a randomised trial. Lancet 2002;359:2053–2058.
    1. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L, Asahara T, et al. Clinical evidence of angiogenesis after arterial gene transfer of phVEGF165 in patient with ischaemic limb. Lancet 1996;348:370–374.
    1. Baumgartner I, Pieczek A, Manor O, Blair R, Kearney M, Walsh K, et al. Constitutive expression of phVEGF165 after intramuscular gene transfer promotes collateral vessel development in patients with critical limb ischemia. Circulation 1998;97:1114–1123.
    1. Powell RJ, Simons M, Mendelsohn FO, Daniel G, Henry TD, Koga M, et al. Results of a double-blind, placebo-controlled study to assess the safety of intramuscular injection of hepatocyte growth factor plasmid to improve limb perfusion in patients with critical limb ischemia. Circulation 2008;118:58–65.
    1. Mäkinen K, Manninen H, Hedman M, Matsi P, Mussalo H, Alhava E, et al. Increased vascularity detected by digital subtraction angiography after VEGF gene transfer to human lower limb artery: a randomized, placebo-controlled, double-blinded phase II study. Mol Ther 2002;6:127–133.
    1. Rajagopalan S, Mohler ER 3rd, Lederman RJ, Mendelsohn FO, Saucedo JF, Goldman CK, et al. Regional angiogenesis with vascular endothelial growth factor in peripheral arterial disease: a phase II randomized, double-blind, controlled study of adenoviral delivery of vascular endothelial growth factor 121 in patients with disabling intermittent claudication. Circulation 2003;108:1933–1938.
    1. Tateishi-Yuyama E, Matsubara H, Murohara T, Ikeda U, Shintani S, Masaki H, et al. Therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-marrow cells: a pilot study and a randomised controlled trial. Lancet 2002;360:427–435.
    1. Kastrup J, Jørgensen E, Drvota V. Vascular growth factor and gene therapy to induce new vessels in the ischemic myocardium. Therapeutic angiogenesis. Scand Cardiovasc J 2001;35:291–296.
    1. Post MJ, Laham R, Sellke FW, Simons M. Therapeutic angiogenesis in cardiology using protein formulations. Cardiovasc Res 2001;49:522–531.
    1. Khurana R, Simons M. Insights from angiogenesis trials using fibroblast growth factor for advanced arteriosclerotic disease. Trends Cardiovasc Med 2003;13:116–122.
    1. Dor Y, Djonov V, Abramovitch R, Itin A, Fishman GI, Carmeliet P, et al. Conditional switching of VEGF provides new insights into adult neovascularization and pro-angiogenic therapy. EMBO J 2002;21:1939–1947.
    1. Schwarz ER, Speakman MT, Patterson M, Hale SS, Isner JM, Kedes LH, et al. Evaluation of the effects of intramyocardial injection of DNA expressing vascular endothelial growth factor (VEGF) in a myocardial infarction model in the rat--angiogenesis and angioma formation. J Am Coll Cardiol 2000;35:1323–1330.
    1. Syed IS, Sanborn TA, Rosengart TK. Therapeutic angiogenesis: a biologic bypass. Cardiology 2004;101:131–143.
    1. Idris NM, Haider HK, Goh MW, Sim EK. Therapeutic angiogenesis for treatment of peripheral vascular disease. Growth Factors 2004;22:269–279.
    1. Annex BH, Simons M. Growth factor-induced therapeutic angiogenesis in the heart: protein therapy. Cardiovasc Res 2005;65:649–655.
    1. Markkanen JE, Rissanen TT, Kivelä A, Ylä-Herttuala S. Growth factor-induced therapeutic angiogenesis and arteriogenesis in the heart--gene therapy. Cardiovasc Res 2005;65:656–664.
    1. Emanueli C, Madeddu P. Therapeutic angiogenesis: translating experimental concepts to medically relevant goals. Vascul Pharmacol 2006;45:334–339.
    1. Mandai M, Watanabe A, Kurimoto Y, Hirami Y, Morinaga C, Daimon T, et al. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med 2017;376:1038–1046.
    1. Bennett J, Wellman J, Marshall KA, McCague S, Ashtari M, DiStefano-Pappas J, et al. Safety and durability of effect of contralateral-eye administration of AAV2 gene therapy in patients with childhood-onset blindness caused by RPE65 mutations: a follow-on phase 1 trial. Lancet 2016;388:661–672.
    1. Sato T, Kusaka S, Shimojo H, Fujikado T. Simultaneous analyses of vitreous levels of 27 cytokines in eyes with retinopathy of prematurity. Ophthalmology 2009;116:2165–2169.
    1. Zhao M, Xie WK, Bai YJ, Huang LZ, Wang B, Liang JH, et al. Expression of total vascular endothelial growth factor and the anti-angiogenic VEGF 165 b isoform in the vitreous of patients with retinopathy of prematurity. Chin Med J (Engl) 2015;128:2505–2509.
    1. Yenihayat F, Özkan B, Kasap M, Karabaş VL, Güzel N, Akpınar G, et al. Vitreous IL-8 and VEGF levels in diabetic macular edema with or without subretinal fluid. Int Ophthalmol 2019;39:821–828.
    1. Citirik M, Kabatas EU, Batman C, Akin KO, Kabatas N. Vitreous vascular endothelial growth factor concentrations in proliferative diabetic retinopathy versus proliferative vitreoretinopathy. Ophthalmic Res 2012;47:7–12.
    1. Ohlmann A, Scholz M, Goldwich A, Chauhan BK, Hudl K, Ohlmann AV, et al. Ectopic norrin induces growth of ocular capillaries and restores normal retinal angiogenesis in Norrie disease mutant mice. J Neurosci 2005;25:1701–1710.
    1. Ohlmann A, Seitz R, Braunger B, Seitz D, Bösl MR, Tamm ER. Norrin promotes vascular regrowth after oxygen-induced retinal vessel loss and suppresses retinopathy in mice. J Neurosci 2010;30:183–193.
    1. Tokunaga CC, Chen YH, Dailey W, Cheng M, Drenser KA. Retinal vascular rescue of oxygen-induced retinopathy in mice by norrin. Invest Ophthalmol Vis Sci 2013;54:222–229.
    1. Zeilbeck LF, Müller BB, Leopold SA, Senturk B, Langmann T, Tamm ER, et al. Norrin mediates angiogenic properties via the induction of insulin-like growth factor-1. Exp Eye Res 2016;145:317–326.
    1. Cho CH, Kammerer RA, Lee HJ, Yasunaga K, Kim KT, Choi HH, et al. Designed angiopoietin-1 variant, COMP-Ang1, protects against radiation-induced endothelial cell apoptosis. Proc Natl Acad Sci U S A 2004;101:5553–5558.
    1. Lee J, Kim KE, Choi DK, Jang JY, Jung JJ, Kiyonari H, et al. Angiopoietin-1 guides directional angiogenesis through integrin αvβ5 signaling for recovery of ischemic retinopathy. Sci Transl Med 2013;5:203ra127
    1. Butler JM, Guthrie SM, Koc M, Afzal A, Caballero S, Brooks HL, et al. SDF-1 is both necessary and sufficient to promote proliferative retinopathy. J Clin Invest 2005;115:86–93.
    1. Chang KH, Chan-Ling T, McFarland EL, Afzal A, Pan H, Baxter LC, et al. IGF binding protein-3 regulates hematopoietic stem cell and endothelial precursor cell function during vascular development. Proc Natl Acad Sci U S A 2007;104:10595–10600.
    1. Caballero S, Sengupta N, Afzal A, Chang KH, Li Calzi S, Guberski DL, et al. Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes 2007;56:960–967.
    1. Medina RJ, O'Neill CL, Humphreys MW, Gardiner TA, Stitt AW. Outgrowth endothelial cells: characterization and their potential for reversing ischemic retinopathy. Invest Ophthalmol Vis Sci 2010;51:5906–5913.
    1. Prasain N, Lee MR, Vemula S, Meador JL, Yoshimoto M, Ferkowicz MJ, et al. Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony-forming cells. Nat Biotechnol 2014;32:1151–1157.
    1. Wang D, Zhang B, Shi H, Yang W, Bi MC, Song XF, et al. Effect of endothelial progenitor cells derived from human umbilical cord blood on oxygen-induced retinopathy in mice by intravitreal transplantation. Int J Ophthalmol 2016;9:1578–1583.
    1. Li Calzi S, Shaw LC, Moldovan L, Shelley WC, Qi X, Racette L, et al. Progenitor cell combination normalizes retinal vascular development in the oxygen-induced retinopathy (OIR) model. JCI Insight 2019;4:e129224
    1. Otani A, Kinder K, Ewalt K, Otero FJ, Schimmel P, Friedlander M. Bone marrow-derived stem cells target retinal astrocytes and can promote or inhibit retinal angiogenesis. Nat Med 2002;8:1004–1010.
    1. Lin Y, Weisdorf DJ, Solovey A, Hebbel RP. Origins of circulating endothelial cells and endothelial outgrowth from blood. J Clin Invest 2000;105:71–77.
    1. O'Leary OE, Canning P, Reid E, Bertelli PM, McKeown S, Brines M, et al. The vasoreparative potential of endothelial colony-forming cells in the ischemic retina is enhanced by cibinetide, a non-hematopoietic erythropoietin mimetic. Exp Eye Res 2019;182:144–155.
    1. Nomi M, Miyake H, Sugita Y, Fujisawa M, Soker S. Role of growth factors and endothelial cells in therapeutic angiogenesis and tissue engineering. Curr Stem Cell Res Ther 2006;1:333–343.

Metrics
Share
Figures

1 / 2

Tables

1 / 3

Funding Information
PERMALINK