Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Oct 1.
Published in final edited form as: Ocul Surf. 2018 Jun 20;16(4):398–414. doi: 10.1016/j.jtos.2018.06.004

Current and Upcoming Therapies for Corneal Neovascularization

Danial Roshandel 1,#, Medi Eslani 2,3,#, Alireza Baradaran-Rafii 1, Albert Y Cheung 3, Khaliq Kurji 3, Sayena Jabbehdari 1,2, Alejandra Maiz 2, Setareh Jalali 1, Ali R Djalilian 2, Edward J Holland 3
PMCID: PMC6461401  NIHMSID: NIHMS1526153  PMID: 29908870

Abstract

The cornea is unique because of its complete avascularity. Corneal neovascularization (CNV) can result from a variety of etiologies including contact lens wear; corneal infections; and ocular surface diseases due to inflammation, chemical injury, and limbal stem cell deficiency. Management is focused primarily on the etiology and pathophysiology causing the CNV and involves medical and surgical options. Because inflammation is a key factor in the pathophysiology of CNV, corticosteroids and other anti-inflammatory medications remain the mainstay of treatment. Anti-VEGF therapies are gaining popularity to prevent CNV in a number of etiologies. Surgical options including vessel occlusion and ocular surface reconstruction are other options depending on etiology and response to medical therapy. Future therapies should provide more effective treatment options for the management of CNV.

Keywords: Eye, Cornea, Vascularization, Neovascularization, Angiogenesis, Limbal stem cell deficiency, Inflammation

INTRODUCTION

Corneal neovascularization (CNV) can result from a variety of etiologies such as contact lens wear; corneal infections; and ocular surface inflammation and injury including limbal stem cell deficiency (LSCD).13 CNV may result in decreased visual acuity from the sequelae of blood vessels invading the cornea and causing opacification of the stroma and irregularity of the corneal surface. Surface irregularity results in higher order aberrations; this can be accompanied by extravasation of fluid and lipids, which leads to corneal edema and lipid keratopathy as well as alteration of the stromal architecture.4 These changes minimize corneal clarity and thus impede vision. In a comprehensive ophthalmology clinic, CNV was found in 4.14% of patients, of which 12% showed a decrease in visual acuity.5 While CNV can be helpful in certain circumstances (e.g. uncontrolled corneal infections, conditions involving stromal necrosis), it is more often a pathologic consequence of various ocular surface and corneal disorders. Notably, CNV is an important risk factor for corneal graft rejection and subsequent failure.6

DEFINITION AND TERMINOLOGY

The cornea is unique because it is completely avascular and alymphatic, which is essential for its clarity and optimal vision. When blood and lymphatic vessels from the pericorneal vascular plexus grow into the cornea, the result is a pathologic condition termed corneal hemangiogenesis and corneal lymphangiogenesis, respectively.

It has been proposed that the term corneal vascularization is appropriate in contrast to neovascularization, as the latter refers to a condition in which new blood vessels arise from pre-existing ones. To avoid confusion with choroidal neovascularization and due to the absence of pre-existing blood vessels in the cornea, the term “corneal vascularization” should be used for corneal vessel formation.7 However, in reviewing the literature, majority of studies term this pathology “corneal neovascularization”. To follow prior studies in this field, the term “corneal neovascularization (CNV)” is used throughout this review.

In many pathologic conditions, lymphatic vessels grow into the cornea parallel to blood vessels. Lymphangiogenesis plays a critical role in many processes such as immunity, infection, and metastasis.8

VASCULOGENESIS VERSUS ANGIOGENESIS

Vasculogenesis comprises the de novo formation of vessels from vascular endothelial precursor cells (i.e. hemangioblasts and angioblasts) which are derived from mesodermal precursors (via mesodermal induction).9 In contrast, angiogenesis is a process in which endothelial cells of pre-existing vessels proliferate and form new vessels.9 In CNV, the endothelial cells of newly formed corneal vessels originate from pre-existing limbal vessels (i.e. angiogenesis). However, pericytes, another crucial cell type in blood vessel formation, originate from bone-marrow derived precursors (i.e. vasculogenesis).10 Ozerdem and colleagues believe that both angiogenesis and vasculogenesis are involved in CNV and that targeting both mechanisms would be most effective in managing this condition.10 Similar to blood vessels, lymphatic vessels may arise de novo from bone-marrow derived cells (i.e. CD11b-positive macrophages) or they may extend from pre-existing limbal lymphatic vessels.8, 11

CORNEAL VASCULAR PRIVILEGE

Previous studies have identified a number of mechanism(s) by which the limbal vascular plexus does not invade the cornea under normal physiologic conditions. It is believed that an imbalance between angiogenic and anti-angiogenic mechanisms in the cornea results in CNV.12

The first proposed mechanism for CNV was proposed by Cogan, who claimed corneal swelling and subsequent disintegration of the corneal lamellae were the sole factors responsible for CNV.13 However, further investigation revealed that corneal swelling is necessary but not sufficient for the development of CNV.14, 15

While there is no anatomical boundary between the limbal vascular plexus and the clear cornea, the angiostatic function of the limbus has been proposed as a mechanism for corneal avascularity, especially since LSCD is often associated with CNV.1618 It is unclear whether the limbus exerts its barrier function via a physical or functional mechanism, or both. The physical barrier effect of the limbus has been proposed by Friedenwald as a “growth pressure theory,” in which continuous self­renewal of the limbal stem cells prevents invasion of the conjunctival epithelium and subsequent vascularization of the cornea.19 However, using a murine hemilimbal corneal injury model, Tobaigy showed factors other than the limbal barrier are involved to maintain corneal avascularity.20

Although earlier reports supported the angiogenic properties of corneal epithelium,21, 22 the predominantly anti-angiogenic role of the corneal epithelium has been widely accepted in more recent studies.23 Clinically, the association of a persistent corneal epithelial defect (PED) with CNV and its resolution after epithelial transplantation further supports the role of corneal epithelium in preventing CNV.24

Interestingly, the corneal epithelium releases pro-angiogenic factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF), which are then sequestrated by the basement membrane (BM) under normal conditions.22, 25 For example, Ambati and colleagues found that the cornea contains a high quantity of VEGF-A, a potent pro-angiogenic molecule. However, it is almost completely bound to the soluble VEGF-receptor 1 (also known as soluble fms-like tyrosine kinase-1 sflt-1), thus preventing its angiogenic effects.26 They concluded that sflt-1 is a crucial factor in corneal avascularity.26 Ambati and colleagues have also reported that expression of sflt-1 is significantly lower in vascularized corneas (secondary to alkali burn, ocular cicatricial pemphigoid, interstitial keratitis, and aniridia) when compared to normal human corneas.27 Inhibitory PAS (Per/Arnt/Sim) domain protein is another corneal epithelial derived factor with antiangiogenic properties, specifically against hypoxia inducible factor (HIF)/Hypoxia induced CNV.28 In addition, VEGF receptor 3, which is constitutively expressed by the corneal epithelium, is an inhibitor of corneal angiogenesis.29

The corneal epithelial BM also contains anti-angiogenic factors such as tissue inhibitor of metalloproteinase 3 (TIMP-3) and collagen XVIII/endostatin.30, 31 Angiostatin, restin, arrestin, endostatin, canstatin, tumstatin, thrombospondins, interleukin-1 receptor antagonist, pigment epithelial derived factor (PEDF), vasoactive intestinal peptide (VIP) and α-melanocyte stimulating hormone (α-MSH) are also anti-angiogenic molecules, which have been found in the cornea and/or the aqueous humor.4, 3234 Given that the cornea contains both angiogenic and anti­angiogenic factors, damage to the basement membrane (BM) due to LSCD or persistent epithelial defects may result in the release of pro-angiogenic factors and loss of anti-angiogenic factors, and thus lead to CNV.35

Several molecules with anti-lymphangiogenic properties have been identified in the cornea and aqueous humor. These include alternatively spliced VEGF receptor-2 (soluble VEGFR-2), tumor necrosis factor superfamily member 10 (Tnfsf10/Trail), tissue plasminogen activator (tPA), and thrombospondin 1 in the cornea as well as VIP and α-MSH in the aqueous humor.33, 3638

HEMANGIOGENESIS VERSUS LYMPHANGIOGENESIS

The lymphatic system is a network of vessels throughout the body that allows lymphatic fluid to return to the systemic blood circulation. In addition to lacking blood vessels, the cornea is also devoid of lymphatic vessels. The paucity of blood vessels prevents immune cells from accessing corneal antigens, and the lack of lymphatic vessels prevents cellular and cytokine traffic to the regional lymph nodes.39 While several anti- (hem) angiogenic factors are known to be present in the normal cornea (as mentioned above), the anti- (lymph) angiogenic factors are still yet unknown. However, in vascularized human corneas, the degree of lymphangiogenesis is significantly correlated with the degree of hemangiogenesis.40, 41 In contrast, corneal lymphangiogenesis may also occur in an avascular cornea. Transient physiologic lymphangiogenesis has been reported to reduce corneal edema and increase corneal transparency in cases with corneal edema.42

Lymphatic vessels can provide a drainage pathway for both antigenic material (cells, cellular debris) and more importantly antigen presenting cells.43, 44 Besides enabling the transport itself, lymphatic vessels enhance the speed and amount of antigenic material or antigen presenting cells that reach the regional lymph nodes.45 This is particularly important after corneal transplantation.46 Corneal lymphangiogenesis provides a route of exit from the graft to regional lymph nodes, which has been shown to be essential in promoting alloimmunization and subsequent graft rejection.47, 48 Corneal lymphatics in vascularized human host beds adjacent to grafted tissue could enhance the traffic of graft-derived antigens to regional lymph nodes, thereby promoting rejection.39

MECHANISMS OF CORNEAL NEOVASCULARIZATION

Cogan originally described histologic evidence of a mechanism following corneal inciting injury, which led to local corneal edema accompanied by aneurysmal engorgement of venules and capillaries. After a few days, these were replaced by tiny spicule-like masses of hemorrhages in a radiating pattern which could later either form capillary channels or regress /disappear. The channels typically formed an extension in front of a pre-existing loop. With repetition of this cycle, there was progressive movement of the vessels toward the central cornea.13 Conversely, Shi and colleagues described three pivotal steps to CNV in five models (suture-mediated, alkali injury, fungal infection, or implantation of immunogen or tumor cells).49 These pivotal steps included a sprout period, a vigorous stage, and a regressive stage. Although in all of the models the initial findings (corneal edema and vascular dilation) were similar to Cogan’s investigations, Shi and colleagues identified more details of the events leading to CNV. First, the BM of the perilimbal capillary network is degraded by proteases released by endothelial cells. Thereafter, endothelial cells (ECs) migrate and invade the extracellular matrix (ECM) and begin to proliferate. Finally, the lumen of the new vessels forms, and the BM is remodeled.49

The mounting evidence in literature clearly show CNV and corneal lymphangiogenesis are the result of sprouting from pre-existing limbal blood and lymphatic vascular arcade.50, 51 Furthermore, there are also reports that corneal edema is neither necessary nor sufficient for CNV.52, 53

INFLAMMATION

Inflammation is the core mechanism of CNV induced by any etiology including chemical injury, infection (Figure 1A), immune disorders (Figure 1C), LSCD (Figure 2), and hypoxia.12, 54 There is an interplay between inflammatory cells (especially macrophages and neutrophils) and angiogenic growth factors (i.e. VEGF family) in inflammation induced CNV. VEGF-A mediated recruitment of macrophages may initiate a process called “immune amplification cascade” that promotes corneal hemangiogenesis and lymphangiogenesis.50 Inflammatory cells that are recruited during corneal injury, especially macrophages, produce pro-angiogenic factors and proteolytic enzymes which promote limbal vascular endothelial cell proliferation and migration.50, 5557 Pro-inflammatory cytokines and chemokines are strong mediators of angiogenesis in humans and are over-expressed in corneal inflammation.5866

Figure 1.

Figure 1.

Slit lamp photographs demonstrating CNV. Necrotic corneal ulcer and persistent epithelial defect with surrounding ring infiltrate and CNV secondary to acanthamoeba (1A, photograph courtesy of Marius Miron). Stromal CNV from acute stromal rejection of a deep anterior lamellar keratoplasty (1B). Regressing deep CNV with mild surrounding haze and lipid secondary to HSV immune stromal keratitis (1C). Deep frond of CNV with accompanying lipid deposition from long-term rigid gas permeable contact lens abuse (1D).

Figure 2.

Figure 2.

Slit lamp photographs of CNV secondary to limbal stem cell deficiency from various etiologies are depicted: Stevens Johnson Syndrome (2A), alkaline chemical injury (2B), mucous membrane pemphigoid (2C), and long-term soft contact lens abuse (2D).

IL-1 is a key cytokine in inflammatory angiogenesis.64 It is released mainly from injured corneal epithelium and directly stimulates proliferation as well as migration of endothelial cells.58, 66 IL-1 also enhances production of strong pro-angiogenic molecules VEGF and bFGF,67 increases the expression of adhesion molecules and inflammatory mediators by human corneal epithelial cells, and recruits leukocytes via production of chemokines.68 IL-6 is another proinflammatory cytokine which can promote CNV by increasing VEGF production in corneal and inflammatory cells in herpes simplex virus (HSV) keratitis.61 Recombinant human IL-8 (rhIL-8) under physiologic concentrations proved to be a potent stimulator of CNV in vivo.65,69 IL-17A has also been implicated in the pathogenesis of HSV infection associated with CNV.69

Matrix metalloproteinases (MMPs) have a dual role in corneal angiogenesis. Release of MMPs (especially MMP-2/gelatinase A) from injured corneal epithelium and leukocytes degrades the basement membrane,70 resulting in the release of sequestrated pro-angiogenic factors as previously discussed. MMPs also degrade the ECM components, creating a physical space and facilitating endothelial cell migration during angiogenesis.71 Simultaneously, some MMPs show anti-angiogenic activity by catalyzing the production of angiostatic mediators such as endostatin and angiostatin from their precursors.7274

LIMBAL STEM CELL DEFICIENCY (LSCD)

LSCD, whether congenital or acquired, is commonly associated with CNV.75 According to Friedenwald’s theory of growth pressure, loss of the physical barrier effect of the limbus is the main cause of CNV following LSCD.19 However, Tobaigy and Azar demonstrated that when half of the limbus and more than half of the corneal epithelium were removed, vessels grew from the opposite side of the defect.20 This observation led to the conclusion that factors other than the physical barrier are responsible for the limbal angiostatic effect.20

Previous studies have shown that damage to limbal stem cells (LSCs) induces long-standing inflammation and recruits macrophages,76 which are important sources of VEGF.77 Additionally, LSCD is followed by amplification of tissue growth factor beta (TGF-β) signaling which exacerbates inflammation and VEGF production.63, 78 The vicious cycle of increasing inflammation causes destruction of the remaining LSCs and progression towards total LSCD. The amplification of VEGF and TGF- β, key angiogenic factors, contributes to CNV. Finally, the establishment of CNV in the absence of the limbal barrier leads to the invasion of the conjunctival epithelium toward the corneal surface, resulting in corneal conjunctivalization.

HYPOXIA

Hypoxia is a classic stimulator of angiogenesis. In the hypoxic cornea, VEGF is expressed by corneal epithelial and endothelial cells, as well as the endothelium of limbal vessels in an attempt to enhance the supply of oxygen to the cornea.77, 79 However, in an animal model of closed-eye contact lens-induced CNV, increased expression of VEGF did not correlate with inflammation.80 Since VEGF is present in the tear film of the normal non-vascularized cornea and is almost completely inactivated by sflt-1, other factors may also mediate CNV in the hypoxic state.80 Hypoxia inducible factor-1 alpha (HIF-1α) plays an essential role in the response to hypoxia and contributes to angiogenesis.81, 82 Similar to VEGF, there is an inhibitory mechanism for HIF-1α in hypoxic conditions which prevents HIF-1α induced angiogenesis in the cornea.28 Although bFGF, a second angiogenic growth factor, was not found to play a significant role in hypoxia-induced corneal angiogenesis,83, 84 other angiogenic cytokines and chemokines that are upregulated by hypoxia may contribute to hypoxic CNV.85 While nitric oxide (NO) is a potent vasodilator which is upregulated by hypoxia and has both pro- and anti-angiogenic activities in a dose dependent manner,86, 87 the effects of hypoxia on corneal NO production and angiogenesis have not yet been fully elucidated.

Inflammation induced by hypoxia might possibly be the most relevant factor for corneal angiogenesis. Hypoxia induces the production of potent inflammatory metabolites of arachidonic acid by the corneal epithelium via the cytochrome p450 (CYP) pathway.88 The most important metabolites are 12-hydroxy-5,8,11,14-eicosatetraenoic acid (12-HETE) which promotes corneal edema and 12-hydroxy-5,8,14-eicosatrienoic acid (12-HETrE) which incites an inflammatory cascade resulting in neutrophil chemotaxis and CNV.89

IMPAIRED CORNEAL INNERVATION

Ferrari and colleagues elucidated the relationship between corneal innervation and angiogenesis in mice models of bFGF-induced corneal vascularization. They concluded that when vessel growth is stimulated, nerves disappear and, conversely, denervation induces angiogenesis. They suggested that this phenomenon is mediated, at least in part, by the reduction of angiostatic molecules (including epithelial-derived PEDF and epithelial VEGFR3) constitutively expressed under normal physiologic conditions by the cornea.90

MOLECULAR PATHWAYS

VEGF

The VEGF family comprises five members that regulate vasculogenesis, angiogenesis, and lymphangiogenesis.91 VEGF-A (also known as VEGF) is a crucial factor for vessel formation and maturation in embryotic and adult tissues. It is over­expressed in various vasculogenic corneal pathologies including LSCD92, inflammation93, chemical injuries94, hypoxia95, edema96 and infections.97 The main sources of VEGF in the human cornea are epithelial, stromal, and endothelial cells as well as macrophages/inflammatory cells, vascular ECs, and pericytes.77

VEGF exerts its angiogenic effects via two major receptor tyrosine kinase (RTK); the VEGFR-1 (flt-1), and VEGFR-2 (kinase insert domain-containing receptor; flk-1/KDR) which are mainly expressed by the vascular ECs. Although VEGFR-1 has higher affinity for the VEGF, its kinase activity is much weaker than VEGFR-2.98, 99 On the other hands, VEGFR-1 is negatively regulated in the cornea by the sflt-1100 and a low affinity VEGF isoform called VEGF165b101 which competes with VEGF-A. Thus, angiogenic activity of the VEGF is mostly related to the VEGFR-2 pathway, while VEGFR-1 has a regulatory rather than angiogenic effect.

Once VEGF binds to its receptor, VEGFR-2, it induces dimerization and autophosphorylation of the receptor at the 1175-phosphotyrosine site which activates phospholipase C, gamma 1 (PLCg1) and finally the PKC-Ca++-c-Raf-MEK-MAPK pathway.102 It leads to activation of the angiogenic cascades including proteolytic activities, proliferation, migration and tube formation of ECs, and maturation of new blood vessels.

VEGFR-1 has no kinase activity in vascular ECs and does not stimulate EC proliferation directly.103 In turn, its activation in macrophages activates the receptor for activated C kinase 1 (RACK1) dependent PI3-AKT pathway104, which stimulates macrophage migration and promotes tissue remodeling which is essential for tube formation and maturation of the vessels.

There are also non-tyrosine kinase-type receptors such as neuropilins (NRP)105 heparin sulfate proteoglycans (HSPGs)106 that regulate VEGF activity via receptors.

PDGF

The Platelet-derived growth factor (PDGF) family comprises 5 dimeric ligands including PDGF-AA, -AB, -BB, -CC and –DD. Although platelet granules are the primary source of PDGF, it can also be released by other cells such as monocytes, vascular endothelium, and vascular smooth muscle cells.107 It can also be expressed by corneal fibroblasts, epithelial, and endothelial cells.108 PDGF receptor (PDGFR) is a tyrosine kinase receptor, composed of alpha and beta chains, that is dimerized upon stimulation by the related ligand. Once PDGF attaches to its receptor, the kinase activity leads to receptor autophosphorylation, which in turn activates multiple signaling molecules including steroid receptor coactivator (SRC), Phosphoinositide 3-kinase (PI3K), and phospholipase Cγ 1 (PLCγ).109 PDGF-BB and PDGFR-β, which are the predominant subtypes in corneal cells, have the greatest role in the angiogenic process.110

The most important physiologic role of PDGF in angiogenesis is recruitment, proliferation, and viability of pericytes.111, 112 Furthermore, it stimulates expression of VEGF by pericytes which can enhance endothelial cell survival.113 Inhibition of the PDGF signaling pathway by PDGFR-β or PI3K inhibitors results in loss of pericytes and decrease of corneal vessels which is correlated with reduced expression of PDGF, VEGF, and other angiogenic molecules.114 In addition, PDGF inhibitors might increase susceptibility of the corneal vessels to anti-VEGF therapy through pericyte detachment and decreased endothelial pericyte coverage.115

BASIC FGF

Fibroblast growth factor (FGF) is another angiogenic growth factor with a complex effect on vascular development. Following corneal injury, FGFs, especially FGF-2 (bFGF), are expressed by the corneal epithelial and stromal cells, as well as inflammatory cells,116 and they promote angiogenesis via their interaction with FGF receptors (mainly FGFR-1) of the vascular ECs. The subsequent dimerization of the FGFRs activates multiple signaling pathways including mitogen-activated protein kinase (MAPK), which in turn induces migration, proliferation, and tubule formation of the ECs; there is also increased protease activity and expression of the cell adhesion molecules (CAMs).117 Additionally, bFGF promotes angiogenesis by induction of VEGF expression.118

NITRIC OXIDE

Nitric oxide (NO) is a free radical produced from L-arginine by three enzymes including neuronal NO synthase (nNOS; NOS-I), inducible NO synthase (iNOS; NOS-II), and endothelial NO synthase (eNOS, NOS-III).119 NO, also referred to as endothelium-derived relaxing factor (EDRF), has been implicated in new vessel formation because of its vasodilatory effects, a phenomenon which usually precedes angiogenesis. However, the role of NO in angiogenesis is controversial probably due to the different isoforms of NOSs involved in the applied models.120, 121

Hypoxia and inflammation, which are important factors in promoting CNV, are potent stimulators of iNOS activity.122, 123 In hypoxia induced angiogenesis, NO synthesized by iNOS is downstream to the HIF-1 and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) but upstream to the VEGF and bFGF pathways.122 Studies have shown that iNOS is a crucial factor in angiogenesis related to tumors since its inhibition effectively blocks tumor angiogenesis.124 Similar results were reported in a cauterization-induced CNV model in mice.125 On the other hand, it has been shown that iNOS is overexpressed in alkali-burn corneas and plays an inhibitory role in CNV.126 The other isoenzyme eNOS is the predominant type of NOS in endothelial cells. It has been shown to be an important modulator of angiogenesis in ischemic conditions by regulating proliferation and migration of endothelial cells.127 Upregulation of eNOS activity in ischemic conditions can be mediated by VEGF, bFGF, and substance P.86, 128, 129 The effects of NO on the vascular endothelial and smooth muscle cells is mainly driven by the cGMP pathway which results in vascular dilation, permeability, and finally angiogenesis.130

RHO/ROCK PATHWAY

The main effect of Rho-associated protein kinase (ROCK) pathway is regulation of cell polarity and cell migration via stimulation of cellular protrusions and contractions as well as focal adhesions. This translates to a great impact on the angiogenic processes by inducing migration of endothelial cells and tube formation. Tumor angiogenesis has been enhanced by activation of ROCK.131 One of the suggested underlying mechanisms of ROCK-induced angiogenesis is regulating actin-binding proteins including Moesin, Radixin, and Ezrin.132, 133 Sonic Hedgehog (Shh) protein and VEGF are up-stream inducers of the ROCK pathway.134136

The expression of Shh is up-regulated in tissues under ischemia, leading to enhanced vascularization.134, 135 It has been shown that Shh directly induces angiogenesis by up-regulating the expression of pro-angiogenic genes e.g. MMP-9 and osteopontin (OPN) via Rho/ROCK signaling pathway.137 Moreover, the Shh induces the expression of VEGF and Angiopoietin 1 (Ang1).134, 135

The induction of Rho/ROCK pathway with VEGF results in phosphorylation of focal adhesion kinases followed by migration of endothelial cells.138 Treatment of endothelial cells with ROCK inhibitor (Y27632) has led to inhibiting migration and tube formation induced by VEGF.136 In vivo studies have also indicated the role of ROCK in angiogenesis. Y27632 reduces hypoxia-induced angiogenesis in the lung.139 Additionally, the VEGF-induced angiogenesis has been suppressed by fasudil (another ROCK inhibitor) both in vitro and in vivo.140, 141

WNT PATHWAY

The canonical Wnt signaling pathway involves various physiological activities such as cell proliferation, differentiation, migration, and apoptosis via regulating the expression of several genes.142 The role of Wnt signaling in angiogenesis has been well established.143 The signaling pathway of canonical Wnt starts by binding of Wnt ligands to a cell surface receptor complex. It results in cytoplasmic β-catenin stabilization via reducing its phosphorylation. Non-phosphorylated-β-catenin will then translocate into the nucleus and with association with T cell factor lead to activation of Wnt target gene transcription such as VEGF and other angiogenic factors.144, 145

The Wnt/β-catenin signaling pathway also plays a pivotal role in the processes involved in corneal wound healing including stem cell proliferation and angiogenesis.146 In an animal study, alkali corneal burns resulted in increased Wnt signaling activity, which was associated with elevated VEGF level in burned corneal buttons. In this study, inhibition of the Wnt signaling pathway was associated with a significant decrease in Wnt signaling activity, VEGF levels, and CNV.147

NOD1 PATHWAY

Nucleotide-binding oligomerization domain 1 (Nod1) receptor is a member of Nod­like receptors (NLRs) that can be expressed by human corneal epithelial cells and may play an important role in inflammatory responses as well as angiogenesis.148 Upon stimulation, Nod1 activates nuclear factor-kappa B (NF-κB) via a kinase called RICK.149 Activation of NF-κB leads to secretion of inflammatory cytokines/chemokines, which can mediate angiogenesis (as discussed above). Stimulation of Nod1 signaling in alkali-induced CNV models resulted in increased vascularization, while Nod1 blocking reversed the condition.150 Hence, the Nod1 pathway might have an important role in CNV and could be considered as a therapeutic target in the future.

EVALUATION

CLINICAL EVALUATION

Inatomi and colleagues introduced a clinical classification for corneal vascularization which allows one to determine the severity of CNV. Grade 1 is the least severe category and indicates peripheral vascularization. Grade 2 indicates peripheral and mid-peripheral vascularization; grade 3 shows modest vascularization involving the entire cornea; finally, grade 4 is the most severe category and indicates massive vascularization of the entire cornea.151

Faraj and colleagues assessed parameters including location, depth, length, branching pattern, color, lipid leakage, nature of blood flow, and presence of hemorrhage to classify corneal vascularization. According to their study, CNV can be classified as active young, active old, mature, partially regressed, or regressed vessels. Their scoring system also considered the number of involved quadrants, depth, and location of the vessels in the setting of CNV secondary to infectious keratitis. With this grading system, herpes simplex keratitis (HSK) and acanthamoeba keratitis had the most and least severe CNV, respectively.152 Furthermore, some CNV characteristics may be suggestive of specific etiology. For example, lipid keratopathy is more likely to be observed in CNV related to viral keratitis, contact lens use (Figure 1D, 2D), and deep anterior lamellar keratoplasty (DALK) (Figure 1B).152

Different parameters have been used for quantifying CNV, including the number of major thick-walled vessels originating from the limbus and reaching the cornea, vessel caliber, and involved CNV area.153

ANTERIOR SEGMENT IMAGING

Ziche and colleagues created a scoring system for CNV quantification which functions by multiplying the vessel density (number of corneal vessels) by the distance from the limbus (in millimeters).154 Furthermore, image processing software such as ImageJ (available online at https://imagej.nih.gov/ij/) can be used to determine the extent of CNV most accurately.155 With this software, boundaries of vessels may be determined manually with cursors or by a semi-automated approach. The semi-automated method eliminates interobserver and intraobserver variability.

Tatham and colleagues developed a semi-automated software and compared its reproducibility with 3 different clinicians. Results indicated the semi-automated software to be roughly 95% reproducible during assessment of CNV while clinicians were only approximately 10% reproducible. As a result, Tatham and colleagues concluded that computer-aided analysis of corneal photographs provided a more reproducible method for quantifying CNV compared to clinicians.156 In other studies, morphometric image analysis of standardized slit-lamp pictures has been used based on gray filter sampling for semi-automatic, semi-quantitative measurement of the extent and progression of CNV in several phase II and III clinical trials.157, 158 Newer imaging technologies have allowed for anatomical and functional evaluation of non-visible and small corneal vessels that cannot be detected with high quality images. Anijeet and colleagues used anterior segment angiography with fluorescein and indocyanine green (ICGA), which showed three to four-fold more visibility of corneal vessels compared to color images. Results also indicated that fluorescein is more effective than ICGA to show leakage from vessel apices.159 Kirwan et al also assessed fluorescein and ICGA findings in patients with keratitis. To do so, four parameters were used: vessel area, diameter, tortuosity, and dye leakage. Fluorescein was useful for detecting vessel leakage, while ICGA was excellent in vessel delineation even in the presence of a stromal scar. Both methods were significantly more sensitive in detecting corneal vessels compared with color images.160

More recently, optical coherence tomography angiography (OCTA) has been shown to provide a feasible, less invasive, more rapid method for evaluation of ocular vasculature including corneal vessels. Using a combination of OCT imaging and angiography has the advantage of determining the depth of vessels and acquisition of en-face views.161, 162

In vivo confocal microscopy (IVCM) has been shown to be useful in assessment of depth and activity of both blood and lymphatic vessels in the cornea. Based on these images, intravascular red blood cell traffic was present in active but not in inactive (ghost) vessels. Moreover, active vessels demonstrated barely visible walls and contained nucleated cells.163

Corneal lymphatic vessels are more difficult to visualize by non-invasive imaging. However, there are reports of detection of corneal lymphatics using IVCM164 and more recently microscopic OCT (mOCT).165 In both techniques, lymphatics were characterized as vessel-like structures with sparse, slowly moving immune cells.

HISTOLOGIC EVALUATION

Histologic features of CNV vary according to the pathologic condition and the chronologic stage of vessel formation. Disruption of the lamellar corneal architecture is a consistent finding in almost all cases of CNV. Cellular infiltration is another common feature of vascularized corneas, which may occur due to the underlying inflammatory disease or increased vascular permeability. A rabbit CNV model revealed that in early stages the vessel wall is composed of a thin layer of cells without muscle and nerve supply, indicating that the vessels are capillaries rather than arterioles or venules. However, in later stages, venules, veins, and finally arterioles are seen.166 Grading or scoring of CNV in a histologic section can be performed by counting the number of vessels per square millimeter167 or by measuring the total vascularized area and the percentage of the cornea covered by vessels.115

Corneal vessels can be identified by hematoxylin-eosin staining, and cell surface markers can be used to determine corneal vessels in corneal buttons. The most commonly used markers for determining blood and lymphatic vessels in corneal buttons include CD31, LYVE-1 and podoplanin. It has been shown that CD31+/LYVE1+++/Podoplanin+ vessels are lymphatics, whereas CD31+++/LYVE-1-/Podplanin- vessels are blood vessels.41, 168 Cursiefen and colleagues found histologic evidence of CNV in roughly 20% of corneal buttons obtained from patients who underwent keratoplasty.169

MANAGEMENT STRATEGIES

Management is focused primarily on the underlying etiology and pathophysiology causing the CNV; for example, if inflammation is the primary mechanism, the best treatment is to control the inflammation. It is important to discontinue contact lens use if contact lens–related hypoxia is suspected. Generally, management consists of medical and surgical strategies.

MEDICAL OPTIONS

Table 1 summarizes some of the medications for potential use as anti-angiogenic therapy although most of them have only been used in experimental studies.

Table 1.

Novel agents, with potential antiangiogenic effects, used for treatment of corneal neovascularization in animal models.

Agentref Model Route Proposed Mechanism(s)
Diospyros kaki Extract (EEDK)309 Alkali burn Oral Suppression of VEGF, FGF, IL-6, and MMP-2.
Recombinant PEDF310 Chemical injury S.C. Inhibits VEGF, bFGF and IL-8.
PEDF311 Chemical injury Topical Downregulates VEGF expression.
Gold nanoparticles312 Chemical injury Topical Decreases VEGFR-2 levels, inhibits ERK phosphorylation.
interferon-induced protein of 10 kDa (IP-10)313 Chemical injury Topical Decreases VEGF and bFGF expression, EC proliferation and tube formation.
Netrin 1314 Chemical injury Topical Laminin-related protein; Decrease inflammation, decrease VEGF, increase PEDF.
Netrin-4315 Chemical injury Topical Laminin-related protein Decreases leukocyte infiltration and VEGF and NF-κB signaling and EC migration, invasion and tube formation, increases PEDF.
LCB54–0009316 Suture induced and chemical injury S.C. Imidazole-based alkaloid Derivative; Antioxidant; decreases VEGF-A and HIF-1α level, inhibits VEGFR-2 and NF-κB signaling, decrease MMP-1, −2, −3 and −9 activity.
Epigallocatechin gallate317 Suture induced Topical Flavonoid; Decreases expression of VEGF and COX-2.
Largazole318 Chemical injury Topical Histone deacetylase inhibitor; Decreases expression of VEGF, b-FGF, TGFβ1 and EGF.
Serine Proteinase Inhibitor A3K (SERPINA3K)319 Suture induced Topical Inhibits Wnt signaling pathway and VEGF
TC14012320 Chemical injury S.C. CXCR4 antagonist and CXCR7 agonist; Reduce CXCR4, CXCR7, VEGF and MMP-2 and −9 mRNA levels*
Itraconazole321 Chemical injury Topical, S.C., I.P. Inhibits cholesterol biosynthesis, endothelial cell proliferation and capillary tube formation.
Lanepitant322 Chemical injury and Suture induced Topical NK1 receptor antagonist; Reduces corneal substance P level and leukocyte infiltration.
Low-molecular-weight heparin-taurocholate 7 (LHT7)323 Chemical injury S.C. Blocks VEGF-VEGFR.
Recombinant C-terminal fragment BIGH3 protein324 Micropocket assay Local Blocks phosphorylation of PI3K/Akt and ERK, inhibits tube formation, increase EC apoptosis.
Celastrol325 Suture induced Topical Reduce the expression of VEGF, MMP-9, and MCP-1.
ADP-ribosylation factor326 Chemical injury Topical Ras-related small GTPase; Downregulates corneal VEGF expression, increases EC apoptosis.
SB-328437327 Chemical injury Topical CCR3 antagonist; Reduces intracorneal MCP-1 and MCP-3 mRNA expression.
Ascorbic acid328 Suture induced Topical Anti-VEGF and anti-MMP.
Cetuximab329 Chemical injury S.C. Anti-EGFR mab; Inhibits EGFR, modulate VEGF and IL-8.
Peroxiredoxin-6330 Ultraviolet radiation Topical Anti-oxidant; Inhibits NF-κB, decrease VEGF expression, increase PEDF expression.
CL9189AP331 Chemical injury Anti-SDF-α1 mab; Inhibits SDF-α1/CXCR4 pathway, down regulates VEGF and C-Kit expression.
Methotrexate332 Suture induced Topical, S.C. Cytotoxic agent; Decreases VEGF and IL-6 expression.
N-acetyl-L-cysteine333 Chemical injury I.P. Anti-oxidant; Down-regulates NF-κB pathway.
Suramab334 Chemical injury I.V. Combined Bevacizumab and Suramin; Inhibits VEGF, bFGF, PDGF, IGF, TGF-β.
Canstatin335 Chemical injury I.P. NC1 domain of the α2 chain of type IV collagen; Inhibits VEGF, HIF-α and TNF-α, prevents EC migration and tube formation.
AMD3100336 Chemical injury S.C. Antagonist of CXCR4; Decreases inflammation, VEGFR-2 expression and EC proliferation.
Dihydroartemisinin337 Suture induced Topical Novel anti-malarial drug; Reduces expression of and phosphorylation of VEGF and VEGFR-2, ERK1/2 and p38.
Parstatin338 Chemical injury S.C. Proteinase-activated receptor 1; Inhibits FGF-2, VEGF, ERK1/2 and MAPK.
PTK/ZK, ZK991339 Suture induced Oral VEGFR-tyrosine kinase inhibitors; Block VEGF receptors.
IMD0354340 Suture induced Systemic Selective blocker of the IKK complex IκB kinase β (IKK2). Decreases inflammatory cell invasion, suppressed CCL2, CXCL5, Cxcr2, and TNF-a and VEGF-A expression.
IL-1R antagonist341 Suture induced Topical Inhibits IL-1 induced angiogenesis.
H-KI20342 Suture induced and Micropocket assay Topical A 20-amino acid peptide from HGF with anti-inflammatory and antiangiogenic preoperties.
Angiopoietin-like protein 2 (ANGPTL2)343 Chemical injury Topical Short heparin RNA inhibiting ANGPTL2 induced inflammation and angiogenesis.
Tissue inhibitors of matrix metalloproteinases344 Micropocket assay Topical Inhibits proliferation and migration of human ECs.
suberoylanilide hydroxamic acid (SAHA)345 Chemical injury Topical Downregulates the expression of VEGF, bFGF, TGFβ1 and EGF and inhibits migration, proliferation, and tube formation by ECs.
Suramin346 Suture induced S.C. Heparin analog; Decreases expression of VEGF, PDGF and bFGF.
FND347 Suture induced FND FND destroys not only blood but also lymphatic vessels, thereby promotes corneal high-risk graft survival.
PDT and verteporfin348 Suture induced PDT after I.V. injection
of verteporfin
Corneal PDT after i.v. verteporfin injection time-dependently regresses mature corneal BV and LV and promotes allograft survival after subsequent high-risk corneal transplantation.
UVA crosslinking and riboflavin308 Suture induced Topical riboflavin and then UVA light exposure Corneal crosslinking with UVA light and riboflavin regressed both preexisting blood and lymphatic vessels significantly via induction of apoptosis in vascular endothelial cells. In addition, macrophages and CD45+ cell counts were significantly reduced.

- ref = reference number, S.C. =subconjunctival, EC = endothelial cell, I.P. = intraperitoneal, I.V. = intravenous, BIGH3 = TGF-β1-inducible gene-h3, SDF = stromal-cell derived factor, LHT7 = Low-molecular-weight heparin (LMWH)–taurocholate derivative, HGF= hepatocyte growth factor, FND= fine-needle diathermy, PDT= photodynamic therapy, UVA= ultraviolet light A.

*

Increases CNV in early stages and decreases in later stages.

ANTI-INFLAMMATORY AND IMMUNOSUPPRESSIVE AGENTS

Inflammation is a key factor in the process of corneal angiogenesis in most pathologic conditions.170 Inflammatory cells that infiltrate the injured cornea are the major source of potent angiogenic mediators such as IL-1, TNF-α, and VEGF.8,34 Also, products of inflammatory reactions, such as prostaglandins, have angiogenic properties.171

Corticosteroids are potent inhibitors of inflammation and have been used for their anti-inflammatory and anti-angiogenic properties in CNV.172 Despite their side effects, corticosteroids are one of the most potent medications clinically available to control inflammation and consequently CNV. Thus, they can be considered first-line treatment for CNV, especially in the setting of inflammation. Corticosteroids vary in their potency and routes of administration; commonly available corticosteroids include difluprednate, fluorometholone, rimexolone, loteprednol, prednisolone, dexamethasone, triamcinolone, and fluocinolone. While these conventional corticosteroids often have undesirable ocular side effects, newer agents such as anecortave acetate have been reported to be equally effective yet safer.173 We recommend aggressively treating inflammatory conditions associated with CNV such as HSV immune stromal keratitis and keratoplasty rejection with frequent potent corticosteroids (i.e. difluprednate every 1 hour). The topical corticosteroids are then tapered very slowly based on clinical response (over the course of months in the case of HSV immune stromal keratitis). It is essential to closely monitor the patients on long-term corticosteroids for early diagnosis and management of potential adverse effects.174

NSAIDs are also anti-inflammatory agents with known anti-angiogenesis properties. Both cyclooxygenase 1 (COX-1) and COX-2 are found in the cornea and have a role in angiogenesis. Indomethacin and ketoprofen are non-selective COX-1 and COX-2 inhibitors that have been found to significantly suppress bFGF and VEGF-induced angiogenesis in the mouse cornea.175 Similar to corticosteroids, long-term use of topical NSIADs is limited by potential corneal side effects which necessitates close monitoring.176 We do not use NSAIDs for CNV and to the best of our knowledge, there is no clinical study evaluating the efficacy of NSAID to prevent or management of CNV.

Cyclosporine is an immunomodulatory agent which is used for ocular surface immune disorders. Systemic cyclosporine A (CsA) can inhibit migration of primary endothelial cells and angiogenesis induced by VEGF.177 In a rabbit model, the efficacy of topical CsA 0.05% was better than bevacizumab but less than dexamethasone for the treatment of immune-mediated CNV.178 However, in a second study, a high dose subconjunctival CsA implant did not have a significant effect on CNV in human corneal transplants.179

Tacrolimus is another potent immunosuppressant which effectively blocks production of cytokines by T-cells and immunoglobulins by B-cells. Systemic tacrolimus has also been recommended.180, 181 Turgut and colleagues reported that both systemic and topical administration of tacrolimus are effective in prevention of CNV in an experimental model.182 Besides inhibiting VEGF production, tacrolimus also decreases the production of a number of angiogenic factors, such as FGF, epidermal growth factor (EGF), histamine, PDGF, prostaglandin E2 (PGE2), TNF-α, MMP-9 and MMP-13, IL-1, IL-6, and HIF.182

Sirolimus (rapamycin) and its derivative everolimus inhibit T-cell activity by preventing cell cycle progression from G1 to S phase, thereby blocking proliferation. In addition, rapamycin can promote T-cell anergy independently of the inhibition of proliferation even in the presence of T-cell receptor activation and co-stimulation by CD28 and IL-2.183, 184 Systemic and topical administration of sirolimus has been associated with a significant decrease of corneal opacity and vascularization as well as decreased corneal IL-6 and TGF-β 1 levels after alkali ocular injury in mice.185 Moreover, topical everolimus significantly decreased CNV induced with silver nitrate in rats, by decreasing the expression of VEGFR-2 and ERK 1/2.186

Tocilizumab, an IL-6 receptor antagonist, was effective in reducing CNV in animal models by decreasing corneal inflammation and VEGF expression.187 Subconjunctival tocilizumab (2.5 mg) was proven as effective as the same dose of subconjunctival bevacizumab in reducing CNV area and VEGF levels.188

TNF-α inhibitors have been used in experimental studies for the treatment of CNV because of their simultaneous anti-inflammatory and antiangiogenic activities.189 Infliximab is an anti-TNF-α monoclonal antibody which can bind both the monomeric and active trimeric form of TNF-α, thus blocking its activity. Topical infliximab was effective in reducing CNV and was correlated with TNF-α and VEGF activity.190 It can also prevent corneal lymphangiogenesis and conjunctivalization after alkali-induced injury.34, 191 In another study, a single intraperitoneal injection of infliximab 15 minutes after a corneal alkali burn was associated with markedly reduced CNV.192 Finally, etanercept is a recombinant TNF receptor which neutralizes both TNF-α and TNF-β. Subconjunctival etanercept had both anti-inflammatory and antiangiogenic effects in an animal model, which were enhanced when administered in combination with subconjunctival bevacizumab.193

ANTI-VEGF AGENTS

Several anti-VEGF agents such as bevacizumab have been primarily approved for use in multiple cancers for their antiangiogenic properties and are being used off-label for the treatment of ocular angiogenesis, including CNV. However, ranibizumab was primarily approved the FDA in 2006 for wet age-related macular degeneration. These agents are most effective for the treatment of actively growing vessels and have limited efficacy in well-established CNV.194, 195

Topical bevacizumab has been successfully used for the reduction of CNV in cases unresponsive to conventional anti-inflammatory medications in animal studies and clinical trials.196 Topical bevacizumab (commonly 5 mg/ml-five times/ day) has demonstrated a beneficial effect by decreasing the affected area of vascularization.197 Although topical bevacizumab has poor penetration into the cornea when the epithelium is intact, it penetrates a vascularized cornea well.198 This same study showed that subconjunctival bevacizumab penetrates the stroma even with an intact epithelium198 and has been used to treat CNV in a human case series as well as its associated lipid keratopathy.199 However, following chemical injury in rats, topical bevacizumab demonstrated longer standing anti-angiogenic effects than subconjunctival bevacizumab.200 A separate study has shown that lower doses of bevacizumab are required for subconjunctival injections compared to topical administration for equal efficacy.201 In addition, results of a meta-analysis on seven clinical and 18 experimental studies revealed a significant reduction of CNV after treatment with either topical or subconjunctival bevacizumab.202 However, CNV may recur following successful management with subconjunctival bevacizumab necessitating repeated injections, especially in cases with lipid deposition.203

Ranibizumab, another monoclonal antibody against VEGF-A, has also been used to reduce CNV. In an animal model, early subconjunctival administration of ranibizumab inhibited alkali-induced corneal vascularization. It not only significantly reduced VEGF levels in the cornea and bulbar conjunctiva, but also in the aqueous humor and the iris.204 In another experimental model, both topical and subconjunctival administration of ranibizumab were associated with significant improvement of CNV.205 In one clinical study, topical ranibizumab effectively reduced vessel caliber but not invasion area.206 There is also a report of successful management of CNV due to HSV keratitis with subconjunctival and intrastromal injection of ranibizumab in a case refractory to bevacizumab.207 However, in a different clinical study, subconjunctival ranibizumab proved to be less effective than bevacizumab in reducing CNV.208210

FD006 is a novel anti-VEGF-A monoclonal antibody with strong antiangiogenic activity. It is a full-length IgG antibody (similar to bevacizumab), obtained by using antibody phage display technology.211 Subconjunctival injection of FD006 significantly decreased the expression of VEGF, VEGFR-1, VEGFR-2, ICAM-1, and MMP-9 in alkali burned rat corneas. FD006 was found to be slightly superior to subconjunctival bevacizumab in reducing CNV in animal models.211 This might be due to higher affinity and a slower dissociation rate of FD006 compared to bevacizumab.

Pegaptanib has been approved by the food and drug administration (FDA) for treatment of choroidal neovascularization associated with AMD, but there is little evidence for its use in CNV. Akar and colleagues compared the efficacy of subconjunctival injection of bevacizumab, ranibizumab, and pegaptanib in the treatment of CNV in an animal model and found significant improvement of CNV in all treated groups. The outcome was best in the bevacizumab group followed by pegaptanib and ranibizumab.210

Aflibercept (VEGF TrapR1R2) has strong anti-VEGF activity and is a soluble fusion protein with binding domains for both VEGFR-1 and 2. It was approved by the FDA in 2011 for intravitreal injection in treatment of diabetic macular edema. Systemic administration of VEGF TrapR1R2 inhibited bFGF induced CNV.212 More recently, topical use of the 0.1% and 0.01% concentrations of aflibercept was associated with a significant decrease in CNV and VEGF expression in suture induced CNV in rabbits.213 In addition, in another experimental study, subconjunctival injection of Aflibercept at the time of corneal grafting was associated with significant reduction of donor vascularization and improvement of graft survival in a high-risk graft murine model.214

So far, the only topical antiangiogenic agent tested in phase II and III trials is aganirsen. It is an antisense oligonucleotide that inhibits CNV via preventing insulin receptor substrate-1 (IRS-1) expression as well as downregulating expression of VEGF and IL-1 β.215 Administration of topical aganirsen eye drops (86 μg/day/eye) in patients with keratitis-related progressive CNV led to significant amelioration of CNV and reduced need for corneal transplantation.158

There are some concerns about the safety of VEGF neutralization at the ocular surface. Delayed epithelial wound healing and increased expression of MMPs have been reported with topical bevacizumab in a rat model of epithelial injury.216 Furthermore, topical administration of bevacizumab was associated with increased risk of persistent epithelial defects (PEDs) that was dependent on the dose and duration of treatment.217, 218 Most clinical studies have reported no serious side effects with subconjunctival bevacizumab. However, a few complications have been reported such as subconjunctival hemorrhage and PEDs.219

Overall, anti-VEGF agents have provided a new therapeutic approach for the clinical management of CNV. As expected based on the biology of angiogenesis, these agents are most effect in the setting of active CNV with “immature” vessels. As clinical experience has shown, their efficacy is limited in longstanding CNV where the vessels have matured and are less dependent on VEGF signaling.209

MMP INHIBITORS

Tetracyclines, such as doxycycline, are potent inhibitors of collagenase activity and MMP-induced extracellular matrix degradation. Doxycycline is also known for its anti-neovascular effects in the cornea.220 Topical preparations of doxycycline including a 2% neutralized solution, temperature-sensitive hydrogel, and, more recently, eye drops have been used successfully for inhibiting CNV in animal and human studies.221, 222 Doxycycline inhibits angiogenesis by modulation of the PI3K/Akt-eNOS pathway in an MMP-independent mechanism.223 Furthermore, it was shown to enhance the anti-VEGF effects of bevacizumab by decreasing the expression of VEGF and its receptors.224

Minocycline is a semisynthetic tetracycline with anti-collagenase and anti-angiogenic properties. In an animal model of alkali ocular injury, systemic minocycline was effective in promoting epithelial healing and decreasing CNV. In this study, corneal VEGF, VEGFR-1, VEGFR-2, bFGF, IL-1β, IL-6, MMP-2, MMP-9, and MMP-13 levels were significantly lower in the treated group.225

Tigecycline, another member of the tetracycline family, has also been used as a topical and subconjunctival injection for inhibiting CNV in a rat chemical burn model.167 Results were promising for both routes, although subconjunctival injection was more effective than topical administration.167

MULTIKINASE INHIBITORS

Sunitinib is a multi-targeted receptor tyrosine kinase (RTK) inhibitor (TKI) which selectively and potently inhibits four RTKs involved in angiogenesis including VEGFR-2, PDGFR-β, FGFR-1, and EGFR.226 It has been approved by the FDA for treatment of metastatic tumors because of its remarkable antiangiogenic activity.227 Oral administration of sunitinib in inflammation-induced CNV in mice was effective in reducing angiogenesis, most likely by blocking the VEGF-A/VEGFR-2 pathway.228 Topical application of 0.5 mg/ml sunitinib was also associated with a significant decrease of VEGFR-2 levels and CNV in rats.186 Topical sunitimib is reported to be 3-fold more effective than bevacizumab in the treatment of CNV due to its inhibitory effect on both VEGF and PDGF pathways.229 Furthermore, greater results were obtained by topical administration as compared with subconjunctival injection.230 However, the safe dose of the topical sunitinib remains to be determined due to the possibility of epithelial cytotoxicity that was seen at concentrations greater than 3.3 μg/ml in an in vitro study.231

Pazopanib is a small TKI active against VEGF and PDGF that is approved for use in renal cell and soft tissue carcinoma. The safety and efficacy of a topical preparation of pazopanib in treatment of CNV due to different underlying etiologies were studied in phase I and II clinical trials and showed promising results.232

Sorafenib is another multikinase inhibitor which blocks both VEGFR2 and PDGFR.233 Its short-term oral administration was effective in reducing experimental CNV most likely via inhibition of ERK and VEGFR-2 phosphorylation.234 However, it was more toxic to corneal epithelial cells in vitro compared to sunitinib.231 Similarly, regorafenib inhibits multiple RTKs including VEGFR-1, −2 and −3; PDGFR-β; and FGFR.235 Topical regorafenib (1 mg/ml) decreased epithelial and endothelial VEGF levels as well as the percentage of CNV area in an alkali-burn CNV model.236

Lapatinib blocks 2 RTKs, including human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR), and it is a part of the treatment for early stages of HER2-positive breast cancer.237 Oral administration of lapatinib effectively reduced CNV by decreasing corneal epithelial and stromal VEGF expression in an animal model. It was more effective than trastuzumab, which only blocks HER2.238

ROCK INHIBITORS

Fasudil hydrochloride is a potent inhibitor of the Rho/ROCK pathway and has demonstrated antiangiogenic properties. In vitro studies showed that fasudil attenuated the Rho/ROCK pathway and VEGF-dependent phosphorylation of extracellular signal regulated kinase 1/2 (ERK1/2) and Akt.140 In alkali burn-induced CNV mice models, administration of topical fasudil (100 μM) was associated with decreased expressions of VEGF, TNF-α, MMP-8, and MMP-9 as well as decreased incidence of CNV in treated mice. It also caused decreased inflammatory cell infiltration (especially PMNs) and reactive oxygen species production.239

AMA0526 is a newer, more specific, locally acting ROCK inhibitor with minimal systemic side effects.240 In vitro studies demonstrated significant inhibition of vascular endothelial proliferation and migration. In animal models, its topical application was comparable to bevacizumab and dexamethasone in regards to preventing corneal vascularization and scarring, respectively.241

SURGICAL OPTIONS

CORNEAL VESSEL OCCLUSION

Several methods have been described for corneal vessel occlusion including conjunctival resection, cryotherapy, laser thermal cauterization, fine needle diathermy (FND), electrolysis-needle cauterization, and photodynamic therapy (PDT). Although these methods are most effective in the treatment of established CNV, they can also be applied to active vascularization in adjunction with anti-VEGF agents.242

Laser thermal cauterization using Argon, yellow dye, and Nd:YAG lasers have been used to ablate CNV in experimental243245 and clinical246248 studies. Histopathologic studies revealed disruption and degeneration of endothelial cell and clot formation in occluded vessels as well as intracorneal hemorrhage followed by increased cellularity and distortion of corneal lamella by red blood cells in treated corneas.243, 244 In a recent study, frequency-doubled Nd:YAG laser (532 nm) was used to treat 40 eyes with quiescent CNV.248 After 3 months, complete occlusion was observed in 53% of vessels, while 37% of vessels were re-canalized.248 Complications include corneal hemorrhage, corneal thinning, vascularization exacerbation and vessel lumen reopening.248250

FND was successful in treating lipid keratopathy associated with CNV in more than 80% of cases in a study.251 Additionally, more than 80% of grafts survived longer than one year in vascularized corneas that were pre-treated with FND. The authors concluded FND can be considered an effective, low-cost, easy, and safe method for treatment of established CNV.251 The long-term efficacy and visual acuity improvement of FND in treatment of HSK-associated CNV persisted for a mean 18.9 months after initial treatment.252 Since FND upregulates VEGFs, it has been suggested that FND should be combined with topical anti-VEGFs to reduce recurrence rates.253

PDT with verteporfin is another approach for the selective occlusion of abnormal, leaky CNV vessels. This procedure creates a highly specific tissue damage that seals off the vessel. A case series showed evidence of vascular thrombosis and decreased CNV in about two thirds of eyes that underwent PDT.254 Resolution of bilateral CNV and associated lipid keratopathy after PDT has also been reported.255 PDT has limited clinical use due to high costs and potential complications related to laser irradiation and generation of reactive oxygen species.

OCULAR SURFACE RECONSTRUCTION

Amniotic membrane transplantation (AMT) has been shown to be effective in preventing inflammation and angiogenesis following ocular injuries. Temporary or permanent transplantation of human amniotic membrane (HAM) in acute chemical injury promotes re-epithelialization and reduces both inflammation and vascularization.256 Chondrocyte-derived extracellular matrix (CDECM) has been introduced as an alternative to amniotic membrane in ocular surface reconstruction because of its potent anti-fibrotic and antiangiogenic effects.257 Transplantation of CDECM in the acute phase of alkali injury of the cornea resulted in significant reduction of CNV and opacity in a rabbit model.257

Autologous or allogenic ocular surface stem cell transplantation (OSST) is the current standard method of managing unilateral or bilateral total LSCD. However, less invasive procedures might be used for CNV and/or conjunctivalization associated with partial LSCD. Sequential sector conjunctival epitheliectomy (SSCE) with or without AMT has been used successfully for management of CNV in eyes with partial LSCD.258,259

To treat total LSCD, limbal stem cells can be harvested from autologous or nonautologous sources. A conjunctival limbal autograft (CLAU) taken from the healthy fellow eye is considered the most effective surgical procedure in patients with total unilateral LSCD. It produces excellent results, often with complete regression of corneal neovascularization.260262 Cultivated limbal epithelial transplantation (CLET) is a suitable alternative in cases of total unilateral LSCD or in cases of bilateral LSCD when the damage is more severe in one eye.263265 Living-related conjunctival limbal allograft (lr-CLAL), keratolimbal allograft (KLAL), or combined procedures (i.e. Cincinnati procedure) are surgical alternatives in patients with bilateral LSCD.266268 A method that provides fresh tissue from a patient’s first-degree blood relative, lr-CLAL utilizes tissue from one eye (or occasionally both eyes) of the best available HLA-matched donor. Lr-CLAL also has the advantage of providing viable conjunctival tissue, which may be used in patients with severe conjunctival deficiency. In comparison, KLAL utilizes cadaveric tissue, is more accessible, and offers more stem cells because two corneoscleral rims are used.269 Simple limbal epithelial transplantation (SLET), cultivated oral mucosal epithelial transplantation (COMET), or allogeneic CLET are other surgical options for total LSCD.270272

Typically, superficial corneal vessels regress after successful OSST, indicating successful grafting. However, re-growth of these vessels may be a marker of graft failure.273 Moreover, deeper vessels may not disappear with OSST only, so concomitant treatment modalities including keratoplasty may be required.274

STEM CELL-BASED THERAPY

In the last decade, there has been intense focus on stem cells for regenerative therapies. Many sources of stem cells including bone marrow, fat, umbilical cord, dental pulp, hair follicle, and induced pluripotent stem cells have shown promising results in experimental models of ocular surface injury.275, 276 Among these, mesenchymal stem cells (MSCs) hold the most promise for clinical application. MSCs are found in most adult tissues, including the limbus, and play animportant role in tissue repair and maintenance. A number of clinical trials aimedat evaluating the safety and efficacy of MSCs in the promotion of cardiac tissue regeneration, modulation of systemic immune diseases, and healing of soft tissue injuries are currently underway.277, 278 In animal models of chemical injury, MSCs have been shown to accelerate corneal wound healing, attenuate inflammation, and modulate corneal neovascularization.279284 These effects have been shown to be mediated in part through secreted factors such as TSG-6.285 Furthermore, in a recent study we showed cornea-derived MSCs (cMSCs) are directly antiangiogenic mainly through sflt-1 and PEDF.286 Further study revealed cMSCs can change the immunophenotype and angiogenic function of the macrophages mainly through cMSC-derived PEDF.287 The so-called cMSC-educated macrophages expressed significantly higher levels of anti-inflammatory and anti-angiogenic factors compared with control macrophages, both in vitro and in vivo.287 Overall, stem cell-based therapies are likely to replace tissue transplantation in the future for certain ocular surface diseases that lead to CNV.

FUTURE DIRECTION

Although several novel agents have been used successfully to treat CNV in animal models and clinical studies, the need for an FDA-approved potent topical antiangiogenic agent is still unmet.288290 The only topical antiagiogenic agent that has made it to the phase III trial is aganirsen, an example of small interfering RNA (siRNA).158, 215 Agents/interventions which are under investigation for possible application in treatment of CNV have been listed in table 1 for experimental and table 2 for clinical studies.

Table 2.

Interventions for treatment of CNV investigated or under investigation in clinical studies.

Intervention Study design Result Ref./ID
Bevacizumab (T) Single group Reduced corneal NV within the first month. Increased risk of epithelial defect in the second month. Kim, 2008349
Single group Reduced CNV in nearly two thirds of the eyes treated. Ocular side effects including eyelid swelling/chalazion and superficial punctate keratitis. Waisbourd, 2013350
Single group Significant reduction in mean neovascular area (47%) and vessel caliber (54%) Dastjerdi, 2009153
Single group Significant reduction in mean neovascular area (47%) and vessel caliber (36%) Cheng, 2012196
Bevacizumab (S.C.) RCT Regression of recent-onset CNV in all treated eyes. Petsoglou, 2013219
Single group Regression of CNV in all eyes at 1 week continued to decrease for 1 month. Benayoun, 2012351
RCT Ongoing NCT00555594
Bevacizumab (I.S.) Single group Complete regression of CNV in 16/25 eyes and reduced opacity and improved visual acuity in 5/25 eyes. Sarah, 2016352
Bevacizumab, (S.C. vs. T) RCT Reduced recurrence of CNV and increased graft survival in cases of high-risk corneal transplants. Less effective when used topically. Bhatti, 2013353
Bevacizumab (S.C. and I.S.) Single group Significant regression of CNV in all eyes. Yeung, 201 1354
Bevacizumab (S.C. followed by T) RCT Ongoing NCT01072357
Ranibizumab (T) Single group Significant decrease in neovascular area lasted through 16 weeks. Ferrari, 2013206
Bevacizumab vs. Ranibizumab (S.C. and I.S.) Comparative interventiona case series CNV regressed in both group. Bevacizumab had more effective and stable regression of CNV. Kim, 2013208
Doxycycline (T) Single group Vessels disappeared or attenuated, shortened and less dense in five of six patients. Jovanovic, 2014222
Topical heparin and steroid* Single group All eyes showed complete regression of CNV within 5 months. Michels, 2012355
Topical Pazopanib Single group Neovascular area, invasion area and vessel caliber significantly decreased. Amparo, 2013232
Aganirsen RCT Significantly reduced the relative corneal neovascular area. Decreased the need for corneal transplantation. Cursiefen, 2014158
Topical IL-1R antagonist RCT, crossover assignment Ongoing NCT00915590
FND Single group Complete regression of vessels in all patients. Reduced graft rejection in high-risk graft. Pillai, 2000356
FND Single group Reduced lipid deposition, prevented rejection episodes, reduced intraoperative bleeding Faraj, 2014251
Angiography guided FND Single group Decreased neovascular area. Corneal angiography enables selective treatment to the afferent vessels. Spiteri, 2015357
FND and I.S. Bevacizumab Single group 8/9 eyes had complete resolution of CNV and lipid deposition. Elbaz, 2015242
Fluorescein-potentiated argon laser Single group Decreased corneal edema, CNV, and lipid keratopathy. Gordon, 2002358
dihematoporphyrin ether (DHE) PDT Single group Immediate reduction in CNV in all patients. Three patients suffered significant systemic short-term phototoxicity reactions. Sheppard, 2006359
PDT with verteporfin Single group Significant decrease of corneal vessel length and CNV in 90% of cases. Verdiguel-Sotelo, 2010360
Single group Complete vascular occlusion in 42.4% and partial occlusion in 24.2% of eyes. Al-Torbak, 2012254
Single group 32% total occlusion, 60% partial occlusion and 8 % worsening. Significant decrease of mean neovascular area. Diaz-Davalos, 2016361
PDT and S.C. Bevacizumab Single group Complete regression in 62.5% and partial regression in 37.5% of eyes. Yoon, 2017305
Single group All eyes showed a significant decrease in CNV. After 1-year, complete and partial occlusion was achieved in 66.7% and 25.0% of eyes, respectively. You, 2011304
PDT with topical dihematoporphyrin derivative RCT Ongoing NCT00004430
Topical bevacizumab (Avastin™) Single group The mean reduction in vascularized area during treatment was 61%. The mean reduction in vessel diameter under topical Avastin™ therapy was 24%. Koenig, 2009197
FND and bevacizumab Single group The vascularized area was reduced significantly (P < 0.05). Combined subconjunctival and eye drop antivascular endothelial growth factor treatment was significantly more effective in reducing the vascularized area compared with antivascular endothelial growth factor eye drop therapy alone (P < 0.05). Koenig, 2012253

CinicalTrials.gov identifier

*

Remixolone or dexamethasone.

RCT= randomized clinical trial, S.C.= subconjunctival, T= topical, I.S.= intrastromal, FND= fine-needle diathermy, PDT= photodynamic therapy.

Gene-based antiangiogenic therapy is an emerging strategy that encompasses a variety of target genes and gene-delivery methods.291 Adeno-associated viral (AAV) vectors have been utilized to transfect corneal epithelial cells with antiangiogenic genes such as endostatin and angiostatin, which successfully reduced CNV in animal models.292, 293 AAV vectors have been reported to be an effective and safe mean for gene delivery to the cornea.294 Nanoparticles are effective, non-viral, non-toxic, and a sustainable form of gene delivery. Nanoparticle-based vectors containing an expression plasmid of sflt-1295 or small hairpin RNA (shRNA) against VEGF-A have been reported to regress CNV in experimental studies.296 Downregulating proangiogenic genes using gene-silencing techniques is an evolving alternative for inhibition of ocular angiogenesis. Besides the example of aganirsen (mentioned above), siRNA targeting VEGF has demonstrated inhibition of CNV in other animal models.156, 211, 297, 298 CRISPR-Cas9-mediated genome editing is a novel approach for manipulating the genome of living cells.299 This rapidly evolving technique has great potential with unlimited applicability.

Using combined strategies targeting different mechanisms might be more effective in treatment of CNV. Combination therapy with a vessel occlusion method (PDT), an anti-VEGF agent (subconjunctival bevacizumab), and a corticosteroid (triamcinolone acetonide) or immunosuppressive agent (cyclosporine A) have been shown to be effective in two cases with extensive superficial and deep CNV.300303 Combination of PDT and subconjunctival bevacizumab was associated with significant improvement of CNV refractory to conventional treatments in two case series.304, 305 Likewise, combination of FND and bevasizumab significantly reduced CNV prior to keratoplasty.253 In addition, combination of two or more anti-angiogenic agents with different mechanisms of action (e.g. anti-VEGFs, TKIs, immunomodulators, MMP inhibitors, etc.) might have additional effects compared to monotherapy.186, 193, 224, 306, 307 Recently, corneal crosslinking using ultraviolet A light (UVA) and topical riboflavin has shown promising results to regress both preexisting blood and lymphatic vessels.308 However, randomized clinical trials are needed to investigate the short­term and long-term safety and efficacy of these methods.

SUMMARY

CNV is a relatively common adverse consequence of severe ocular surface and corneal pathologies that can result in significant visual disability if not addressed promptly. Moreover, since it is an important risk factor for corneal graft rejection, its treatment before any keratoplasty procedure is recommended. Recent advances in elucidating the molecular pathways, clinical staging, anterior segment imaging, and therapeutic strategies have led to improved treatment strategies.

Studies show that the formation and invasion of vessels is the result of a complex mechanism which includes inflammation, hypoxia, corneal edema, innervation abnormalities, and LSCD. Although VEGF is the core molecule responsible for endothelial proliferation, other important mediators such as PDGF, bFGF, nitric oxide, MMPs, and proinflammatory cytokines have a critical role in the maturation, establishment, and invasion of vessels.

Semi-automated and automated analysis software which have been optimized for interpreting corneal photographs are useful tools for grading and classifying CNV, as well as evaluating interventions. Application of IVCM, AS-OCT, fluorescein, and ICG have led to increased understanding of the origin, nature, and course of vessels on the cornea, as well as improved targeted treatment and assessment of outcomes.

Currently, anti-inflammatory and anti-VEGF medications are the mainstay therapy of active CNV, while FND is used for ablation of established vessels. The advent of newer, more potent anti-angiogenic agents such as multi-kinase inhibitors, ROCK inhibitors, MMP inhibitors, and immunosuppressive medications may lead to improved treatment outcomes. However, targeting multiple angiogenic pathways may have a greater synergistic effect and should be considered the goal of future drug designs. The concomitant use of angiography guided thermo-ablation and photodynamic therapy may also increase the success rate of medical therapies which target CNV. In case of severe total LSCD with deep stromal CNV, the best strategy is an OSST followed by a keratoplasty. MSC-based cellular therapy and gene-based antiangiogenic therapy are novel promising alternatives.

Financial Disclosures:

The authors have no financial interest in the products discussed and did not receive funding for any of the work. EJH has consulted for Alcon Laboratories, Allergan, Bausch & Lomb, Katena, Kala Pharmaceuticals, Mati Pharmaceuticals, Omeros, PRN, TearLab, TearScience, Senju Pharmaceuticals, and Shire.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Lee P, Wang CC, Adamis AP. Ocular neovascularization: an epidemiologic review. Surv Ophthalmol. 1998;43(3):245–69. [DOI] [PubMed] [Google Scholar]
  • 2.Hamill CE, Bozorg S, Peggy Chang HY, et al. Corneal alkali burns: a review of the literature and proposed protocol for evaluation and treatment. Int Ophthalmol Clin.. 2013;53(4):185–94. [DOI] [PubMed] [Google Scholar]
  • 3.Lim P, Fuchsluger TA, Jurkunas UV. Limbal stem cell deficiency and corneal neovascularization. Seminars in ophthalmology: Informa UK Ltd UK, 2009; v. 24. [DOI] [PubMed] [Google Scholar]
  • 4.Azar DT. Comeal angiogenic privilege: angiogenic and antiangiogenic factors in corneal avascularity, vasculogenesis, and wound healing (an American Ophthalmological Society thesis). Trans Am Ophthalmol Soc. 2006;104:264–302. [PMC free article] [PubMed] [Google Scholar]
  • 5.Colby K, Adamis A. Prevalence of corneal neovascularization in a general eye service population. INVESTIGATIVE OPHTHALMOLOGY & VISUAL SCIENCE: LIPPINCOTT-RAVEN PUBL 227 EAST WASHINGTON SQ, PHILADELPHIA, PA 19106, 1996; v. 37. [Google Scholar]
  • 6.Bachmann B, Taylor RS, Cursiefen C. Corneal neovascularization as a risk factor for graft failure and rejection after keratoplasty: an evidence-based meta-analysis. Ophthalmology. 2010;117(7):1300–5. e7. [DOI] [PubMed] [Google Scholar]
  • 7.Faraj LA, Said DG, Dua HS. Evaluation of corneal neovascularisation. Br J Ophthalmol. 2011;95(10):1343–4. [DOI] [PubMed] [Google Scholar]
  • 8.Maruyama K, Ii M, Cursiefen C, et al. Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages. J Clin Invest. 2005;115(9):2363–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Risau W Mechanisms of angiogenesis. Nature. 1997;386(6626):671–4. [DOI] [PubMed] [Google Scholar]
  • 10.Ozerdem U, Alitalo K, Salven P, Li A. Contribution of bone marrow-derived pericyte precursor cells to corneal vasculogenesis. Invest Ophthalmol Vis Sci. 2005;46(10):3502–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Park PJ, Chang M, Garg N, et al. Corneal lymphangiogenesis in herpetic stromal keratitis. Surv Ophthalmol. 2015;60(1):60–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Abdelfattah NS, Amgad M, Zayed AA, et al. Molecular underpinnings of corneal angiogenesis: advances over the past decade. Int J Ophthalmol. 2016;9(5):768–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cogan DG. Vascularization of the cornea; ats experimental induction by small lesions and a new theory of its pathogenesis. Arch Ophthal. 1949;41(4):406–16. [PubMed] [Google Scholar]
  • 14.Maurice DM, Zauberman H, Michaelson IC. The stimulus to neovascularization in the cornea. Exp Eye Res. 1966;5(3):168–84. [DOI] [PubMed] [Google Scholar]
  • 15.Levene R, Shapiro A, Baum J. Experimental Corneal Vascularization. Arch Ophthalmol. 1963;70(2):242–9. [DOI] [PubMed] [Google Scholar]
  • 16.Nishida K, Kinoshita S, Ohashi Y, et al. Ocular surface abnormalities in aniridia. Am J Ophthalmol. 1995;120(3):368–75. [DOI] [PubMed] [Google Scholar]
  • 17.Vera LS, Gueudry J, Delcampe A, et al. In vivo confocal microscopic evaluation of corneal changes in chronic Stevens-Johnson syndrome and toxic epidermal necrolysis. Cornea. 2009;28(4):401–7. [DOI] [PubMed] [Google Scholar]
  • 18.Ruff AL, Jarecke AJ, Hilber DJ, et al. Development of a mouse model for sulfur mustard-induced ocular injury and long-term clinical analysis of injury progression. Cutan Ocul Toxicol. 2013;32(2):140–9. [DOI] [PubMed] [Google Scholar]
  • 19.Friedenwald JS. Growth pressure and metaplasia of conjunctival and corneal epithelium. Doc Ophthalmol. 1951;5–6(1):184–92. [DOI] [PubMed] [Google Scholar]
  • 20.Tobaigy FM, Azar DT. Hemilimbal Deficiency Model of Corneal Neovascularization: Possible Invalidity of the Limbal Barrier Concept. Saudi Journal of Ophthalmology. 2009;23(1). [Google Scholar]
  • 21.Eliason JA, Elliott JP. Proliferation of vascular endothelial cells stimulated in vitro by corneal epithelium. Invest Ophthalmol Vis Sci. 1987;28(12):1963–9. [PubMed] [Google Scholar]
  • 22.Van Setten GB. Vascular endothelial growth factor (VEGF) in normal human corneal epithelium: detection and physiological importance. Acta Ophthalmol Scand. 1997;75(6):649–52. [DOI] [PubMed] [Google Scholar]
  • 23.Ma DH, Tsai RJ, Chu WK, et al. Inhibition of vascular endothelial cell morphogenesis in cultures by limbal epithelial cells. Invest Ophthalmol Vis Sci. 1999;40(8):1822–8. [PubMed] [Google Scholar]
  • 24.Sejpal K, Ali MH, Maddileti S, et al. Cultivated limbal epithelial transplantation in children with ocular surface burns. JAMA Ophthalmol. 2013;131(6):731–6. [DOI] [PubMed] [Google Scholar]
  • 25.Adamis AP, Meklir B, Joyce NC. In situ injury-induced release of basic-fibroblast growth factor from corneal epithelial cells. Am J Pathol. 1991;139(5):961–7. [PMC free article] [PubMed] [Google Scholar]
  • 26.Ambati BK, Nozaki M, Singh N, et al. Corneal avascularity is due to soluble VEGF receptor-1. Nature. 2006;443(7114):993–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ambati BK, Patterson E, Jani P, et al. Soluble vascular endothelial growth factor receptor-1 contributes to the corneal antiangiogenic barrier. Br J Ophthalmol. 2007;91(4):505–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Makino Y, Cao R, Svensson K, et al. Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature. 2001;414(6863):550–4. [DOI] [PubMed] [Google Scholar]
  • 29.Cursiefen C, Chen L, Saint-Geniez M, et al. Nonvascular VEGF receptor 3 expression by corneal epithelium maintains avascularity and vision. Proc Natl Acad Sci U S A. 2006;103(30): 11405–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kenney MC, Chwa M, Alba A, et al. Localization of TIMP-1, TIMP-2, TIMP-3, gelatinase A and gelatinase B in pathological human corneas. Curr Eye Res. 1998;17(3):238–46. [DOI] [PubMed] [Google Scholar]
  • 31.Lin HC, Chang JH, Jain S, et al. Matrilysin cleavage of corneal collagen type XVIII NC1 domain and generation of a 28-kDa fragment. Invest Ophthalmol Vis Sci. 2001;42(11):2517–24. [PubMed] [Google Scholar]
  • 32.Sheibani N, Sorenson CM, Cornelius LA, Frazier WA. Thrombospondin-1, a natural inhibitor of angiogenesis, is present in vitreous and aqueous humor and is modulated by hyperglycemia. Biochem Biophys Res Commun. 2000;267(1):257–61. [DOI] [PubMed] [Google Scholar]
  • 33.Bock F, Onderka J, Braun G, et al. Identification of Novel Endogenous Anti(lymph)angiogenic Factors in the Aqueous Humor. Invest Ophthalmol Vis Sci. 2016;57(15):6554–60. [DOI] [PubMed] [Google Scholar]
  • 34.Bock F, Maruyama K, Regenfuss B, et al. Novel anti(lymph)angiogenic treatment strategies for corneal and ocular surface diseases. Prog Retin Eye Res. 2013;34:89–124. [DOI] [PubMed] [Google Scholar]
  • 35.Ma DH, Chen JK, Zhang F, et al. Regulation of corneal angiogenesis in limbal stem cell deficiency. Prog Retin Eye Res. 2006;25(6):563–90. [DOI] [PubMed] [Google Scholar]
  • 36.Albuquerque RJ, Hayashi T, Cho WG, et al. Alternatively spliced vascular endothelial growth factor receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth. Nat Med. 2009;15(9):1023–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Regenfuss B, Dreisow ML, Hos D, et al. The Naive Murine Cornea as a Model System to Identify Novel Endogenous Regulators of Lymphangiogenesis: TRAIL and rtPA. Lymphat Res Biol. 2015;13(2):76–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cursiefen C, Maruyama K, Bock F, et al. Thrombospondin 1 inhibits inflammatory lymphangiogenesis by CD36 ligation on monocytes. J Exp Med. 2011;208(5):1083–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Cursiefen C, Chen L, Dana MR, Streilein JW. Corneal lymphangiogenesis: evidence, mechanisms, and implications for corneal transplant immunology. Cornea. 2003;22(3):273–81. [DOI] [PubMed] [Google Scholar]
  • 40.Chung ES, Saban DR, Chauhan SK, Dana R. Regulation of blood vessel versus lymphatic vessel growth in the cornea. Invest Ophthalmol Vis Sci. 2009;50(4):1613–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cursiefen C, Schlotzer-Schrehardt U, Kuchle M, et al. Lymphatic vessels in vascularized human corneas: immunohistochemical investigation using LYVE-1 and podoplanin. Invest Ophthalmol Vis Sci. 2002;43(7):2127–35. [PubMed] [Google Scholar]
  • 42.Hos D, Bukowiecki A, Horstmann J, et al. Transient Ingrowth of Lymphatic Vessels into the Physiologically Avascular Cornea Regulates Corneal Edema and Transparency. Sci Rep. 2017;7(1):7227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Dana MR. Angiogenesis and lymphangiogenesis-implications for corneal immunity. Semin Ophthalmol. 2006;21(1):19–22. [DOI] [PubMed] [Google Scholar]
  • 44.Chen L, Hamrah P, Cursiefen C, et al. Vascular endothelial growth factor receptor-3 mediates induction of corneal alloimmunity. Nat Med. 2004;10(8):813–5. [DOI] [PubMed] [Google Scholar]
  • 45.Regenfuss B, Bock F, Cursiefen C. Corneal angiogenesis and lymphangiogenesis. Curr Opin Allergy Clin Immunol. 2012;12(5):548–54. [DOI] [PubMed] [Google Scholar]
  • 46.Cursiefen C, Cao J, Chen L, et al. Inhibition of hemangiogenesis and lymphangiogenesis after normal-risk corneal transplantation by neutralizing VEGF promotes graft survival. Invest Ophthalmol Vis Sci. 2004;45(8):2666–73. [DOI] [PubMed] [Google Scholar]
  • 47.Patel SP, Dana R. Corneal lymphangiogenesis: implications in immunity. Semin Ophthalmol. 2009;24(3):135–8. [DOI] [PubMed] [Google Scholar]
  • 48.Dietrich T, Bock F, Yuen D, et al. Cutting edge: lymphatic vessels, not blood vessels, primarily mediate immune rejections after transplantation. J Immunol. 2010;184(2):535–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shi W, Liu J, Li M, et al. Expression of MMP, HPSE, and FAP in stroma promoted corneal neovascularization induced by different etiological factors. Curr Eye Res. 2010;35(11):967–77. [DOI] [PubMed] [Google Scholar]
  • 50.Cursiefen C, Chen L, Borges LP, et al. VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment. J Clin Invest. 2004; 113(7): 1040–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chang LK, Garcia-Cardena G, Farnebo F, et al. Dose-dependent response of FGF-2 for lymphangiogenesis. Proc Natl Acad Sci US A. 2004;101(32): 11658–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gothard TW, Hardten DR, Lane SS, et al. Clinical findings in Brown-McLean syndrome. Am J Ophthalmol. 1993;115(6):729–37. [DOI] [PubMed] [Google Scholar]
  • 53.Kenyon BM, Voest EE, Chen CC, et al. A model of angiogenesis in the mouse cornea. Invest Ophthalmol Vis Sci. 1996;37(8):1625–32. [PubMed] [Google Scholar]
  • 54.Abdelfattah NS, Amgad M, Zayed AA, et al. Clinical correlates of common corneal neovascular diseases: a literature review. Int J Ophthalmol. 2015;8(1):182–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Amano S, Rohan R, Kuroki M, et al. Requirement for vascular endothelial growth factor in wound-and inflammation-related corneal neovascularization. Investigative Ophthalmology and Visual Science. 1998;39(1):18–22. [PubMed] [Google Scholar]
  • 56.Sunderkotter C, Steinbrink K, Goebeler M, et al. Macrophages and angiogenesis. J Leukoc Biol. 1994;55(3):410–22. [DOI] [PubMed] [Google Scholar]
  • 57.Kvanta A, Sarman S, Fagerholm P, et al. Expression of matrix metalloproteinase-2 (MMP-2) and vascular endothelial growth factor (VEGF) in inflammation-associated corneal neovascularization. Exp Eye Res. 2000;70(4):419–28. [DOI] [PubMed] [Google Scholar]
  • 58.Mantovani A, Bussolino F, Dejana E. Cytokine regulation of endothelial cell function. FASEB J. 1992;6(8):2591–9. [DOI] [PubMed] [Google Scholar]
  • 59.Hayashi T, Matsuoka K, Saitoh M, et al. Influence of alpha-tumor necrosis factor and beta-interleukin-1 on production of angiogenetic factors and thymidine phosphorylase activity in immortalized human decidual fibroblasts in vitro. J Obstet Gynaecol Res. 2006;32(1):15–22. [DOI] [PubMed] [Google Scholar]
  • 60.Vinores S, Xiao W, Zimmerman R, et al. Upregulation of vascular endothelial growth factor (VEGF) in the retinas of transgenic mice overexpressing interleukin-1ß (IL-1ß) in the lens and mice undergoing retinal degeneration. Histology and histopathology. 2003;18(3):797–810. [DOI] [PubMed] [Google Scholar]
  • 61.Biswas PS, Banerjee K, Kinchington PR, Rouse BT. Involvement of IL-6 in the paracrine production of VEGF in ocular HSV-1 infection. Exp Eye Res. 2006;82(1):46–54. [DOI] [PubMed] [Google Scholar]
  • 62.Yoshida S, Ono M, Shono T, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol. 1997;17(7):4015–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Nakagawa T, Li JH, Garcia G, et al. TGF-β induces proangiogenic and antiangiogenic factorsvia parallel but distinct Smad pathways1. Kidney international. 2004;66(2):605–13. [DOI] [PubMed] [Google Scholar]
  • 64.Nakao S, Kuwano T, Tsutsumi-Miyahara C, et al. Infiltration of COX-2–expressing macrophages is a prerequisite for IL-1β–induced neovascularization and tumor growth. Journal of Clinical Investigation. 2005; 115(11):2979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Strieter R, Kunkel S, Elner V, et al. Interleukin-8. A corneal factor that induces neovascularization. The American journal of pathology. 1992;141(6):1279. [PMC free article] [PubMed] [Google Scholar]
  • 66.Sotozono C, He J, Matsumoto Y, et al. Cytokine expression in the alkali-burned cornea. Curr Eye Res. 1997;16(7):670–6. [DOI] [PubMed] [Google Scholar]
  • 67.Salven P, Hattori K, Heissig B, Rafii S. Interleukin-1alpha promotes angiogenesis in vivo via VEGFR-2 pathway by inducing inflammatory cell VEGF synthesis and secretion. FASEB J. 2002;16(11):1471–3. [DOI] [PubMed] [Google Scholar]
  • 68.Narayanan S, Glasser A, Hu YS, McDermott AM. The effect of interleukin-1 on cytokine gene expression by human corneal epithelial cells. Exp Eye Res. 2005;80(2):175–83. [DOI] [PubMed] [Google Scholar]
  • 69.Suryawanshi A, Veiga-Parga T, Reddy PB, et al. IL-17A differentially regulates corneal vascular endothelial growth factor (VEGF)-A and soluble VEGF receptor 1 expression and promotes corneal angiogenesis after herpes simplex virus infection. The Journal of Immunology. 2012;188(7):3434–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li W, He H, Kuo CL, et al. Basement membrane dissolution and reassembly by limbal corneal epithelial cells expanded on amniotic membrane. Invest Ophthalmol Vis Sci. 2006;47(6):2381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Raffetto JD, Khalil RA. Matrix metalloproteinases and their inhibitors in vascular remodeling and vascular disease. Biochemical pharmacology 2008;75(2):346–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ferreras M, Felbor U, Lenhard T, et al. Generation and degradation of human endostatin proteins by various proteinases. FEBS letters. 2000;486(3):247–51. [DOI] [PubMed] [Google Scholar]
  • 73.Wen W, Moses MA, Wiederschain D, et al. The generation of endostatin is mediated by elastase. Cancer Res. 1999;59(24):6052–6. [PubMed] [Google Scholar]
  • 74.Patterson BC, Sang QA. Angiostatin-converting enzyme activities of human matrilysin (MMP-7) and gelatinase B/type IV collagenase (MMP-9). J Biol Chem. 1997;272(46):28823–5. [DOI] [PubMed] [Google Scholar]
  • 75.Ahmad S Concise review: limbal stem cell deficiency, dysfunction, and distress. Stem Cells Transl Med. 2012;1(2):110–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Sonoda KH, Nakao S, Nakamura T, et al. Cellular events in the normal and inflamed cornea. Cornea. 2005;24(8 Suppl):S50–S4. [DOI] [PubMed] [Google Scholar]
  • 77.Philipp W, Speicher L, Humpel C. Expression of vascular endothelial growth factor and its receptors in inflamed and vascularized human corneas. Invest Ophthalmol Vis Sci. 2000;41(9):2514–22. [PubMed] [Google Scholar]
  • 78.Abraham S, Sawaya BE, Safak M, et al. Regulation of MCP-1 gene transcription by Smads and HIV-1 Tat in human glial cells. Virology. 2003;309(2):196–202. [DOI] [PubMed] [Google Scholar]
  • 79.Singh N, Amin S, Richter E, et al. Flt-1 intraceptors inhibit hypoxia-induced VEGF expression in vitro and corneal neovascularization in vivo. Invest Ophthalmol Vis Sci. 2005;46(5):1647–52. [DOI] [PubMed] [Google Scholar]
  • 80.Mastyugin V, Mosaed S, Bonazzi A, et al. Corneal epithelial VEGF and cytochrome P450 4B1 expression in a rabbit model of closed eye contact lens wear. Curr Eye Res. 2001;23(1):1–10. [DOI] [PubMed] [Google Scholar]
  • 81.Manalo DJ, Rowan A, Lavoie T, et al. Transcriptional regulation of vascular endothelial cell responses to hypoxia by HIF-1. Blood. 2005;105(2):659–69. [DOI] [PubMed] [Google Scholar]
  • 82.Zhang SX, Ma JX. Ocular neovascularization: Implication of endogenous angiogenic inhibitors and potential therapy. Prog Retin Eye Res. 2007;26(1):1–37. [DOI] [PubMed] [Google Scholar]
  • 83.Sunderkotter C, Roth J, Sorg C. Immunohistochemical detection of bFGF and TNF-alpha in the course of inflammatory angiogenesis in the mouse cornea. Am J Pathol. 1990;137(3):511–5. [PMC free article] [PubMed] [Google Scholar]
  • 84.Edelman JL, Castro MR, Wen Y. Correlation of VEGF expression by leukocytes with the growth and regression of blood vessels in the rat cornea. Invest Ophthalmol Vis Sci. 1999;40(6):1112–23. [PubMed] [Google Scholar]
  • 85.Kallinikos P, Morgan P, Efron N. Assessment of stromal keratocytes and tear film inflammatory mediators during extended wear of contact lenses. Cornea. 2006;25(1):1–10. [DOI] [PubMed] [Google Scholar]
  • 86.Ziche M, Morbidelli L, Masini E, et al. Nitric oxide mediates angiogenesis in vivo and endothelial cell growth and migration in vitro promoted by substance P. J Clin Invest. 1994;94(5):2036–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Sennlaub F, Courtois Y, Goureau O. Nitric oxide synthase-II is expressed in severe corneal alkali burns and inhibits neovascularization. Investigative Ophthalmology and Visual Science. 1999;40:2773–9. [PubMed] [Google Scholar]
  • 88.Vafeas C, Mieyal PA, Urbano F, et al. Hypoxia stimulates the synthesis of cytochrome P450-derived inflammatory eicosanoids in rabbit corneal epithelium. J Pharmacol Exp Ther. 1998;287(3):903–10. [PubMed] [Google Scholar]
  • 89.Mieyal PA, Bonazzi A, Jiang H, et al. The effect of hypoxia on endogenous corneal epithelial eicosanoids. Invest Ophthalmol Vis Sci. 2000;41(8):2170–6. [PubMed] [Google Scholar]
  • 90.Ferrari G, Hajrasouliha AR, Sadrai Z, et al. Nerves and neovessels inhibit each other in the cornea. Investigative ophthalmology & visual science. 2013;54(1):813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Shibuya M Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. Journal of biochemistry. 2013;153(1):13–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kadar T, Amir A, Cohen L, et al. Anti-VEGF therapy (bevacizumab) for sulfur mustard-induced corneal neovascularization associated with delayed limbal stem cell deficiency in rabbits. Curr Eye Res. 2014;39(5):439–50. [DOI] [PubMed] [Google Scholar]
  • 93.Leonardi A, Sathe S, Bortolotti M, et al. Cytokines, matrix metalloproteases, angiogenic and growth factors in tears of normal subjects and vernal keratoconjunctivitis patients. Allergy. 2009;64(5):710–7. [DOI] [PubMed] [Google Scholar]
  • 94.Yan J, Zeng Y, Jiang J, et al. The expression patterns of vascular endothelial growth factor and thrombospondin 2 after corneal alkali burn. Colloids Surf B Biointerfaces. 2007;60(1):105–9. [DOI] [PubMed] [Google Scholar]
  • 95.Chen P, Yin H, Wang Y, et al. Inhibition of VEGF expression and corneal neovascularization by shRNA targeting HIF-1alpha in a mouse model of closed eye contact lens wear. Mol Vis. 2012;18:864–73. [PMC free article] [PubMed] [Google Scholar]
  • 96.Shoshani Y, Pe′er J, Doviner V, et al. Increased expression of inflammatory cytokines and matrix metalloproteinases in pseudophakic corneal edema. Invest Ophthalmol Vis Sci. 2005;46(6):1940–7. [DOI] [PubMed] [Google Scholar]
  • 97.Zheng M, Deshpande S, Lee S, et al. Contribution of vascular endothelial growth factor in the neovascularization process during the pathogenesis of herpetic stromal keratitis. J Virol. 2001;75(20):9828–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Sawano A, Takahashi T, Yamaguchi S, et al. Flt-1 but not KDR/Flk-1 tyrosine kinase is a receptor for placenta growth factor, which is related to vascular endothelial growth factor. Cell Growth Differ. 1996;7(2):213–21. [PubMed] [Google Scholar]
  • 99.Waltenberger J, Claesson-Welsh L, Siegbahn A, et al. Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor. J Biol Chem. 1994;269(43):26988–95. [PubMed] [Google Scholar]
  • 100.Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci US A. 1993;90(22):10705–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Bates DO, Cui TG, Doughty JM, et al. VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, is down-regulated in renal cell carcinoma. Cancer Res. 2002;62(14):4123–31. [PubMed] [Google Scholar]
  • 102.Takahashi T, Ueno H, Shibuya M. VEGF activates protein kinase C-dependent, but Ras-independent Raf-MEK-MAP kinase pathway for DNA synthesis in primary endothelial cells. Oncogene. 1999;18(13):2221–30. [DOI] [PubMed] [Google Scholar]
  • 103.Hiratsuka S, Minowa O, Kuno J, et al. Flt-1 lacking the tyrosine kinase domain is sufficient for normal development and angiogenesis in mice. Proc Natl Acad Sci U S A. 1998;95(16):9349–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Wang F, Yamauchi M, Muramatsu M, et al. RACK1 regulates VEGF/Flt1-mediated cell migration via activation of a PI3K/Akt pathway. J Biol Chem. 2011;286(11):9097–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhou R, Curry JM, Roy LD, et al. A novel association of neuropilin-1 and MUC1 in pancreatic ductal adenocarcinoma: role in induction of VEGF signaling and angiogenesis. Oncogene. 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Le Jan S, Hayashi M, Kasza Z, et al. Functional overlap between chondroitin and heparan sulfate proteoglycans during VEGF-induced sprouting angiogenesis. Arterioscler Thromb Vasc Biol. 2012;32(5):1255–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Ross R, Raines EW, Bowen-Pope DF. The biology of platelet-derived growth factor. Cell. 1986;46(2):155–69. [DOI] [PubMed] [Google Scholar]
  • 108.Kim W-J, Mohan RR, Wilson S. Effect of PDGF, IL-1alpha, and BMP2/4 on corneal fibroblast chemotaxis: expression of the platelet-derived growth factor system in the cornea. Investigative ophthalmology & visual science. 1999;40(7):1364–72. [PubMed] [Google Scholar]
  • 109.Tallquist M, Kazlauskas A. PDGF signaling in cells and mice. Cytokine & growth factor reviews. 2004;15(4):205–13. [DOI] [PubMed] [Google Scholar]
  • 110.Hoppenreijs V, Pels E, Vrensen G, et al. Platelet-derived growth factor: receptor expression in corneas and effects on corneal cells. Investigative ophthalmology & visual science. 1993;34(3):637–49. [PubMed] [Google Scholar]
  • 111.Betsholtz C Insight into the physiological functions of PDGF through genetic studiesin mice. Cytokine & growth factor reviews. 2004;15(4):215–28. [DOI] [PubMed] [Google Scholar]
  • 112.Hellstrom M, Lindahl P, Abramsson A, Betsholtz C. Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth muscle cells and pericytes during embryonic blood vessel formation in the mouse. Development. 1999;126(14):3047–55. [DOI] [PubMed] [Google Scholar]
  • 113.Reinmuth N, Liu W, Jung YD, et al. Induction of VEGF in perivascular cells definesa potential paracrine mechanism for endothelial cell survival. The FASEB Journal. 2001;15(7):1239–41. [DOI] [PubMed] [Google Scholar]
  • 114.Dell S, Peters S, Muther P, et al. The role of PDGF receptor inhibitors and PI3-kinase signaling in the pathogenesis of corneal neovascularization. Investigative ophthalmology & visual science. 2006;47(5):1928–37. [DOI] [PubMed] [Google Scholar]
  • 115.Chaoran Z, Zhirong L, Gezhi X. Combination of vascular endothelial growth factor receptor/platelet-derived growth factor receptor inhibition markedly improves the antiangiogenic efficacy for advanced stage mouse corneal neovascularization. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2011;249(10):1493–501. [DOI] [PubMed] [Google Scholar]
  • 116.Gan L, Fagerholm P, Palmblad J. Expression of basic fibroblast growth factor inrabbit corneal alkali wounds in the presence and absence of granulocytes. Acta Ophthalmol Scand. 2005;83(3):374–8. [DOI] [PubMed] [Google Scholar]
  • 117.Bikfalvi A, Klein S, Pintucci G, Rifkin DB. Biological roles of fibroblast growth factor-2. Endocr Rev. 1997;18(1):26–45. [DOI] [PubMed] [Google Scholar]
  • 118.Ellenberg D, Azar DT, Hallak JA, et al. Novel aspects of corneal angiogenic and lymphangiogenic privilege. Prog Retin Eye Res. 2010;29(3):208–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Knowles RG, Moncada S. Nitric oxide synthases in mammals. Biochem J. 1994;298 (Pt 2):249–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Papapetropoulos A, Desai KM, Rudic RD, et al. Nitric oxide synthase inhibitors attenuate transforming-growth-factor-beta 1-stimulated capillary organization in vitro. The American journal of pathology. 1997;150(5):1835. [PMC free article] [PubMed] [Google Scholar]
  • 121.Sakkoula E, Pipili-Synetos E, Maragoudakis M. Involvement of nitric oxide in the inhibition of angiogenesis by interleukin-2. British journal of pharmacology. 1997;122(5):793–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.VINCENZOCHIARUGI LMA, GALLO O. Cox-2, iNOS and p53 as play-makers of tumor angiogenesis (review). International journal of molecular medicine. 1998;2:715–9. [DOI] [PubMed] [Google Scholar]
  • 123.Kleinert H, Pautz A, Linker K, Schwarz PM. Regulation of the expression ofinducible nitric oxide synthase. Eur J Pharmacol. 2004;500(1–3):255–66. [DOI] [PubMed] [Google Scholar]
  • 124.Gallo O, Fini-Storchi I, Vergari WA, et al. Role of nitric oxide in angiogenesis and tumor progression in head and neck cancer. Journal of the National Cancer Institute. 1998;90(8):587–96. [DOI] [PubMed] [Google Scholar]
  • 125.Fujita N, Nishimoto S, Miyamoto T, et al. Loss Of Nitric Oxide Synthase Type Ii Inhibits Ocular Neovascularization In Mice; Corneal Neovascularization And Argon Laser-induced Choroidal Neovascularization. Investigative Ophthalmology & Visual Science. 2011;52(14):6402-. [Google Scholar]
  • 126.Sennlaub F, Courtois Y, Goureau O. Nitric Oxide Synthase–II Is Expressed in Severe Corneal Alkali Burns and Inhibits Neovascularization. Investigative ophthalmology & visual science. 1999;40(12):2773–9. [PubMed] [Google Scholar]
  • 127.Murohara T, Asahara T, Silver M, et al. Nitric oxide synthase modulates angiogenesis in response to tissue ischemia. Journal of Clinical Investigation. 1998;101(11):2567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hood JD, Meininger CJ, Ziche M, Granger HJ. VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells. American Journal of Physiology-Heart and Circulatory Physiology. 1998;274(3):H1054–H8. [DOI] [PubMed] [Google Scholar]
  • 129.Ziche M, Morbidelli L, Masini E, et al. Nitric oxide promotes DNA synthesis and cyclic GMP formation in endothelial cells from postcapillary venules. Biochemical and biophysical research communications. 1993;192(3):1198–203. [DOI] [PubMed] [Google Scholar]
  • 130.Roy B, Garthwaite J. Nitric oxide activation of guanylyl cyclase in cells revisited. Proceedings of the National Academy of Sciences. 2006; 103(32): 12185–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Croft DR, Sahai E, Mavria G, et al. Conditional ROCK activation in vivo induces tumor cell dissemination and angiogenesis. Cancer Res. 2004;64(24):8994–9001. [DOI] [PubMed] [Google Scholar]
  • 132.Menager C, Vassy J, Doliger C, et al. Subcellular localization of RhoA and ezrin at membrane ruffles of human endothelial cells: differential role of collagen and fibronectin. Exp Cell Res. 1999;249(2):221–30. [DOI] [PubMed] [Google Scholar]
  • 133.Simoncini T, Scorticati C, Mannella P, et al. Estrogen receptor alpha interacts with Galpha13 to drive actin remodeling and endothelial cell migration via the RhoA/Rho kinase/moesin pathway. Mol Endocrinol. 2006;20(8):1756–71. [DOI] [PubMed] [Google Scholar]
  • 134.Pola R, Ling LE, Silver M, et al. The morphogen Sonic hedgehog is an indirect angiogenic agent upregulating two families of angiogenic growth factors. Nat Med. 2001;7(6):706–11. [DOI] [PubMed] [Google Scholar]
  • 135.Kusano KF, Pola R, Murayama T, et al. Sonic hedgehog myocardial gene therapy: tissue repair through transient reconstitution of embryonic signaling. Nat Med.2005;11(11): 1197–204. [DOI] [PubMed] [Google Scholar]
  • 136.Bryan BA, Dennstedt E, Mitchell DC, et al. RhoA/ROCK signaling is essential for multiple aspects of VEGF-mediated angiogenesis. FASEB J. 2010;24(9):3186–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Renault MA, Roncalli J, Tongers J, et al. Sonic hedgehog induces angiogenesis via Rho kinase-dependent signaling in endothelial cells. J Mol Cell Cardiol. 2010;49(3):490–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Le Boeuf F, Houle F, Sussman M, Huot J. Phosphorylation of focal adhesion kinase (FAK) on Ser732 is induced by rho-dependent kinase and is essential for proline-rich tyrosine kinase-2-mediated phosphorylation of FAK on Tyr407 in response to vascular endothelial growth factor. Mol Biol Cell. 2006;17(8):3508–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hyvelin JM, Howell K, Nichol A, et al. Inhibition of Rho-kinase attenuates hypoxia-induced angiogenesis in the pulmonary circulation. Circ Res. 2005;97(2):185–91. [DOI] [PubMed] [Google Scholar]
  • 140.Hata Y, Miura M, Nakao S, et al. Antiangiogenic properties of fasudil, a potent Rho-Kinase inhibitor. Jpn J Ophthalmol. 2008;52(1):16–23. [DOI] [PubMed] [Google Scholar]
  • 141.Yin L, Morishige K, Takahashi T, et al. Fasudil inhibits vascular endothelial growth factor-induced angiogenesis in vitro and in vivo. Mol Cancer Ther. 2007;6(5):1517–25. [DOI] [PubMed] [Google Scholar]
  • 142.Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer. 2008;8(5):387–98. [DOI] [PubMed] [Google Scholar]
  • 143.Reis M, Liebner S. Wnt signaling in the vasculature. Exp Cell Res. 2013;319(9):1317–23. [DOI] [PubMed] [Google Scholar]
  • 144.Dejana E The role of wnt signaling in physiological and pathological angiogenesis. Circ Res. 2010;107(8):943–52. [DOI] [PubMed] [Google Scholar]
  • 145.Kinnunen K, Yla-Herttuala S. Vascular endothelial growth factors in retinal and choroidal neovascular diseases. Ann Med. 2012;44(1):1–17. [DOI] [PubMed] [Google Scholar]
  • 146.Chang J-H, Huang Y-H, Cunningham CM, et al. Matrix metalloproteinase 14 modulates signal transduction and angiogenesis in the cornea. Survey of ophthalmology. 2016;61(4):478–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wang Z, Cheng R, Lee K, et al. Nanoparticle-mediated expression of a Wnt pathway inhibitor ameliorates ocular neovascularization. Arteriosclerosis, thrombosis, and vascular biology. 2015:ATVBAHA. 114.304627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Benko S, Tozser J, Miklossy G, et al. Constitutive and UV-B modulated transcription of Nod-like receptors and their functional partners in human corneal epithelial cells. 2008. [PMC free article] [PubMed] [Google Scholar]
  • 149.Hasegawa M, Fujimoto Y, Lucas PC, et al. A critical role of RICK/RIP2 polyubiquitination in Nod-induced NF-κB activation. The EMBO journal. 2008;27(2):373–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kim SJ, Lee JW, Yu S-L, et al. The role of Nod1 signaling in corneal neovascularization. Cornea. 2013;32(5):674–9. [DOI] [PubMed] [Google Scholar]
  • 151.Inatomi T, Nakamura T, Koizumi N, et al. Midterm results on ocular surface reconstruction using cultivated autologous oral mucosal epithelial transplantation. Am J Ophthalmol. 2006;141(2):267–75. [DOI] [PubMed] [Google Scholar]
  • 152.Faraj LA, Said DG, Al-Aqaba M, et al. Clinical evaluation and characterisation of corneal vascularisation. Br J Ophthalmol. 2016;100(3):315–22. [DOI] [PubMed] [Google Scholar]
  • 153.Dastjerdi MH, Al-Arfaj KM, Nallasamy N, et al. Topical bevacizumab in the treatment of corneal neovascularization: results of a prospective, open-label, noncomparative study. Arch Ophthalmol. 2009;127(4):381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ziche M, Maglione D, Ribatti D, et al. Placenta growth factor-1 is chemotactic, mitogenic, and angiogenic. Lab Invest. 1997;76(4):517–31. [PubMed] [Google Scholar]
  • 155.Kim TI, Kim SW, Kim S, et al. Inhibition of experimental corneal neovascularization by using subconjunctival injection of bevacizumab (Avastin). Cornea. 2008;27(3):349–52. [DOI] [PubMed] [Google Scholar]
  • 156.Tatham A, Tatham E, Prydal J. Validation of a semi-automated computer-aided technique for quantifying corneal vascularisation and scarring. Br J Ophthalmol. 2011;95(10):1379–84. [DOI] [PubMed] [Google Scholar]
  • 157.Cursiefen C, Bock F, Horn FK, et al. GS-101 antisense oligonucleotide eye drops inhibit corneal neovascularization: interim results of a randomized phase II trial. Ophthalmology. 2009; 116(9):1630–7. [DOI] [PubMed] [Google Scholar]
  • 158.Cursiefen C, Viaud E, Bock F, et al. Aganirsen antisense oligonucleotide eye drops inhibit keratitis-induced corneal neovascularization and reduce need for transplantation: the I-CAN study. Ophthalmology. 2014;121(9):1683–92. [DOI] [PubMed] [Google Scholar]
  • 159.Anijeet DR, Zheng Y, Tey A, et al. Imaging and evaluation of corneal vascularization using fluorescein and indocyanine green angiography. Invest Ophthalmol Vis Sci. 2012;53(2):650–8. [DOI] [PubMed] [Google Scholar]
  • 160.Kirwan RP, Zheng Y, Tey A, et al. Quantifying changes in corneal neovascularization using fluorescein and indocyanine green angiography. Am J Ophthalmol. 2012;154(5):850–8.e2. [DOI] [PubMed] [Google Scholar]
  • 161.Ang M, Sim DA, Keane PA, et al. Optical Coherence Tomography Angiography for Anterior Segment Vasculature Imaging. Ophthalmology 2015;122(9):1740–7. [DOI] [PubMed] [Google Scholar]
  • 162.Ang M, Cai Y, Shahipasand S, et al. En face optical coherence tomography angiography for corneal neovascularisation. Br J Ophthalmol. 2016;100(5):616–21. [DOI] [PubMed] [Google Scholar]
  • 163.Romano V, Steger B, Zheng Y, et al. Angiographic and In Vivo Confocal Microscopic Characterization of Human Corneal Blood and Presumed Lymphatic Neovascularization: A Pilot Study. Cornea. 2015;34(11):1459–65. [DOI] [PubMed] [Google Scholar]
  • 164.Peebo BB, Fagerholm P, Traneus-Rockert C, Lagali N. Cellular-level characterization of lymph vessels in live, unlabeled corneas by in vivo confocal microscopy. Invest Ophthalmol Vis Sci. 2010;51(2):830–5. [DOI] [PubMed] [Google Scholar]
  • 165.Horstmann J, Schulz-Hildebrandt H, Bock F, et al. Label-Free In Vivo Imaging of Corneal Lymphatic Vessels Using Microscopic Optical Coherence Tomography. Invest Ophthalmol Vis Sci. 2017;58(13):5880–6. [DOI] [PubMed] [Google Scholar]
  • 166.Burger PC, Chandler DB, Klintworth GK. Experimental corneal neovascularization: biomicroscopic, angiographic, and morphologic correlation. Cornea. 1985;4(1):35–41. [PubMed] [Google Scholar]
  • 167.Goktas S, Erdogan E, Sakarya R, et al. Inhibition of corneal neovascularization by topical and subconjunctival tigecycline. Journal of ophthalmology. 2014;2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Schroedl F, Kaser-Eichberger A, Schlereth SL, et al. Consensus statement on the immunohistochemical detection of ocular lymphatic vessels. Invest Ophthalmol Vis Sci. 2014;55(10):6440–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Cursiefen C, Kuchle M, Naumann GO. Angiogenesis in corneal diseases: histopathologic evaluation of 254 human corneal buttons with neovascularization. Cornea. 1998;17(6):611–3. [DOI] [PubMed] [Google Scholar]
  • 170.Clements JL, Dana R. Inflammatory corneal neovascularization: etiopathogenesis. Semin Ophthalmol. 2011;26(4–5):235–45. [DOI] [PubMed] [Google Scholar]
  • 171.Liclican EL, Nguyen V, Sullivan AB, Gronert K. Selective activation of the prostaglandin E2 circuit in chronic injury-induced pathologic angiogenesis. Invest Ophthalmol Vis Sci. 2010;51(12):6311–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.McNatt LG, Weimer L, Yanni J, Clark AF. Angiostatic activity of steroids in the chick embryo CAM and rabbit cornea models of neovascularization. J Ocul Pharmacol Ther. 1999;15(5):413–23. 173. [DOI] [PubMed] [Google Scholar]
  • 173.Clark AF. Mechanism of action of the angiostatic cortisene anecortave acetate. Surv Ophthalmol. 2007;52 Suppl 1:S26–34. [DOI] [PubMed] [Google Scholar]
  • 174.Carnahan MC, Goldstein DA. Ocular complications of topical, peri-ocular, and systemic corticosteroids. Curr Opin Ophthalmol. 2000;11(6):478–83. [DOI] [PubMed] [Google Scholar]
  • 175.Pakneshan P, Birsner AE, Adini I, et al. Differential suppression of vascular permeability and corneal angiogenesis by nonsteroidal anti-inflammatory drugs. Invest Ophthalmol Vis Sci. 2008;49(9):3909–13. [DOI] [PubMed] [Google Scholar]
  • 176.Flach AJ. Corneal melts associated with topically applied nonsteroidal anti­inflammatory drugs. Trans Am Ophthalmol Soc. 2001;99:205–10; discussion 10–2. [PMC free article] [PubMed] [Google Scholar]
  • 177.Hernandez GL, Volpert OV, Iniguez MA, et al. Selective inhibition of vascular endothelial growth factor-mediated angiogenesis by cyclosporin A: roles of the nuclear factor of activated T cells and cyclooxygenase 2. J Exp Med. 2001;193(5):607–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Bucak YY, Erdurmus M, Terzi EH, et al. Inhibitory effects of topical cyclosporine A 0.05% on immune-mediated corneal neovascularization in rabbits. Graefes Arch Clin Exp Ophthalmol. 2013;251(11):2555–61. [DOI] [PubMed] [Google Scholar]
  • 179.Bock F, Matthaei M, Reinhard T, et al. High-dose subconjunctival cyclosporine a implants do not affect corneal neovascularization after high-risk keratoplasty. Ophthalmology. 2014; 121 (9): 1677–82. [DOI] [PubMed] [Google Scholar]
  • 180.Joseph A, Raj D, Shanmuganathan V, et al. Tacrolimus immunosuppression in high-risk corneal grafts. Br J Ophthalmol. 2007;91(1):51–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Sloper CM, Powell RJ, Dua HS. Tacrolimus (FK506) in the management of high-risk corneal and limbal grafts. Ophthalmology. 2001;108(10):1838–44. [DOI] [PubMed] [Google Scholar]
  • 182.Turgut B, Guler M, Akpolat N, et al. The impact of tacrolimus on vascular endothelial growth factor in experimental corneal neovascularization. Curr Eye Res. 2011;36(1):34–40. [DOI] [PubMed] [Google Scholar]
  • 183.Baroja-Mazo A, Revilla-Nuin B, Ramirez P, Pons JA. Immunosuppressive potency of mechanistic target of rapamycin inhibitors in solid-organ transplantation. World J Transplant. 2016;6(1):183–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Gidfar S, Milani FY, Milani BY, et al. Rapamycin Prolongs the Survival of Corneal Epithelial Cells in Culture. Sci Rep. 2017;7:40308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Shin YJ, Hyon JY, Choi WS, et al. Chemical injury-induced corneal opacity and neovascularization reduced by rapamycin via TGF-beta1/ERK pathways regulation. Invest Ophthalmol Vis Sci. 2013;54(7):4452–8. [DOI] [PubMed] [Google Scholar]
  • 186.Cakmak H, Ergin K, Bozkurt G, et al. The effects of topical everolimus and sunitinib on corneal neovascularization. Cutan Ocul Toxicol. 2016;35(2):97–103. [DOI] [PubMed] [Google Scholar]
  • 187.Sari ES, Yazici A, Aksit H, et al. Inhibitory effect of sub-conjunctival tocilizumab on alkali burn induced corneal neovascularization in rats. Curr Eye Res. 2015;40(1):48–55. [DOI] [PubMed] [Google Scholar]
  • 188.Yoo AR, Chung SK. Effects of Subconjunctival Tocilizumab Versus Bevacizumab in Treatment of Corneal Neovascularization in Rabbits. Cornea. 2014;33(10):1088–94. [DOI] [PubMed] [Google Scholar]
  • 189.Ferrari G, Bignami F, Rama P. Tumor necrosis factor-alpha inhibitors as a treatment of corneal hemangiogenesis and lymphangiogenesis. Eye Contact Lens. 2015;41(2):72–6. [DOI] [PubMed] [Google Scholar]
  • 190.Kim JW, Chung SK. The effect of topical infliximab on corneal neovascularization in rabbits. Cornea. 2013;32(2):185–90. [DOI] [PubMed] [Google Scholar]
  • 191.Ferrari G, Bignami F, Giacomini C, et al. Safety and Efficacy of Topical Infliximab in a Mouse Model of Ocular Surface ScarringSafety and Efficacy of Topical Infliximab. Investigative ophthalmology & visual science. 2013;54(3):1680–8. [DOI] [PubMed] [Google Scholar]
  • 192.Cade F, Paschalis EI, Regatieri CV, et al. Alkali burn to the eye: protection using TNF-α inhibition. Cornea. 2014;33(4):382–9. [DOI] [PubMed] [Google Scholar]
  • 193.Ozdemir O, Altintas O, Altintas L, et al. Effects of subconjunctivally injected bevacizumab, etanercept, and the combination of both drugs on experimental corneal neovascularization. Canadian Journal of Ophthalmology/Journal Canadien d’Ophtalmologie. 2013;48(2):115–20. [DOI] [PubMed] [Google Scholar]
  • 194.Chang JH, Garg NK, Lunde E, et al. Corneal neovascularization: an anti-VEGF therapy review. Surv Ophthalmol. 2012;57(5):415–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Keating AM, Jacobs DS. Anti-VEGF Treatment of Corneal Neovascularization. Ocul Surf. 2011;9(4):227–37. [DOI] [PubMed] [Google Scholar]
  • 196.Cheng SF, Dastjerdi MH, Ferrari G, et al. Short-term topical bevacizumab in the treatment of stable corneal neovascularization. Am J Ophthalmol. 2012;154(6):940–8.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Koenig Y, Bock F, Horn F, et al. Short- and long-term safety profile and efficacy of topical bevacizumab (Avastin) eye drops against corneal neovascularization. Graefes Arch Clin Exp Ophthalmol. 2009;247(10):1375–82. [DOI] [PubMed] [Google Scholar]
  • 198.Dastjerdi MH, Sadrai Z, Saban DR, et al. Corneal penetration of topical and subconjunctival bevacizumab. Invest Ophthalmol Vis Sci. 2011;52(12):8718–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Chu HS, Hu FR, Yang CM, et al. Subconjunctival injection of bevacizumab in the treatment of corneal neovascularization associated with lipid deposition. Cornea. 2011;30(1):60–6. [DOI] [PubMed] [Google Scholar]
  • 200.Kim J, Kim D, Kim ES, et al. Topically administered bevacizumab had longer standing anti-angiogenic effect than subconjunctivally injected bevacizumab in rat corneal neovacularization. Int J Ophthalmol. 2013;6(5):588–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Oner V, Kucukerdonmez C, Akova YA, et al. Topical and subconjunctival bevacizumab for corneal neovascularization in an experimental rat model. Ophthalmic Res. 2012;48(3):118–23. [DOI] [PubMed] [Google Scholar]
  • 202.Papathanassiou M, Theodoropoulou S, Analitis A, et al. Vascular endothelial growth factor inhibitors for treatment of corneal neovascularization: a meta-analysis. Cornea. 2013;32(4):435–44. [DOI] [PubMed] [Google Scholar]
  • 203.Chu HS, Chen TC, Hu FR, Chen WL. Recurrence of corneal neovascularization associated with lipid deposition after subconjunctival injection of bevacizumab. Cornea. 2013;32(11):1446–53. [DOI] [PubMed] [Google Scholar]
  • 204.Liarakos VS, Papaconstantinou D, Vergados I, et al. The effect of subconjunctival ranibizumab on corneal and anterior segment neovascularization: study on an animal model. Eur J Ophthalmol. 2014;24(3):299–308. [DOI] [PubMed] [Google Scholar]
  • 205.Turkcu FM, Cinar Y, Turkcu G, et al. Topical and subconjunctival ranibizumab (lucentis) for corneal neovascularization in experimental rat model. Cutan Ocul Toxicol. 2014;33(2):138–44. [DOI] [PubMed] [Google Scholar]
  • 206.Ferrari G, Dastjerdi MH, Okanobo A, et al. Topical ranibizumab as a treatment of corneal neovascularization. Cornea. 2013;32(7):992–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Ahn YJ, Hwang HB, Chung SK. Ranibizumab injection for corneal neovascularization refractory to bevacizumab treatment. Korean J Ophthalmol. 2014;28(2):177–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Kim JH, Seo HW, Han HC, et al. The effect of bevacizumab versus ranibizumab in the treatment of corneal neovascularization: a preliminary study. Korean J Ophthalmol. 2013;27(4):235–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Stevenson W, Cheng SF, Dastjerdi MH, et al. Corneal neovascularization and the utility of topical VEGF inhibition: ranibizumab (Lucentis) vs bevacizumab (Avastin). Ocul Surf. 2012;10(2):67–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Akar EE, Oner V, Kucukerdonmez C, Aydin Akova Y. Comparison of subconjunctivally injected bevacizumab, ranibizumab, and pegaptanib for inhibition of corneal neovascularization in a rat model. Int J Ophthalmol. 2013;6(2):136–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Wang Q, Yang J, Tang K, et al. Pharmacological characteristics and efficacy of a novel anti-angiogenic antibody FD006 in corneal neovascularization. BMC biotechnology. 2014;14(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Oliveira HB, Sakimoto T, Javier JA, et al. VEGF Trap(R1R2) suppresses experimental corneal angiogenesis. Eur J Ophthalmol. 2010;20(1):48–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Park Y-R, Chung SK. Inhibitory effect of topical aflibercept on corneal neovascularization in rabbits. Cornea. 2015;34(10):1303–7. [DOI] [PubMed] [Google Scholar]
  • 214.Dohlman TH, Omoto M, Hua J, et al. VEGF-trap aflibercept significantly improves long-term graft survival in high-risk corneal transplantation. Transplantation. 2015;99(4):678–86. [DOI] [PubMed] [Google Scholar]
  • 215.Berdugo M, Andrieu-Soler C, Doat M, et al. Downregulation of IRS-1 expression causes inhibition of corneal angiogenesis. Investigative ophthalmology & visual science. 2005;46(11):4072–8. [DOI] [PubMed] [Google Scholar]
  • 216.Kim EC, Ryu HW, Lee HJ, Kim MS. Bevacizumab eye drops delay corneal epithelial wound healing and increase the stromal response to epithelial injury in rats. Clin Exp Ophthalmol. 2013;41(7):694–701. [DOI] [PubMed] [Google Scholar]
  • 217.Dekaris I, Gabric N, Draca N, et al. Three-year corneal graft survival rate in high-risk cases treated with subconjunctival and topical bevacizumab. Graefes Arch Clin Exp Ophthalmol. 2015;253(2):287–94. [DOI] [PubMed] [Google Scholar]
  • 218.Kim TI, Chung JL, Hong JP, et al. Bevacizumab application delays epithelial healing in rabbit cornea. Invest Ophthalmol Vis Sci. 2009;50(10):4653–9. [DOI] [PubMed] [Google Scholar]
  • 219.Petsoglou C, Balaggan KS, Dart JK, et al. Subconjunctival bevacizumab induces regression of corneal neovascularisation: a pilot randomised placebo-controlled double­masked trial. Br J Ophthalmol. 2013;97(1):28–32. [DOI] [PubMed] [Google Scholar]
  • 220.Dan L, Shi-long Y, Miao-li L, et al. Inhibitory effect of oral doxycycline on neovascularization in a rat corneal alkali burn model of angiogenesis. Curr Eye Res. 2008;33(8):653–60. [DOI] [PubMed] [Google Scholar]
  • 221.Su W, Li Z, Lin M, et al. The effect of doxycycline temperature-sensitive hydrogel on inhibiting the corneal neovascularization induced by BFGF in rats. Graefes Arch Clin Exp Ophthalmol. 2011;249(3):421–7. [DOI] [PubMed] [Google Scholar]
  • 222.Jovanovic V, Nikolic L. The effect of topical doxycycline on corneal neovascularization. Curr Eye Res. 2014;39(2):142–8. [DOI] [PubMed] [Google Scholar]
  • 223.Su W, Li Z, Li F, et al. Doxycycline-mediated inhibition of corneal angiogenesis: an MMP-independent mechanism. Invest Ophthalmol Vis Sci. 2013;54(1):783–8. [DOI] [PubMed] [Google Scholar]
  • 224.Su W, Li Z, Li Y, et al. Doxycycline enhances the inhibitory effects of bevacizumab on corneal neovascularization and prevents its side effects. Invest Ophthalmol Vis Sci. 2011;52(12):9108–15. [DOI] [PubMed] [Google Scholar]
  • 225.Xiao O, Xie ZL, Lin BW, et al. Minocycline inhibits alkali burn-induced corneal neovascularization in mice. PLoS One. 2012;7(7):e41858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Sun L, Liang C, Shirazian S, et al. Discovery of 5-[5-fluoro-2-oxo-1, 2-dihydroindol-(3 Z)-ylidenemethyl]-2, 4-dimethyl-1 H-pyrrole-3-carboxylic acid (2-diethylaminoethyl) amide, a novel tyrosine kinase inhibitor targeting vascular endothelial and platelet-derived growth factor receptor tyrosine kinase. Journal of medicinal chemistry. 2003;46(7): 1116–9. [DOI] [PubMed] [Google Scholar]
  • 227.Goodman VL, Rock EP, Dagher R, et al. Approval summary: sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clinical Cancer Research. 2007;13(5):1367–73. [DOI] [PubMed] [Google Scholar]
  • 228.Detry B, Blacher S, Erpicum C, et al. Sunitinib Inhibits Inflammatory Corneal LymphangiogenesisSunitinib Inhibits Corneal Neovascularization. Investigative ophthalmology & visual science. 2013;54(5):3082–93. [DOI] [PubMed] [Google Scholar]
  • 229.Pérez-Santonja JJ, Campos-Mollo E, Lledó-Riquelme M, et al. Inhibition of corneal neovascularization by topical bevacizumab (anti-VEGF) and sunitinib (anti-VEGF and anti-PDGF) in an animal model. American journal of ophthalmology 2010;150(4):519–28.e1. [DOI] [PubMed] [Google Scholar]
  • 230.Ko BY, Kim YS, Baek SG, et al. Inhibition of corneal neovascularization by subconjunctival and topical bevacizumab and sunitinib in a rabbit model. Cornea. 2013;32(5):689–95. [DOI] [PubMed] [Google Scholar]
  • 231.Bayyoud T, Hofmann J, Spitzer M, et al. Cytotoxic properties of sunitinib and sorafenib on human corneal epithelial cells. Curr Eye Res. 2014;39(2): 149–54. [DOI] [PubMed] [Google Scholar]
  • 232.Amparo F, Sadrai Z, Jin Y, et al. Safety and efficacy of the multitargeted receptor kinase inhibitor pazopanib in the treatment of corneal neovascularization. Invest Ophthalmol Vis Sci. 2013;54(1):537–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Wilhelm SM, Carter C, Tang L, et al. BAY 43–9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer research. 2004;64(19):7099–109. [DOI] [PubMed] [Google Scholar]
  • 234.Seo JW, Chung S-H, Choi J-S, Joo C-K. Inhibition of corneal neovascularization in rats by systemic administration of sorafenib. Cornea. 2012;31(8):907–12. [DOI] [PubMed] [Google Scholar]
  • 235.Eisen T, Joensuu H, Nathan PD, et al. Regorafenib for patients with previously untreated metastatic or unresectable renal-cell carcinoma: a single-group phase 2 trial. The lancet oncology. 2012; 13(10): 1055–62. [DOI] [PubMed] [Google Scholar]
  • 236.Onder HI, Erdurmus M, Bucak YY, et al. Inhibitory effects of regorafenib, a multiple tyrosine kinase inhibitor, on corneal neovascularization. International journal of ophthalmology 2014;7(2):220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.den Hollander P, Savage MI, Brown PH. Targeted therapy for breast cancer prevention. Frontiers in oncology 2013;3:250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Kaya MK, Demir T, Bulut H, et al. Effects of lapatinib and trastuzumab on vascular endothelial growth factor in experimental corneal neovascularization. Clinical & experimental ophthalmology. 2015;43(5):449–57. [DOI] [PubMed] [Google Scholar]
  • 239.Zeng P, Pi R-b, Li P, et al. Fasudil hydrochloride, a potent ROCK inhibitor, inhibits corneal neovascularization after alkali burns in mice. Molecular vision.. 2015;21:688. [PMC free article] [PubMed] [Google Scholar]
  • 240.Boland S, Bourin A, Alen J, et al. Design, synthesis, and biological evaluation of novel, highly active soft ROCK inhibitors. J Med Chem. 2015;58(10):4309–24. [DOI] [PubMed] [Google Scholar]
  • 241.Sijnave D, Van Bergen T, Castermans K, et al. Inhibition of rho-associated kinase prevents pathological wound healing and neovascularization after corneal trauma. Cornea. 2015;34(9):1120–9. [DOI] [PubMed] [Google Scholar]
  • 242.Elbaz U, Mireskandari K, Shen C, Ali A. Corneal Fine Needle Diathermy With Adjuvant Bevacizumab to Treat Corneal Neovascularization in Children. Cornea. 2015;34(7):773–7. [DOI] [PubMed] [Google Scholar]
  • 243.Park SC, Kim JH. Effects of laser photocoagulation on corneal neovascularization in rabbits. J Refract Corneal Surg. 1994;10(6):631–9. [PubMed] [Google Scholar]
  • 244.Hemady RK, Baer JC, Foster CS. Biomicroscopic and histopathologic observations after corneal laser photocoagulation in a rabbit model of corneal neovascularization. Cornea. 1993;12(3):185–90. [DOI] [PubMed] [Google Scholar]
  • 245.Krasnick NM, Spigelman AV. Comparison of yellow dye, continuous wave Nd:YAG, and argon green laser on experimentally induced corneal neovascularization. J Refract Surg. 1995;11(1):45–9. [DOI] [PubMed] [Google Scholar]
  • 246.Goto S Q-switched Nd:YAG laser treatment for corneal neovascularization. Jpn J Ophthalmol. 1992;36(3):291–300. [PubMed] [Google Scholar]
  • 247.Sharma A, Samal A, Narang S, et al. Frequency doubled Nd:YAG (532 nm) laser photocoagulation in corneal vascularisation: efficacy and time sequenced changes. Indian J Ophthalmol. 2001;49(4):235–40. [PubMed] [Google Scholar]
  • 248.Kumar J, Gehra A, Sirohi N. Role of Frequency Doubled Nd: Yag Laser in Treatmentof Corneal Neovascularisation. J Clin Diagn Res. 2016;10(4):Nc01–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Parsa CF, Temprano J, Wilson D, Green WR. Hemorrhage complicating YAG laser feeder vessel coagulation of cornea vascularization. Cornea. 1994;13(3):264–8. [DOI] [PubMed] [Google Scholar]
  • 250.Marsh RJ. Argon laser treatment of lipid keratopathy. Br J Ophthalmol. 1988;72(12):900–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Faraj LA, Elalfy MS, Said DG, Dua HS. Fine needle diathermy occlusion of corneal vessels. Br J Ophthalmol. 2014;98(9):1287–90. [DOI] [PubMed] [Google Scholar]
  • 252.Trikha S, Parikh S, Osmond C, et al. Long-term outcomes of Fine Needle Diathermy for established corneal neovascularisation. Br J Ophthalmol. 2014;98(4):454–8. [DOI] [PubMed] [Google Scholar]
  • 253.Koenig Y, Bock F, Kruse FE, et al. Angioregressive pretreatment of mature corneal blood vessels before keratoplasty: fine-needle vessel coagulation combined with anti-VEGFs. Cornea. 2012;31(8):887–92. [DOI] [PubMed] [Google Scholar]
  • 254.Al-Torbak AA. Photodynamic therapy with verteporfin for corneal neovascularization. Middle East Afr J Ophthalmol. 2012;19(2):185–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Al-Abdullah AA, Al-Assiri A. Resolution of bilateral corneal neovascularization and lipid keratopathy after photodynamic therapy with verteporfin. Optometry. 2011;82(4):212–4. [DOI] [PubMed] [Google Scholar]
  • 256.Kheirkhah A, Johnson DA, Paranjpe DR, et al. Temporary sutureless amniotic membrane patch for acute alkaline burns. Archives of ophthalmology 2008;126(8):1059–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Lee HS, Lee JH, Kim CE, Yang JW. Anti-neovascular effect of chondrocyte-derived extracellular matrix on corneal alkaline burns in rabbits. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2014;252(6):951–61. [DOI] [PubMed] [Google Scholar]
  • 258.Holland EJ, Schwartz GS. The Paton lecture: Ocular surface transplantation: 10 years’ experience. Cornea. 2004;23(5):425–31. [DOI] [PubMed] [Google Scholar]
  • 259.Konomi K, Satake Y, Shimmura S, et al. Long-term results of amniotic membrane transplantation for partial limbal deficiency. Cornea. 2013;32(8): 1110–5. [DOI] [PubMed] [Google Scholar]
  • 260.Eslani M, Baradaran-Rafii A, Djalilian A. Conjunctival-Limbal Autograft (CLAU) In: Thomsen WL, ed. Advances in Eye Research Volume 1 Hauppauge, NY: Nova Biomedical Press, 2011. [Google Scholar]
  • 261.Ozdemir O, Tekeli O, Ornek K, et al. Limbal autograft and allograft transplantations in patients with corneal burns. Eye (Lond). 2004;18(3):241–8. [DOI] [PubMed] [Google Scholar]
  • 262.Cheung AY, Sarnicola E, Holland EJ. Long-Term Ocular Surface Stability in Conjunctival Limbal Autograft Donor Eyes. Cornea. 2017;36(9):1031–5. [DOI] [PubMed] [Google Scholar]
  • 263.Pauklin M, Fuchsluger TA, Westekemper H, et al. Midterm results of cultivated autologous and allogeneic limbal epithelial transplantation in limbal stem cell deficiency. Dev Ophthalmol. 2010;45:57–70. [DOI] [PubMed] [Google Scholar]
  • 264.Basu S, Ali H, Sangwan VS. Clinical outcomes of repeat autologous cultivated limbal epithelial transplantation for ocular surface burns. Am J Ophthalmol. 2012;153(4):643–50, 50 e1–2. [DOI] [PubMed] [Google Scholar]
  • 265.Eslani M, Baradaran-Rafii A, Ahmad S. Cultivated limbal and oral mucosal epithelial transplantation. Semin Ophthalmol. 2012;27(3–4):80–93. [DOI] [PubMed] [Google Scholar]
  • 266.Solomon A, Ellies P, Anderson DF, et al. Long-term outcome of keratolimbal allograft with or without penetrating keratoplasty for total limbal stem cell deficiency. Ophthalmology. 2002; 109(6): 1159–66. [DOI] [PubMed] [Google Scholar]
  • 267.Daya SM, Ilari FA. Living related conjunctival limbal allograft for the treatment of stem cell deficiency. Ophthalmology 2001;108(1):126–33; discussion 33–4. [DOI] [PubMed] [Google Scholar]
  • 268.Holland EJ. Epithelial transplantation for the management of severe ocular surface disease. Trans Am Ophthalmol Soc. 1996;94:677–743. [PMC free article] [PubMed] [Google Scholar]
  • 269.Baradaran-Rafii A, Eslani M, Haq Z, et al. Current and Upcoming Therapies for Ocular Surface Chemical Injuries. Ocul Surf. 2017;15(1):48–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Pellegrini G, Traverso CE, Franzi AT, et al. Long-term restoration of damagedcorneal surfaces with autologous cultivated corneal epithelium. Lancet. 1997;349(9057):990–3. [DOI] [PubMed] [Google Scholar]
  • 271.Sangwan VS, Basu S, Vemuganti GK, et al. Clinical outcomes of xeno-free autologous cultivated limbal epithelial transplantation: a 10-year study. Br J Ophthalmol. 2011;95(11):1525–9. [DOI] [PubMed] [Google Scholar]
  • 272.Sangwan VS, Basu S, MacNeil S, Balasubramanian D. Simple limbal epithelial transplantation (SLET): a novel surgical technique for the treatment of unilateral limbal stem cell deficiency. Br J Ophthalmol. 2012;96(7):931–4. [DOI] [PubMed] [Google Scholar]
  • 273.Guarnieri A, Moreno-Montanes J, Alfonso-Bartolozzi B, et al. Quantification of corneal neovascularization after ex vivo limbal epithelial stem cell therapy. Int J Ophthalmol. 2014;7(6):988–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Baradaran-Rafii A, Delfazayebaher S, Aghdami N, et al. Midterm outcomes of penetrating keratoplasty after cultivated oral mucosal epithelial transplantation in chemical burn. Ocul Surf. 2017;15(4):789–94. [DOI] [PubMed] [Google Scholar]
  • 275.Yao L, Bai H. Review: mesenchymal stem cells and corneal reconstruction. Mol Vis. 2013;19:2237–43. [PMC free article] [PubMed] [Google Scholar]
  • 276.Espandar L, Afshari N. Adult Corneal Stem Cells and Alternative Sources for Regenerative Therapy for the Cornea. CML – Ophthalmology. 2013;23(1):1–6. [Google Scholar]
  • 277.Le Blanc K, Frassoni F, Ball L, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. Lancet.2008;371 (9624): 1579–86. [DOI] [PubMed] [Google Scholar]
  • 278.Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Yao L, Li ZR, Su WR, et al. Role of mesenchymal stem cells on cornea wound healing induced by acute alkali burn. PLoS One. 2012;7(2):e30842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Javorkova E, Trosan P, Zajicova A, et al. Modulation of the Early Inflammatory Microenvironment in the Alkali-Burned Eye by Systemically Administered Interferon-gamma-Treated Mesenchymal Stromal Cells. Stem Cells Dev. 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Jiang TS, Cai L, Ji WY, et al. Reconstruction of the corneal epithelium with induced marrow mesenchymal stem cells in rats. Mol Vis. 2010;16:1304–16. [PMC free article] [PubMed] [Google Scholar]
  • 282.Lin HF, Lai YC, Tai CF, et al. Effects of cultured human adipose-derived stem cells transplantation on rabbit cornea regeneration after alkaline chemical burn. Kaohsiung J Med Sci. 2013;29(1):14–8. [DOI] [PubMed] [Google Scholar]
  • 283.Cejkova J, Trosan P, Cejka C, et al. Suppression of alkali-induced oxidative injury in the cornea by mesenchymal stem cells growing on nanofiber scaffolds and transferred onto the damaged corneal surface. Exp Eye Res. 2013;116:312–23. [DOI] [PubMed] [Google Scholar]
  • 284.Bray LJ, Heazlewood CF, Munster DJ, et al. Immunosuppressive properties of mesenchymal stromal cell cultures derived from the limbus of human and rabbit corneas. Cytotherapy. 2014;16(1):64–73. [DOI] [PubMed] [Google Scholar]
  • 285.Oh JY, Roddy GW, Choi H, et al. Anti-inflammatory protein TSG-6 reduces inflammatory damage to the cornea following chemical and mechanical injury. Proc Natl Acad Sci USA. 2010;107(39):16875–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Eslani M, Putra I, Shen X, et al. Corneal Mesenchymal Stromal Cells Are Directly Antiangiogenic via PEDF and sFLT-1. Invest Ophthalmol Vis Sci. 2017;58(12):5507–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Eslani M, Putra I, Shen X, et al. Cornea-Derived Mesenchymal Stromal Cells Therapeutically Modulate Macrophage Immunophenotype and Angiogenic Function. Stem Cells. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Cursiefen C, Colin J, Dana R, et al. Consensus statement on indications for anti-angiogenic therapy in the management of corneal diseases associated with neovascularisation: outcome of an expert roundtable. Br J Ophthalmol. 2012;96(1):3–9. [DOI] [PubMed] [Google Scholar]
  • 289.Syed ZA, Dana R. Novel Treatments for Corneal Angiogenesis. Int Ophthalmol Clin. 2017;57(4):31–8. [DOI] [PubMed] [Google Scholar]
  • 290.Maddula S, Davis DK, Maddula S, et al. Horizons in therapy for corneal angiogenesis. Ophthalmology. 2011;118(3):591–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Liu S, Romano V, Steger B, et al. Gene-based antiangiogenic applications for corneal neovascularization. Surv Ophthalmol. 2018;63(2):193–213. [DOI] [PubMed] [Google Scholar]
  • 292.Lai LJ, Xiao X, Wu JH. Inhibition of corneal neovascularization with endostatin delivered by adeno-associated viral (AAV) vector in a mouse corneal injury model. J Biomed Sci. 2007;14(3):313–22. [DOI] [PubMed] [Google Scholar]
  • 293.Cheng HC, Yeh SI, Tsao YP, Kuo PC. Subconjunctival injection of recombinant AAV-angiostatin ameliorates alkali burn induced corneal angiogenesis. Mol Vis. 2007;13:2344–52. [PubMed] [Google Scholar]
  • 294.Sharma A, Ghosh A, Hansen ET, et al. Transduction efficiency of AAV 2/6, 2/8 and 2/9 vectors for delivering genes in human corneal fibroblasts. Brain Res Bull. 2010;81(2–3):273–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Iriyama A, Usui T, Yanagi Y, et al. Gene transfer using micellar nanovectors inhibits corneal neovascularization in vivo. Cornea. 2011;30(12): 1423–7. [DOI] [PubMed] [Google Scholar]
  • 296.Qazi Y, Stagg B, Singh N, et al. Nanoparticle-mediated delivery of shRNA.VEGF-a plasmids regresses corneal neovascularization. Invest Ophthalmol Vis Sci. 2012;53(6):2837–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Wang YL, Gao GP, Wang YQ, et al. Inhibitory effects of S100A4 gene silencing on alkali burn-induced corneal neovascularization: an in vivo study. Mol Vis. 2017;23:286–95. [PMC free article] [PubMed] [Google Scholar]
  • 298.Zuo L, Fan Y, Wang F, et al. A siRNA targeting vascular endothelial growth factor-A inhibiting experimental corneal neovascularization. Curr Eye Res. 2010;35(5):375–84. [DOI] [PubMed] [Google Scholar]
  • 299.Kick L, Kirchner M, Schneider S. CRISPR-Cas9: From a bacterial immune system to genome-edited human cells in clinical trials. Bioengineered. 2017;8(3):280–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Veritti D, Vergallo S, Lanzetta P. Triple therapy for corneal neovascularization: acase report. Eur J Ophthalmol. 2012;22 Suppl 7:S126–8. [DOI] [PubMed] [Google Scholar]
  • 301.Kirat OM, Al-Dhibi HA. Regression of aggressive corneal vascularization after photodynamic therapy, subconjunctival Avastin injections and topical cyclosporin-A 1% drops: A case report. Saudi J Ophthalmol. 2010;24(4):151–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Feizi S, Azari AA, Safapour S. Therapeutic approaches for corneal neovascularization. Eye Vis (Lond). 2017;4:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Liu X, Wang S, Wang X, et al. Recent drug therapies for corneal neovascularization. Chem Biol Drug Des. 2017;90(5):653–64. [DOI] [PubMed] [Google Scholar]
  • 304.You IC, Im SK, Lee SH, Yoon KC. Photodynamic therapy with verteporfin combined with subconjunctival injection of bevacizumab for corneal neovascularization. Cornea. 2011;30(1):30–3. [DOI] [PubMed] [Google Scholar]
  • 305.Yoon HJ, Kim MK, Seo KY, et al. Effectiveness of photodynamic therapy with verteporfin combined with intrastromal bevacizumab for corneal neovascularization in Stevens-Johnson syndrome. Int Ophthalmol. 2017. [DOI] [PubMed] [Google Scholar]
  • 306.Kuo CN, Chen CY, Chen SN, et al. Inhibition of corneal neovascularization with the combination of bevacizumab and plasmid pigment epithelium-derived factor-synthetic amphiphile INTeraction-18 (p-PEDF-SAINT-18) vector in a rat corneal experimental angiogenesis model. Int J Mol Sci. 2013;14(4):8291–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307.Huang J, Wang W, Yu J, et al. Combination of dexamethasone and Avastin((R)) by supramolecular hydrogel attenuates the inflammatory corneal neovascularization in rat alkali burn model. Colloids Surf B Biointerfaces. 2017; 159:241–50. [DOI] [PubMed] [Google Scholar]
  • 308.Hou Y, Le VNH, Toth G, et al. UV light crosslinking regresses mature corneal blood and lymphatic vessels and promotes subsequent high-risk corneal transplant survival. Am J Transplant. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.Yang SJ, Jo H, Kim K-A, et al. Diospyros kaki extract inhibits alkali burn-induced corneal neovascularization. Journal of medicinal food. 2016;19(1):106–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 310.Kuerten D, Johnen S, Harmening N, et al. Transplantation of PEDF-transfected pigment epithelial cells inhibits corneal neovascularization in a rabbit model. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2015;253(7):1061–9. [DOI] [PubMed] [Google Scholar]
  • 311.Jin J, Ma J-X, Guan M, Yao K. Inhibition of Chemical Cautery–Induced Corneal Neovascularization by Topical Pigment Epithelium–Derived Factor Eyedrops. Cornea. 2010;29(9):1055–61. [DOI] [PubMed] [Google Scholar]
  • 312.Cho W-K, Kang S, Choi H, Rho CR. Topically administered gold nanoparticles inhibit experimental corneal neovascularization in mice. Cornea. 2015;34(4):456–9. [DOI] [PubMed] [Google Scholar]
  • 313.Liu G, Zhang W, Xiao Y, Lu P. Critical Role of IP-10 on Reducing Experimental Corneal Neovascularization. Current eye research. 2015;40(9):891–901. [DOI] [PubMed] [Google Scholar]
  • 314.Han Y, Shao Y, Lin Z, et al. Netrin-1 simultaneously suppresses corneal inflammation and neovascularization. Investigative ophthalmology & visual science. 2012;53(3):1285–95. [DOI] [PubMed] [Google Scholar]
  • 315.Han Y, Shao Y, Liu T, et al. Therapeutic effects of topical netrin-4 inhibits corneal neovascularization in alkali-burn rats. PloS one. 2015;10(4):e0122951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 316.Kim BH, Lee J, Choi JS, et al. Imidazole-based alkaloid derivative LCB54–0009 suppresses ocular angiogenesis and lymphangiogenesis in models of experimental retinopathy and corneal neovascularization. British journal of pharmacology 2015;172(15):3875–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 317.Koh CH, Lee HS, Chung SK. Effect of topical epigallocatechin gallate on corneal neovascularization in rabbits. Cornea. 2014;33(5):527–32. [DOI] [PubMed] [Google Scholar]
  • 318.Zhou H, Jiang S, Chen J, et al. Largazole, an inhibitor of class I histone deacetylases, attenuates inflammatory corneal neovascularization. European journal of pharmacology 2014;740:619–26. [DOI] [PubMed] [Google Scholar]
  • 319.Zhou T, Chen L, Huang C, et al. Serine Proteinase Inhibitor SERPINA3K Suppresses Corneal Neovascularization Via Inhibiting Wnt Signaling and VEGFSERPINA3K and Corneal Neovascularization. Investigative ophthalmology & visual science. 2014;55(8):4863–72. [DOI] [PubMed] [Google Scholar]
  • 320.Shen M, Yuan F, Jin J, Yuan Y. The effect of TC14012 on alkali burn-inducedcorneal neovascularization in mice. Ophthalmic research. 2014;52(1):17–24. [DOI] [PubMed] [Google Scholar]
  • 321.Goktas S, Sakarya R, Erdogan E, et al. Antiangiogenic Effect of Itraconazole on Corneal Neovascularization: A Pilot Experimental Investigation. Ophthalmic research. 2014;52(4):170–4. [DOI] [PubMed] [Google Scholar]
  • 322.Bignami F, Giacomini C, Lorusso A, et al. NK1 Receptor Antagonists as a New Treatment for Corneal NeovascularizationInhibition of Substance P (NK1) Receptor in Cornea. Investigative ophthalmology & visual science. 2014;55(10):6783–94. [DOI] [PubMed] [Google Scholar]
  • 323.Yoon SY, Kim JY, Kim E-S, et al. Subconjunctival Injection of Low-Molecular-Weight Heparin–Taurocholate 7 Inhibits Corneal Neovascularization. Cornea. 2013;32(11):1488–92. [DOI] [PubMed] [Google Scholar]
  • 324.Ge H, Tian P, Guan L, et al. A C-terminal fragment BIGH3 protein with an RGDRGD motif inhibits corneal neovascularization in vitro and in vivo. Experimental eye research. 2013;112:10–20. [DOI] [PubMed] [Google Scholar]
  • 325.Li Z, Yao L, Li J, et al. Celastrol nanoparticles inhibit corneal neovascularization induced by suturing in rats. Int J Nanomedicine. 2012;7:1163–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Dai C, Liu G, Li L, et al. ADP-ribosylation factor as a novel target for corneal neovascularization regression. 2012. [PMC free article] [PubMed] [Google Scholar]
  • 327.Zhou W-J, Liu G-Q, Li L-B, et al. Inhibitory effect of CCR3 signal on alkali-induced corneal neovascularization. International journal of ophthalmology. 2012;5(3):251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 328.Lee MY, Chung SK. Treatment of corneal neovascularization by topical application of ascorbic acid in the rabbit model. Cornea. 2012;31(10): 1165–9. [DOI] [PubMed] [Google Scholar]
  • 329.Tunik S, Nergiz Y, Keklikci U, Akkus M. The subconjunctival use of cetuximab and bevacizumab in inhibition of corneal angiogenesis. Graefe’s Archive for Clinical and Experimental Ophthalmology. 2012;250(8):1161–7. [DOI] [PubMed] [Google Scholar]
  • 330.Shi H, Yu HJ, Wang HY, et al. Topical Administration of Peroxiredoxin-6 on the Cornea Suppresses Inflammation and Neovascularization Induced by Ultraviolet RadiationPRDX6 Inhibited Inflammation and Neovascularization. Investigative ophthalmology & visual science. 2012;53(13):8016–28. [DOI] [PubMed] [Google Scholar]
  • 331.Liu G-Q, Lu P-R, Li L-B, Zhang X-G. Inhibited experimental corneal neovascularization by neutralizing anti-SDF-1a antibody. International journal of ophthalmology. 2012;5(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Byun Y-S, Chung SK. The effect of methotrexate on corneal neovascularization in rabbits. Cornea. 2011;30(4):442–6. [DOI] [PubMed] [Google Scholar]
  • 333.Kubota M, Shimmura S, Kubota S, et al. Hydrogen and N-acetyl-L-cysteine rescue oxidative stress-induced angiogenesis in a mouse cornealalkali-burn model. Investigative ophthalmology & visual science. 2011;52(1):427–33. [DOI] [PubMed] [Google Scholar]
  • 334.Lopez ES, Rizzo MM, Croxatto JO, et al. Suramab, a novel antiangiogenic agent, reduces tumor growth and corneal neovascularization. Cancer chemotherapy and pharmacology. 2011;67(3):723–8. [DOI] [PubMed] [Google Scholar]
  • 335.Wang Y, Yin H, Chen P, Xie L. Inhibitory effect of canstatin in alkali burn-induced corneal neovascularization. Ophthalmic research. 2011;46(2):66–72. [DOI] [PubMed] [Google Scholar]
  • 336.Peng L-H, Shen W, Yong W, et al. Effects of AMD3100 subconjunctival injection on alkali burn induced corneal neovascularization in mice. International journal of ophthalmology. 2011;4(1):44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Zhong Y-Y, Zhang H-F, Zhong J-X, et al. Topical dihydroartemisinin inhibits suture-induced neovascularization in rat corneas through ERK1/2 and p38 pathways. International journal of ophthalmology. 2011; 4(2): 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 338.Huang H, Vasilakis P, Zhong X, et al. Parstatin suppresses ocular neovascularization and inflammation. Investigative ophthalmology & visual science. 2010;51(11):5825–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Hos D, Bock F, Dietrich T, et al. Inflammatory corneal (lymph)angiogenesis isblocked by VEGFR-tyrosine kinase inhibitor ZK 261991, resulting in improved graft survival after corneal transplantation. Invest Ophthalmol Vis Sci. 2008;49(5):1836–42. [DOI] [PubMed] [Google Scholar]
  • 340.Lennikov A, Mirabelli P, Mukwaya A, et al. Selective IKK2 inhibitor IMD0354 disrupts NF-kappaB signaling to suppress corneal inflammation and angiogenesis. Angiogenesis. 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.Dana MR, Zhu SN, Yamada J. Topical modulation of interleukin-1 activity in corneal neovascularization. Cornea. 1998;17(4):403–9. [DOI] [PubMed] [Google Scholar]
  • 342.Lu Y, Xu Y, Gu Q, Xu X. Inhibition of Pathologic Corneal Neovascularization by Topical Application of a Novel Peptide In Vivo. Cornea. 2015;34(10):1295–302. [DOI] [PubMed] [Google Scholar]
  • 343.Taketani Y, Usui T, Toyono T, et al. Topical Use of Angiopoietin-like Protein 2 RNAi-loaded Lipid Nanoparticles Suppresses Corneal Neovascularization. Mol Ther Nucleic Acids. 2016;5:e292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 344.Ma X, Li J. Corneal neovascularization suppressed by TIMP2 released from human amniotic membranes. Yan Ke Xue Bao. 2005;21(1):56–61. [PubMed] [Google Scholar]
  • 345.Zhou H, Jiang S, Chen J, Su SB. Suberoylanilide hydroxamic acid suppresses inflammation-induced neovascularization. Can J Physiol Pharmacol. 2014;92(10):879–85. [DOI] [PubMed] [Google Scholar]
  • 346.Lee HS, Chung SK. The effect of subconjunctival suramin on corneal neovascularization in rabbits. Cornea. 2010;29(1):86–92. [DOI] [PubMed] [Google Scholar]
  • 347.Le VNH, Schneider AC, Scholz R, et al. Fine Needle-Diathermy Regresses Pathological Corneal (Lymph)Angiogenesis and Promotes High-Risk Corneal Transplant Survival. Sci Rep. 2018;8(1):5707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Hou Y, Le VNH, Clahsen T, et al. Photodynamic Therapy Leads to Time-Dependent Regression of Pathologic Corneal (Lymph) Angiogenesis and Promotes High-Risk Corneal Allograft Survival. Invest Ophthalmol Vis Sci. 2017;58(13):5862–9. [DOI] [PubMed] [Google Scholar]
  • 349.Kim SW, Ha BJ, Kim EK, et al. The effect of topical bevacizumab on corneal neovascularization. Ophthalmology. 2008;115(6):e33–8. [DOI] [PubMed] [Google Scholar]
  • 350.Waisbourd M, Levinger E, Varssano D, et al. High-dose topical bevacizumab for corneal neovascularization. Pharmacology 2013;92(5–6):310–4. [DOI] [PubMed] [Google Scholar]
  • 351.Benayoun Y, Adenis JP, Casse G, et al. Effects of subconjunctival bevacizumab on corneal neovascularization: results of a prospective study. Cornea. 2012;31(8):937–44. [DOI] [PubMed] [Google Scholar]
  • 352.Sarah B, Ibtissam H, Mohammed B, et al. Intrastromal Injection of Bevacizumab in the Management of Corneal Neovascularization: About 25 Eyes. J Ophthalmol. 2016;2016:6084270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Bhatti N, Qidwai U, Hussain M, Kazi A. Efficacy of sub-conjunctival and topical bevacizumab in high-risk corneal transplant survival. J Pak Med Assoc. 2013;63(10):1256–9. [PubMed] [Google Scholar]
  • 354.Yeung SN, Lichtinger A, Kim P, et al. Combined use of subconjunctival and intracorneal bevacizumab injection for corneal neovascularization. Cornea. 2011;30(10):1110–4. [DOI] [PubMed] [Google Scholar]
  • 355.Michels R, Michels S, Kaminski S. Effect of combined topical heparin and steroid on corneal neovascularization in children. Ophthalmic Surg Lasers Imaging. 2012;43(6):452–8. [DOI] [PubMed] [Google Scholar]
  • 356.Pillai CT, Dua HS, Hossain P. Fine needle diathermy occlusion of corneal vessels. Invest Ophthalmol Vis Sci. 2000;41(8):2148–53. [PubMed] [Google Scholar]
  • 357.Spiteri N, Romano V, Zheng Y, et al. Corneal angiography for guiding and evaluating fine-needle diathermy treatment of corneal neovascularization. Ophthalmology. 2015;122(6):1079–84. [DOI] [PubMed] [Google Scholar]
  • 358.Gordon YJ, Mann RK, Mah TS, Gorin MB. Fluorescein-potentiated argon laser therapy improves symptoms and appearance of corneal neovascularization. Cornea. 2002;21(8):770–3. [DOI] [PubMed] [Google Scholar]
  • 359.Sheppard JD Jr., Epstein RJ, Lattanzio FA Jr., et al. Argon laser photodynamic therapy of human corneal neovascularization after intravenous administration of dihematoporphyrin ether. Am J Ophthalmol. 2006; 141 (3): 524–9. [DOI] [PubMed] [Google Scholar]
  • 360.Verdiguel-Sotelo K, Hernandez-Lopez A, Gonzalez-Camarena PI, et al. [Photodynamic therapy with verteporfirin in corneal neovascularization]. Rev Med Inst Mex Seguro Soc. 2010;48(3):313–6. [PubMed] [Google Scholar]
  • 361.Diaz-Davalos CD, Carrasco-Quiroz A, Rivera-Diez D. [Neovascularization corneal regression in patients treated with photodynamic therapy with verteporfin]. Rev Med Inst Mex Seguro Soc. 2016;54(2):164–9. [PubMed] [Google Scholar]

RESOURCES