Skip to main content
Indian Journal of Ophthalmology logoLink to Indian Journal of Ophthalmology
. 2024 Apr 20;72(Suppl 3):S354–S371. doi: 10.4103/IJO.IJO_3043_23

Management of corneal neovascularization: Current and emerging therapeutic approaches

Duoduo Wu 1, Kai En Chan 2, Blanche Xiao Hong Lim 1,2, Dawn Ka-Ann Lim 1,2, Wendy Meihua Wong 1,2, Charmaine Chai 1,2, Ray Manotosh 1,2, Chris Hong Long Lim 1,2,3,4,
PMCID: PMC467007  PMID: 38648452

Abstract

Corneal neovascularization (CoNV) is a sight-threatening condition affecting an estimated 1.4 million people per year, and the incidence is expected to rise. It is a complication of corneal pathological diseases such as infective keratitis, chemical burn, corneal limbal stem cell deficiency, mechanical trauma, and immunological rejection after keratoplasties. CoNV occurs due to a disequilibrium in proangiogenic and antiangiogenic mediators, involving a complex system of molecular interactions. Treatment of CoNV is challenging, and no therapy thus far has been curative. Anti-inflammatory agents such as corticosteroids are the mainstay of treatment due to their accessibility and well-studied safety profile. However, they have limited effectiveness and are unable to regress more mature neovascularization. With the advent of advanced imaging modalities and an expanding understanding of its pathogenesis, contemporary treatments targeting a wide array of molecular mechanisms and surgical options are gaining traction. This review aims to summarize evidence regarding conventional and emerging therapeutic options for CoNV.

Keywords: Anti-VEGF, cornea, corneal neovascularization, gene therapy


Corneal neovascularization (CoNV) is a sight-threatening condition that occurs after pathological insults such as microbial keratitis, herpes simplex keratitis, chemical burn, corneal limbal stem cell deficiency (LSCD), mechanical trauma, and immunological rejection following corneal transplantation. It is characterized by disruption of the cornea’s normal avascularity from the invasion of abnormal blood vessels in the stroma [Fig. 1]. These atypical vessels have increased vascular permeability and can cause chronic corneal edema, lipid and protein exudation, and scar formation leading to disarrangement of the regularly arranged lamellar collagen in the cornea. The loss of transparency can lead to visual impairment and blindness in severe cases. With an estimated annual incidence of 1.4 million people, CoNV represents a significant public health concern.[1]

Figure 1.

Figure 1

Clinical manifestations and etiologies of corneal neovascularization. SJS = Steven–Johnson syndrome, TEN = toxic epidermal necrolysis

Unfortunately, despite its prevalence, no curative therapy is available for this condition. Various treatments including anti-inflammatory agents, immunomodulatory drugs, fine needle diathermy (FND), and laser photocoagulation have been proposed to mitigate CoNV. However, these treatments often have limited effectiveness and may be associated with adverse effects. More recently, the introduction of anti-vascular endothelial growth factor (anti-VEGF) drugs has provided new therapeutic options. There is also growing interest in combining different treatment modalities in a synergistic manner to enhance the management of CoNV. This review aims to provide an overview of the existing treatment options for CoNV and shed light on emerging therapeutic approaches for this debilitating condition.

Methods

A literature search of the PubMed database was performed to include publications from inception to 2023. The key search terms included but were not limited to “corneal neovascularization,” “CoNV,” “anti-VEGF,” “anti-inflammatory,” “immunosuppressant,” “tyrosine kinase inhibitors,” “laser photocoagulation,” “fine needle diathermy,” “photodynamic therapy,” “gene therapy,” as well as other relevant treatment modalities in the titles and abstracts. In addition, an independent search of current literature and a thorough screening of previous reviews were also conducted to maintain a comprehensive search and article inclusion. Both animal and human clinical studies were included in this review.

Pathogenesis

The human cornea is an optically important structure. Together with the tear film, they contribute to 70% of the eye’s total refractive power. It is made up of six distinct layers – the epithelium, Bowman’s membrane, stroma, Dua’s layers, Descemet’s membrane, and the endothelium.[2,3] The stroma contains type I and V collagen fibrils interwoven in a regular fashion, forming layers of stacked lamellae to create an optically transparent media for light to pass through. CoNV is postulated to occur via a disruption in corneal angiogenic privilege due to a disequilibrium in proangiogenic and antiangiogenic factors in the cornea.

Proangiogenic factors involved in the pathogenesis of CoNV include vascular endothelial growth factor (VEGF), platelet-derived growth factor, matrix metalloproteinase (MMP), and inflammatory cytokines such as interleukin-1 (IL-1) and interleukin-6 (IL-6).[4] An extensively studied angiogenic mediator is VEGF-A, which is a member of a family of proteins including VEGF-A, VEGF-B, VEGF-C, VEGF-D, virally encoded VEGF-E, and placental growth factor.[5] VEGF-A is secreted by a wide variety of cells including macrophages, T-cells, fibroblasts, retinal pigment epithelial cells, and corneal cells upon inflammation and insult.[6] VEGF-A plays a dominant role in regulating angiogenesis by interacting with the tyrosine kinase receptors, VEGF-R1 and VEGF-R2.[7] VEGF-C and VEGF-D are secreted by the macrophages, and they bind to VEGF-R3 to stimulate lymphangiogenesis. MMPs are Zn2+-dependent enzymes categorized as either secreted MMPs or membrane-type MMPs. The first transmembrane-containing MMP to be identified was MMP-14. It has been linked to several molecular mechanisms involved in the selective binding and cleaving of VEGF-R1, which induces corneal angiogenesis.[8] MMP-2 and MMP-9 contribute to the degradation of extracellular matrix components and the upregulation of VEGF and tumor growth factor-β.[9] IL-1 and IL-6 are proinflammatory cytokines which upregulate the expression of chemokines and growth factors that lead to neovascularization.[10,11]

Antiangiogenic factors include endostatin and endostatin analogs (neostatin, arrestin, canstatin, and tumstatin), plasminogen inhibitors, serine protease inhibitors (angiostatin and pigment epithelial-derived factor), and soluble VEGF receptors.[12] Angiostatin binds to surface proteins in the vascular endothelial cells to hinder migration and tubule formation.[13] Soluble VEGF receptors block the downstream effects of VEGF ligands by depleting available VEGF molecules and preventing their attachment to membrane-bound VEGF receptors. Soluble VEGF-R1 has a high affinity to VEGF-A and is postulated to mediate corneal avascularity during development.[14] The presence of corneal insult or inflammation disrupts the equilibrium by upregulating proangiogenic factors and downregulating antiangiogenic agents, leading to corneal hemangiogenesis and lymphangiogenesis.

LSCD, a consequence of a dysfunctional limbal stem cell (LSC) population, can also lead to CoNV. LSC has the unique capacity to differentiate into normal corneal epithelial cells. Any damage in LSC leads to cellular apoptosis followed by replacement with abnormal conjunctival epithelial cells. Corneal stem cells play a vital role in maintaining the integrity of corneal surface. When these stem cells are damaged or deficient, the corneal surface becomes compromised, leading to the loss of barrier function that normally prevents blood vessels from growing into the cornea.[15] In LSCD, there is often an increase in inflammatory mediators such as cytokines and growth factors. These mediators create a proinflammatory environment in the cornea, promoting the infiltration of immune cells, which, in turn, release angiogenic factors promoting CoNV.[16] LSCD occurs when there are alterations in corneal tissue homeostasis due to depletion of LSC (such as in chemical injuries and mechanical trauma), absence of LSC (congenital aniridia), or in proinflammatory states (Steven–Johnson syndrome or contact lens–related hypoxia).[17]

Therapeutic approaches

Several treatment modalities have been derived based on the understanding of CoNV pathogenesis. Broadly, they can be categorized based on their mechanism of action. These include anti-inflammatory agents, anti-VEGF agents, and surgical vessel occlusion with laser or diathermy. Contemporary and experimental therapies such as gene therapy and nano-based drug therapy show promise in managing CoNV, but lack clinical studies to support their safety and efficacy. Treatment modalities studied in the literature are summarized in Tables 1–3. Key therapeutic approaches are evaluated below.

Table 1.

Summary of anti-inflammatory and anti-VEGF agents studied for corneal neovascularization

Agent Level of Evidence Mechanism of Action Efficacy and Remarks Studies
Anti-Inflammatory
Corticosteroids Animal studies
Prospective Clinical Studies
Inhibit phospholipase A2
Inhibit proliferation and migration of vascular endothelial cells
Low-cost, highly accessible
Limited regression of mature CoNV
More effective if started early after CoNV develops
Animal:
Hos et al., 2011,[120] Hoffart
et al., 2010[22]
Human: Cursiefen et al., 2001[20]
Long-term use associated with side effects such as
infection and glaucoma
NSAIDs Animal Studies Inhibition of COX-1/2 enzymes Differential effectiveness; COX-2 selective inhibitors (celecoxib, rofecoxib) have limited inhibition of VEGF production Animal:
Cooper et al., 1980,[121]
Pakneshan et al., 2008[122]
Long-term use is associated with side effects such as corneal ulceration and melting
Generally ineffective in treating CoNV
Cyclosporine A Animal Studies
Randomized Controlled Clinical Trial
Calcineurin inhibitor, limits T-Cell response
Inhibition of VEGF- induced primary endothelial angiogenesis
Topical, subconjunctival, and systemic administration has shown to be effective in inhibiting CoNV in experimental models
Inhibitory effect of topical cyclosporine A was found to be significantly more effective than bevacizumab in an immune-mediated rabbit CoNV model
Animal:
Bucak et al., 2013,[26] Ulusoy et al., 2020[123]
Human:
Bock et al., 2014[27]
Limited inhibition of CoNV in post-keratoplasty patients
Tacrolimus Animal Studies Immunosuppressor, calcineurin inhibitor that suppresses T-cell/B-cell activation and proliferation Subconjunctival tacrolimus was shown to be more effective than anti-rat- VEGF in an alkali-burn rat model
Topical application is limited by its poor corneal permeability
Animal:
Chen et al., 2018[31]
Methotrexate Animal Studies Anti-metabolite; Mechanism not well understood
Anti-inflammatory response through reduction of chemokine- induced VCAM
Topical and subconjunctival administration effective in suture-induced CoNV rabbit models with associated reduction of VEGF and IL-6 Animal:
Byun et al., 2011[124]
Anti-angiogenesis associated with inhibition of VEGF and bFGF, similar to thalidomide
Thalidomide/Thalidomide Analogue (CC-3052) Animal Studies Inhibition of VEGF, FGF-2, and TNF-α Effective inhibition of CoNV in experimental models, however, special caution should be exercised regarding dosage safety and efficacy Animal:
Kruse et al., 1998,[125] Abbas et al., 2002,[126]
Lee et al., 2013[127]
Side effects appear to be lower with thalidomide analogues (teratogenicity)
Interleukin Receptor Inhibitors (IL-1ra, IL-17 mAb, tocilizumab) Animal Studies Inhibition of pro- angiogenic activity of Interleukins
Promote angiogenesis either directly – activation of endothelial cells, or indirectly – macrophage polarization and VEGF secretion
IL-1ra was associated with significant inhibition of CoNV in early initiation of therapy after induction of CoNV in a murine model
Efficacy of CoNV inhibition in subconjunctival tocilizumab was comparable to that of bevacizumab in a suture- induced rabbit model
Animal:
Dana et al., 1998,[128] Yoo et al., 2014[35]
TNF-α Inhibitors (Infliximab, Enterecept, adalimumab) Animal Studies Inhibition of TNF-α, a pro- inflammatory cytokine responsible for corneal lymphangiogenesis through the induction of VEGF-C production by macrophages
TNF-α is also responsible
for the expression of angiogenic factors
Experimental models displayed the inhibitory effects of TNF-α Inhibitors on CoNV
Effectiveness of subconjunctival etanercept is enhanced when used in conjunction with bevacizumab in a rat silver nitrate cauterization model
Subconjunctival adalimumab more effectively reduced VEGF expression than bevacizumab in a rat silver/potassium nitrate
cauterization model
Animal:
Ozdemir et al., 2013,[34] Özkaya et al., 2023[129]
Tyrosine Kinase Inhibitors (Sorafenib, Semaxanib, Rivoceranib, Regorafenib, Lapatinib, Axitinib, Dovitinib, Dasatinib, Sunitinib, Vatalanib, Nilotinib, Pazopanib, ZK261991, AG 1296) Animal Studies
Prospective Clinical Studies
Multitarget/Selective TKI in signaling pathways Effectiveness comparable to corticosteroids and anti- VEGF
Requires further evaluation of therapeutic effect and long-term safety in clinical studies
Animal:
Onder et al., 2014[36]
Human: Amparo et al., 2013[130]
Transforming growth factor-β-activated kinase 1 (5Z-7-oxozeaenol) Animal Studies Inhibits cell proliferation via suppression of cell cycle and DNA replication
Inhibition of TNFα- mediated NFκB signaling
Topical administration of 5Z-7-oxozeaenol suppresses corneal neovascularization in a mouse model Animal:
Wang et al., 2022[131]
Effect is enhanced when gelatin- nanoparticles- encapsulated 5Z-7- oxozeaenol are administered, likely due to the extended retention of the drug in the cornea
Anti-VEGF
Bevacizumab Animal Studies Humanized anti-VEGF-A monoclonal antibody Prolonged topical use associated with loss of integrity of corneal epithelium Animal:
Manzano et al., 2007[43]
Randomized Controlled Clinical Trials Large molecular weight and size may impede corneal penetrance
Human:
Pooled meta-analysis suggests significant inhibitory effects of topical and subconjunctival bevacizumab on CoNV in both human and animal studies Dastjerdi et al., 2010,[132]
Petsoglou et al., 2013,[133]
Papathanassiou et al., 2013[48]
Meta-analysis
Ranibizumab Animal Studies
Prospective Clinical Studies
Recombinant humanized anti-VEGF-A monoclonal antibody
Lower molecular weight and size allows greater drug penetration than bevacizumab
Effectiveness in inhibiting CoNV in comparison to bevacizumab remains inconclusive, but ranibizumab appears to induce neovascular reduction earlier than bevacizumab in animal models Animal:
Kim et al., 2014,[134]
Liarakos et al., 2014[51]
Human:
Kim et al., 2013,[52] Ferrari et al., 2013[49]
Topical ranibizumab was found to inhibit CoNV mostly through the reduction of vessel caliber rather than invasion area Comparative inhibitory effect of ranibizumab and bevacizumab in clinical studies remains to be
further validated
Pegaptanib Animal Studies Oligonucleotide aptamer that selectively binds VEGF-165 Inconclusive treatment efficacy in both topical and subconjunctival administration in experimental animal models No inhibitory effect on corneal neovascularization was noted in a rabbit model when compared to topical prednisolone Animal:
Akar et al., 2013,[39] Andrade et al., 2021[53]
Aflibercept Animal Studies
Prospective Clinical Studies
Humanized recombinant fusion protein that binds VEGF and PlGF Inconclusive treatment efficacy in both topical and subconjunctival administration in comparison to other anti- VEGFs in animal models
No significant improvement of corneal neovascularization following subconjunctival administration of aflibercept in six patients
Animal:
Oliveira et al., 2010,[135] Eski et al. 2022[136]
Human:
Sella et al., 2021[58]
Conbercept Animal Studies
Case Series Studies
Humanized recombinant fusion protein that binds specifically to VEGF-B, PlGF and various isoforms of VEGF-A Topical, subconjunctival, and intrastromal conbercept was found to have statistically significant effect on CoNV regression in rabbit models Animal:
Liu et al., 2020,[137] Du et al., 2023[138]
Human:
Sun et al., 2023[139]
Effectiveness of CoNV inhibition has yet to be compared to alternative anti-VEGFs
Laser Photocoagulation Animal Studies
Prospective Clinical Studies
Heat-induced photocoagulation of corneal vasculature More commonly used in chronic CoNV, in part due to the limited efficacy of medical therapies on more established corneal vasculature
Repeated treatments are often required due to recanalization of corneal vessels
May cause damage of surrounding tissues
Animal:
Cherry et al., 1976[140]
Human:
Gordon et al., 2002,[141] Kumar et al., 2016[77]
Photodynamic Therapy Animal Studies
Prospective Clinical Studies
Excitation of photosensitizers to occlude corneal vessels through thrombus formation Relatively selective for corneal neovascular vessels with little damage to surrounding tissue
Repeated treatments are often required for extensive disease
High costs
Animal:
Primbs et al., 1998[142]
Human:
Al-Torbak et al., 2012,[143] Yoon et al., 2007[144]
Fine Needle Diathermy Animal Studies
Prospective Clinical Studies
Coagulating current used to occlude corneal vessels More effective than topical dexamethasone in mature CoNV and lymphatics regression in a sutured- induced CoNV mouse model
Corneal vascularization secondary to thermal
cautery may be reduced with combined anti-VEGF
Animal:
Le et al., 2018[85]
Human:
Romano et al., 2016,[145] Pillai et al., 2000[80]

Legend: CoNV, Corneal Neovascularization; NSAID, Non-Steroidal Anti-Inflammatory Drug; COX; Cyclooxygenase, VEGF, Vascular Endothelial Growth Factor; IL, Interleukin; VCAM, Vascular Cell Adhesion Molecule; FGF, Fibroblast Growth Factor; TNF, Tumor Necrosis Factor; IL-1ra, Interleukin-1 Receptor Antagonist; IL-17 mAb, Interleukin-17 Monoclonal Antibody; TKI, Tyrosine Kinase Inhibitors; PlGF, Placental Growth Factor; Nuclear Factor-κB, NFκB

Anti-inflammatory agents

Inflammation is a key driver of corneal angiogenesis and is mediated by the release of proangiogenic cytokines such as IL-1, tumor necrosis factor-α (TNF-α), and VEGF.[18]

Corticosteroids are potent inhibitors of inflammation and remain widely accepted as first-line therapy for CoNV. By inhibiting vascular endothelial cells, steroids may exert a direct antiangiogenic effect through inhibition of VEGF expression from the vascular smooth muscle cells.[19] They also inhibit the migration of inflammatory cells, such as macrophages and lymphocytes, into the cornea. Inflammatory cells release angiogenic factors that promote neovascularization. By limiting the infiltration of these cells, steroids indirectly inhibit the growth of new blood vessels. Furthermore, steroids stabilize newly formed blood vessels, making them less fragile and less prone to leakage. This stabilization effect can prevent further expansion of the abnormal corneal vascular complex. However, their antiangiogenic effects are weak and insufficient to induce regression of long-term, mature CoNVs.[20]

The efficacy of steroids in suppressing angiogenesis is maximized if they are started immediately following corneal insult. Delays in administration may render them ineffective against subsequent development of CoNV.[21] In a chemical cauterization rat model, early and immediate application of dexamethasone resulted in a significant reduction of new vessel size compared to the control group.[22] Roshandel et al. have suggested prompt and aggressive treatment of CoNV associated with inflammatory conditions such as herpes simplex virus, immune stromal keratitis, and corneal graft rejection with early, frequent corticosteroids, before tapering them slowly based on clinical response.[23] However, the optimal duration of treatment remains controversial. Cursiefen et al.[20] showed that there was no difference in CoNV extent between post-keratoplasty patients who received either 6 months or 12 months of topical steroid therapy. Long-term corticosteroid usage is also associated with ocular and systemic side effects.[24] Despite this, corticosteroids remain the mainstay of treatment due to their low cost, accessibility, and anti-inflammatory properties.

Cyclosporine is an immunomodulatory agent which inhibits the migration of primary endothelial cells. The mechanism behind its antiangiogenic effects are not well characterized, but is thought to be related to an overexpression of HESR1 transcription factor and a downregulation of VEGF-R2.[25] The role of cyclosporine in the treatment of CoNV remains controversial as conflicting evidence on its efficacy have been reported. In one study, topical cyclosporine A 0.05% was found to be more effective than topical bevacizumab 0.5% (24.4% vs. 37.1%, P = 0.03) but less effective than topical dexamethasone 0.1% (24.4% vs. 5.9%, P = 0.02) in reducing the extent of neovascularization in a rabbit cornea model.[26] On the contrary, a randomized, prospective, multicenter clinical trial did not find any significant difference in the incidence of CoNV following high-risk keratoplasty among patients receiving either 5.13 or 7.7 mg subconjunctival cyclosporine A implant when compared to placebo.[27] This was postulated to be related to the strong proangiogenic stimulus after high-risk transplantation and poor drug penetration into the anterior chamber following local administration of cyclosporine.[28]

Tacrolimus is a calcineurin inhibitor which decreases T-cell activation and inflammatory cytokine release. It has a higher potency than cyclosporine, exhibiting similar effects at concentrations 100 times lower.[29] It reduces the production of angiogenic factors such as VEGF, epidermal growth factor, platelet-derived growth factor, prostaglandin E2, TNF-α, MMP-9, MMP-13, IL-1, IL-6, and hypoxia-inducible factor.[30] In a corneal alkali burn model, topical tacrolimus 0.05% administered four times daily was more effective than subconjunctival anti-rat-VEGF (0.5 mg/0.05 ml) in decreasing CoNV after 1 month of therapy.[31] Corneal epithelial defects and opacities were also significantly reduced by topical tacrolimus. However, the efficacy of topically instilled tacrolimus is limited by poor drug penetration across the cornea.[32] Cationic liposomal tacrolimus has been reported to be effective in circumventing this problem. With a surface potential of approximately +30 mV, this formulation binds to the negatively charged mucin layer of the ocular surface to increase the concentration of tacrolimus in the anterior chamber of mice by 20-fold at 90 min post-instillation.[32] Furthermore, this new drug delivery vehicle was more effective than topical tacrolimus in inhibiting CoNV, reducing corneal inflammation, and shortening the duration required for corneal epithelial recovery. However, adherence of topical tacrolimus may be an issue. Some patients may experience temporary discomfort, burning, or stinging in the eyes after applying tacrolimus ointment. Tacrolimus ointment might exacerbate dry eye symptoms in some individuals. Prolonged use of topical tacrolimus may also increase the risk of ocular infection because of local immunosuppression.[33]

Immunomodulatory biologic therapies including IL-6 receptor antagonist, tyrosine kinase inhibitors, and TNF-α inhibitors have been shown in experimental studies to be effective in treating CoNV.[34,35,36] Subconjunctival tocilizumab, an IL-6 receptor antagonist, was as effective as subconjunctival bevacizumab in reducing the extent of CoNV.[35] Subconjunctival sunitinib, a tyrosine kinase inhibitor, reduced neovascularization area in experimental rat corneas at a level comparable to subconjunctival bevacizumab.[37] Subconjunctival etanercept, a recombinant TNF receptor which antagonizes both TNF-α and TNF-β, demonstrated anti-inflammatory and antiangiogenic effects in an animal model, and the effects were further enhanced when given in conjunction with subconjunctival bevacizumab.[34] However, further human clinical studies are required to ascertain their clinical safety and utility.

Anti-vascular endothelial growth factor

Anti-VEGF agents such as pegaptanib, bevacizumab, ranibizumab, and aflibercept have been administered off-label, via topical, subconjunctival, and intrastromal routes, to treat CoNV.[38,39,40,41] Among the anti-VEGF agents, bevacizumab is most commonly used to treat neovascular age-related macular degeneration and diabetic macular edema due to its widespread availability and cost-effectiveness. Bevacizumab is a large, full-length humanized monoclonal antibody that binds to all isoforms of VEGF-A. Owing to its large molecular weight of 149 kD, it is typically unable to penetrate the cornea. However, in experimental animal models, the pathologically vascularized stroma permits penetration of large immunoglobulin molecules and increases the viability of topical administration.[42] In a rat model of chemically induced CoNV, topical bevacizumab 4 mg/ml drops administered twice daily for 1 week was demonstrably more effective than placebo in reducing the area of CoNV (38.2% vs. 63.5%, P < 0.02).[43] A prospective clinical trial of short-term topical bevacizumab 10 mg/ml eyedrops administered two to four times daily for 3 weeks successfully reduced the area of neovascularization (47.5%, P < 0.001) and diameter of neovascular vessels (36.2%, P = 0.003) at 6 months.[44] Although no adverse ophthalmic or systemic events were reported, this study excluded patients with poor corneal epithelial integrity (either existing epithelial defects or previous corneal surgeries) and utilized a shorter treatment course with a lower concentration of topical bevacizumab, compared to other studies. In contrast, separate studies have reported corneal epitheliopathy, approaching 60% of the eyes which received topical bevacizumab 12.5 mg/ml over the course of 3 months, and development of epithelial defects in 16.7% of eyes after administration of topical bevacizumab 5 mg/ml for up to 1 year of therapy.[45,46] Both studies reported improvements in CoNV parameters. Despite improvements in neovascularization, there was no statistically significant improvement in visual acuity.[43,45,46]

Subconjunctival bevacizumab (2.5 mg/0.1 ml) administered monthly for up to 3 months in patients with CoNV has been demonstrated to reduce the extent and density of neovascular vessels (6.0 ± 1.2 clock hours before and 4.6 ± 1.0 clock hours after bevacizumab injection, P = 0.008).[47] A meta-analysis of seven clinical trials of subconjunctival bevacizumab showed a pooled reduction of 32% (95% confidence interval [CI]: 10%–54%) in the neovascularization area.[48] The pooled mean change in best corrected visual acuity in three eligible studies demonstrated a mean improvement of 0.04 log of minimum angle of resolution (logMAR; 95% CI: -0.01–0.09), but this improvement was not statistically significant. The analyses were limited by substantial heterogeneity among the studies (studies for neovascularization area: I2 = 92.8%, best correct visual acuity: I2 = 73%). Interestingly, this meta-analysis demonstrated that the efficacy of subconjunctival bevacizumab was comparable to that of topical bevacizumab, although this was limited by the small population and large heterogeneity between the studies.

Ranibizumab is a recombinant humanized monoclonal antibody that is affinity-matured to optimize VEGF-A binding potential. It has a smaller molecular weight of 48 kD, which theoretically allows better penetration of the cornea than bevacizumab. Topical administration of ranibizumab 10 mg/ml four times daily for 3 weeks has been reported to reduce the neovascularization extent (55.3%, P < 0.001) and the diameter of neovascular vessels (59%, P < 0.001) at 4 months.[49] Similar to studies on topical bevacizumab, there were no statistically significant changes in invasion area or visual acuity. No adverse ophthalmic or systemic events were reported, though patients with previous ocular surgery or corneal epithelial defects were excluded from the study. Comparison of the bevacizumab and ranibizumab studies with similar study population characteristics by Cheng et al. and Ferrari et al. demonstrated earlier improvements in vessel caliber and neovascularization area in the topical ranibizumab group, likely attributable to the smaller molecular size.[50] However, there were no statistically significant differences in neovascularization area, vessel caliber, or invasion area between the two medications at the study endpoint.

Subconjunctival and intrastromal administration of ranibizumab has also been investigated as a potential therapeutic option. In rabbit cornea models, subconjunctival ranibizumab (1 mg/0.1 ml) administered 1 h after alkali burns resulted in smaller vascularization areas at 2 weeks (control: 22.63 ± 4.03% vs. treated: 6.13 ± 4.22%, P = 0.001).[51] VEGF levels were also significantly lower in all the sampled anterior segment tissues including the cornea, iris, aqueous humor, and conjunctiva. The extent of cornea scarring area was, however, similar to the ranibizumab-treated and control corneas. A clinical randomized prospective study of 16 patients with chronic CoNV demonstrated that combined subconjunctival and intrastromal bevacizumab (2.5 mg/0.1 ml) and ranibizumab (1 mg/0.1 ml) both reduced the neovascularization area at 1 month.[52] Notably, bevacizumab resulted in greater reduction in neovascularization area compared to ranibizumab (28.4 ± 9.01% vs. 4.51 ± 11.64%, P = 0.001). Further prospective randomized controlled trials with larger study populations will be required to provide evidence on the long-term efficacy of bevacizumab versus ranibizumab.

Pegaptanib is an oligonucleotide aptamer that selectively binds to VEGF-165. It has shown variable efficacy in treating CoNV in animal models. Topical 0.5% and 1.0% pegaptanib sodium diluted in 15 ml of 0.5% carboxymethylcellulose sodium were both less effective than topical 1.0% prednisolone acetate in reducing neovascularization area, total vascular length, and number of neovascular vessels in a rabbit alkali burn model.[53] Another experimental rat model investigating the efficacy of subconjunctival bevacizumab, ranibizumab, and pegaptanib found that all three agents reduced the area of neovascularization, but bevacizumab produced a larger area of reduction compared to ranibizumab and pegaptanib.[39]

Aflibercept is a soluble fusion protein of VEGF receptors 1 and 2.[54] It is a relatively newer anti-VEGF compared to bevacizumab and ranibizumab, with a wider range of target molecules including VEGF-A, VEGF-B, and placental growth factor.[55] While an animal study showed that topical aflibercept 2 mg/0.5 ml and 2 mg/5 ml were effective in reducing neovascularization surface area, the effect was comparable to topical bevacizumab 2.5 mg/1 ml.[40] In another rat model of chemically induced CoNV, subconjunctival administration of bevacizumab (1.25 mg/0.05 ml) or ranibizumab (0.5 mg/0.05 ml) or aflibercept (1.25 mg/0.05 ml) was effective in inhibiting CoNV, inflammation, fibroblast activity, and reduced VEGF levels in neovascularized arteries.[56] No significant differences in efficacy were noted among the three medications. A comparison of topical aflibercept and topical bevacizumab in corneal chemical burn rat models demonstrated smaller areas of neovascularization at day 7 and day 10 in rat corneas receiving topical aflibercept compared to topical bevacizumab (aflibercept: 21.73 ± 14.59% on day 7 and 31.0 ± 23.61% on day 10 vs. bevacizumab: 51.27 ± 15.50% on day 7 and 54.4 ± 11.33% on day 10, P < 0.001).[57] Clinically, a single subconjunctival injection with 0.08 ml of aflibercept (25 mg/ml) in six eyes with CoNV did not demonstrate significant changes in the extent, density, or centricity of corneal blood vessels up to 3 months after treatment.[58] Further studies are required to ascertain the effectiveness of pegaptanib and aflibercept in treating CoNV.

Intrastromal anti-VEGF therapy was proposed as a more effective drug administration route for CoNV due to the delayed drug elimination from the avascular cornea. Multiple case series and clinical studies have demonstrated the efficacy of intrastromal injections in regression of neovascularization and sustained reduction of VEGF levels.[59,60,61,62] In a suture-induced rabbit cornea neovascularization model, intrastromal bevacizumab (1.25 mg/0.05 ml) and intrastromal aflibercept (2 mg/0.05 ml) reduced CoNV more effectively at 2 weeks (88.1% and 82.5%, respectively) compared to subconjunctival bevacizumab (1.25 mg/0.05 ml) or subconjunctival aflibercept (2 mg/0.05 ml) (64.5% and 69.9%, respectively).[41] However, intrastromal injections may be associated with procedural complications such as Descemet’s membrane perforation or detachment.

Complications associated with topical anti-VEGF therapy include corneal wound healing impairment that manifests as persistent epithelial defects and corneal stromal thinning.[45,46,63] A theoretical side effect is neurotrophic keratopathy due to inhibition of VEGF-mediated neural growth.[64] However, this has yet to be observed clinically. Risk factors for development of corneal epitheliopathy include the use of high concentrations of bevacizumab (>10 mg/ml), extended duration of therapy, pre-existing corneal epithelial defects, and previous corneal surgeries. No systemic complications of topical anti-VEGF, such as cerebrovascular accidents and thromboembolic events, have been reported in the limited number of clinical studies published to date. Nonetheless, it is prudent to take precautions to reduce systemic absorption of the drug, such as performing punctal occlusion or applying punctal plugs before topical administration.[50] Subconjunctival injection of anti-VEGF is safe and well tolerated, with no significant side effects reported in human or animal studies. Common adverse events include subconjunctival hemorrhage (28%), pain (31%), anterior chamber reaction (8%), and irritation (3%).[48,65,66,67]

Although current studies show promise in treating CoNV, anti-VEGF agents only produce a partial and temporary reduction in neovascularization, with most effects occurring within a month of commencing therapy with progressive waning over the subsequent 2 months.[68] Neovascularization may also recur after successful treatment with subconjunctival bevacizumab, necessitating repeated injections.[69] While there are anecdotal reports of subconjunctival bevacizumab successfully treating CoNV, these reports are rare and confounded by the concomitant or preceding administration of other therapeutics such as corticosteroids.[70,71,72] In addition, there is a paucity of long-term clinical outcomes, with most studies reporting follow-up of up to 3 months. The advent of new anti-VEGF drugs, such as brolucizumab and faricimab, further expands the armamentarium of therapies available. Therefore, further large-scale studies and long-term evaluation of anti-VEGF agents are required to ascertain their effectiveness and safety in treating CoNV.

Physical ablation of neovascular vessels

Medical therapy may have limited effectiveness in treating chronic CoNV with mature and well-established vascularization. In such cases, physical occlusion of neovascular vessels can be considered with laser photocoagulation, photodynamic therapy, or FND.

Laser photocoagulation using argon, yellow dye, and Nd:YAG neodymium:yttrium-aluminium-garnet laser has been shown to be effective in treating CoNV in animal[73,74,75] and clinical studies.[76,77] Although laser photocoagulation can obliterate efferent neovascular vessels, as they are wider and have relatively slower blood flow, it is often unable to cauterize narrower and deeper afferent vessels with faster blood flow. A single prospective study of 40 patients with CoNV receiving double-frequency Nd:YAG laser photocoagulation demonstrated improvements in the mean area of CoNV from 31.9% to 17.6% after 3 months of laser treatment.[77] Of 185 corneal vessels, 53.5% were completely occluded, 9.2% were partially occluded, and 37.3% were recanalized at the end of 3 months. Hence, multiple laser photocoagulation sessions may be required for patients with extensive CoNV in view of the possibility of recanalization. Side effects associated with laser photocoagulation include corneal endothelium or crystalline lens damage, corneal hemorrhage, corneal thinning, and iris atrophy.[77]

Photodynamic therapy achieves vessel occlusion via reactive oxygen species generation, which arise from the interaction between light energy and photosensitizers. In the treatment of CoNV, photodynamic therapy with intravenous or intrastromal verteporfin was effective in inducing angiogenic and lymphogenic regression in a rodent suture-induced CoNV model.[78,79] Long-term allograft survival was also improved significantly in photodynamic therapy-treated eyes when compared to controls.[78] A potential advantage of using photodynamic therapy is its selectivity for neovascular vessels, allowing it to induce vascular regression with minimal damage to surrounding tissues.

FND involves the use of high-frequency electrical currents to generate heat. The tip of the fine needle is heated using diathermy and subsequently applied in close proximity to the abnormal blood vessels. The heat generated coagulates the blood vessels and stops blood flow, leading to their closure.[80] Subsequent studies demonstrated that occlusion of corneal vessels using FND among pre-keratoplasty patients and patients with lipid keratopathy was successful in reducing lipid deposition in 82.3% of treated eyes, with 84.6% of high-risk corneal grafts surviving beyond 1 year.[81] Unfortunately, corneal diathermy may induce further CoNV due to thermal damage. In a mouse suture-induced CoNV model, FND monotherapy inhibited angiogenesis at early time points, but was associated with secondary corneal angiogenesis, with elevated proangiogenic factors such as VEGF-A, VEGF-C, and VEGF-D.[82] Hence, combination therapy with anti-VEGF agents has been proposed to complement its usage. In the same study, addition of anti-VEGF agents with FND significantly regressed corneal blood and lymphatic vessels at 1 week and prevented the undesirable effect of angiogenesis observed in the monotherapy group.[82] Clinically, FND combined with subconjunctival bevacizumab treatment before penetrating keratoplasty (PK) in patients with at least one quadrant of CoNV resulted in rejection-free graft survival rates of more than 90% after 1 year and 78% after 3 years.[83] A retrospective review of children who received FND with subconjunctival bevacizumab reported complete CoNV resolution in 88.9% within 1 month of treatment and an improvement in mean corrected distance visual acuity from 0.66 ± 0.31 to 0.50 ± 0.37 logMAR (P = 0.02).[84] Combination of FND and corticosteroids has also been shown to regress both blood vessels and lymphatic vessels in animal models.[85] However, no clinical studies are available to support the efficacy of this combination. Another strategy to reduce excessive thermal damage is to selectively cauterize afferent arterioles, which comprise < 1% of the neovascular vessels.[86] Spiteri et al. identified afferent neovascular vessels with fluorescein and indocyanine green corneal angiography and performed selective FND. They showed that the number of afferent vessels ranged between one and three and that the vessels had a mean diameter of 40 µm. After selective FND, the mean area of neovascularization was reduced by 1.80 ± 1.40 mm2 (P < 0.01) up to 12 weeks postoperatively, demonstrating the viability of this protocol in reducing CoNV while minimizing thermal damage.[87]

LSC transplantation

LSCD is an ocular surface disease caused by abnormal LSCs and can lead to CoNV. Management is challenging and medical therapy is only effective for early or partial LSCD.[88] Surgical management of LSCD can be categorized into allograft, autograft, and cultivated LSC transplantation. LSC grafting techniques include autologous limbal transplant, allogenic limbal transplant, keratolimbal allograft, and simple limbal epithelial transplantation (SLET). A meta-analysis of 40 studies with 2202 eyes demonstrated that autologous limbal transplantations were associated with greater visual acuity improvement (76%) than allogenic limbal transplants (52.3%), cultivated autologous LSC transplants (56.4%), and cultivated allogenic LSC transplants (43.3%) (P < 0.001).[89] SLET utilizes a small limbal tissue autograft that is transplanted from the healthy eye to the diseased eye.[90] This is accomplished by dividing the strip of donor limbal tissue into small pieces and distributing them over an amniotic membrane placed over the diseased cornea. The outcomes of SLET have been favorable and are likely comparable to those of cultivated LSCs – 83% of reported SLET operations in the literature restored the defective corneal epithelium and 69% resulted in improvements in visual acuity.[91] Common complications reported in these studies following SLET include focal recurrence of LSCD, progression of conjunctivalization, progressive symblepharon, and keratitis.[91] No large-scale head-to-head comparison studies of SLET and other LSC transplantation techniques have been performed.

Mitomycin intravascular chemoembolization

Mitomycin C (MMC) intravascular chemoembolization (MICE) is an emerging technique used to treat CoNV and lipid keratopathy. MMC inhibits vascular endothelial cell proliferation and hinders neovascular vessel growth and repair.[92] Mimouni and Ouano described a case series of three patients who received 0.05 ml of MMC (0.4 mg/ml) injected directly into a corneal neovascular vessel.[93] All three patients experienced a regression of neovascularization and lipid keratopathy over a follow-up period of 1 year, with no intraoperative or postoperative complications identified. To avoid possible unwanted loss of limbal vascularization, MMC should not be injected in the direction of the limbus. Another case series reported success in inducing regression of neovascularization in two cases of MICE performed in patients with failed PK.[94] Further clinical studies are warranted to elucidate the long-term outcomes and safety of this treatment.

Corneal cross linking

Corneal cross-linking (CXL) is a procedure that utilizes ultraviolet-A (UVA) and riboflavin (vitamin B2) to treat corneal ectasias.[95] CXL was shown in an in vivo rat model to be able to temporarily suppress suture-induced corneal hemangiogenesis and lymphangiogenesis.[96] This is postulated to occur via the combined effects of UVA and riboflavin – UVA irradiation has been suggested to produce an anti-lymphangiogenic effect at the cellular level, while riboflavin reduces the lipopolysaccharide-induced synthesis of inflammatory cytokines TNF-α, IL-1, and IL-6.[97,98] UVA activation of riboflavin further releases reactive oxygen species, which is theorized to reduce neovascular vessel formation.[99] Further short-term observations of corneal parameters in animal studies displayed a partial effectiveness in treating CoNV. Xu et al. studied 60 rabbits with alkali burn-induced CoNV, which were divided into six groups.[100] Group A (control) was not given any treatment, while groups B, C, and D received 30, 15, and 45 min of CXL immediately after injury, respectively. Groups E and F received CXL for 30 min on day 1 and 3 after the injury, respectively. CXL was performed with riboflavin 0.1% drops dissolved in dextran 20% applied every 2–5 min for 30 min, and with continual administration during UVA irradiation once every 3 min. Irradiation with UVA was performed at an irradiance of 3 mW/cm2 and a surface dose of 5.4 J/cm2. Over the course of 2 weeks, the extent of CoNV increased in a time-dependent manner across all groups. All corneas treated with CXL displayed less neovascularization than the control group, with the most significant reduction observed in corneas receiving 45 min of UVA irradiation. In a separate rabbit alkali burn model, comparison of CXL using Dresden protocol with matrix-regenerating agent also showed improvement in epithelial loss, stromal edema, corneal vascularization, and leukocytic infiltration in both groups.[101] CXL was, however, not able to reduce corneal opacification from alkali burns in animal models.[101,102,103] In a retrospective case series involving five patients, CXL was performed before or in combination with high-risk PK in patients with thin or fragile corneas. While these results may be affected by the accompanying PK, reduction of CoNV was observed in all patients (mean 70.5% ± 22.7%) and revascularization was not observed over a mean follow-up of 16.4 weeks.[104] CXL remains an experimental treatment for CoNV, and additional studies relating to the optimal riboflavin dose and irradiation regimen are required.

Gene therapy

Gene therapy refers to the transfer of genetic material to facilitate genomic augmentation, suppression, or repair, with the aim of providing durable, long-term therapeutic expression or suppression of the edited gene.[105] This attempts to overcome the drawback of conventional pharmacological approaches, where the effects are temporary and require repeated treatment. Gene therapy has been studied to treat various anterior segment diseases such as dry eye disease, corneal and conjunctival scarring, epithelial wound healing, and improve corneal graft survival.[106]

Multiple experimental animal models targeting the angiogenic VEGF pathway via gene transfer of VEGF receptors, Flt-1 and Flk-1, have shown variable success in regressing CoNV. Gene transfer of Flt-1 using recombinant adeno-associated virus and non-viral micellar nano vectors, as well as recombinant adenovirus-mediated delivery of soluble Flk-1 produced favorable inhibition of CoNV.[107,108,109] Subconjunctival administration of lipoplexes carrying the VEGF and roundabout4 transcription factor, GA-binding protein gene, produced temporary improvement on CoNV.[110] Downregulation of VEGF via recombinant adenovirus-driven antisense VEGF RNA and nanoparticle-mediated delivery of shRNA VEGF-A plasmids reduced CoNV.[111,112] Techniques such as adeno-associated virus and equine infectious anemia virus-based gene therapies to upregulate the antiangiogenic factors endostatin and angiostatin to inhibit VEGF and FGF-2 were effective in suppressing CoNV, immune cell infiltration, and corneal opacification.[113,114] More recently, downregulation of MMP-9 using lipid nanoparticles and the use of cholesterol-modified small interfering RNA targeting stromal cell-derived factor-1 have shown promise in treating CoNV.[115,116] Ocular delivery of small interfering RNA via pH-sensitive vehicles had comparable efficacy to that of ranibizumab.[117] The only Phase III clinical gene therapy study is that of aganirsen, an antisense oligonucleotide preventing insulin receptor substrate-1 expression, which has been shown to inhibit CoNV and reduce the need for transplantation in patients with keratitis.[118] Aganirsen has since been granted an Orphan Drug Designation by the US Food and Drug Administration for the prevention of corneal graft rejection in 2016.

Despite advancements, more research is required to address the challenges associated with gene therapy in CoNV. Gene therapy has very limited clinical evidence available regarding its efficacy and side effects. The prominent concern relates to the risk of developing host immune responses that may lead to the generation of neutralizing antibodies against vectors carrying the therapeutic gene, which may reduce therapeutic efficacy. Another concern is the development of unwanted, “off-target” effects both locally and systemically. However, these side effects theoretically carry a lower risk in the case of corneal diseases, due to its inherent immune privilege and avascular environment. Finally, the high cost of research and development associated with gene therapy represents a major hurdle before preclinical studies can progress to clinical trials.

Nanoparticle-based therapy

Nanomedicine and nano-based drug delivery therapy is a new but rapidly developing field, where materials of the nanoscale are employed to enhance drug delivery to provide better therapeutic efficacy. The potential functions of nanoparticles in the treatment of CoNV are wide ranging, but can be broadly classified into three groups: (1) enhancing drug delivery via nanocarriers, (2) acting as vehicles for gene therapy, and (3) functioning as therapeutic agents.[119] Nanoparticle-based therapies studied in the literature are summarized in Table 2. Notably, these therapies are still in the preclinical research stage and their clinical safety and efficacy remains to be determined. Further work is required to determine the potential toxicity of nanoparticles as well as to overcome the high cost of production associated with the large-scale production for clinical use. Nonetheless, nanoparticle-based therapy remains a promising and viable method to treat CoNV.

Table 2.

Summary of surgical, gene-based and nano-based delivery therapy modalities studied for corneal neovascularization

Agent Level of Evidence Mechanism of Action Efficacy and Remarks Studies
LSC
Transplantation
Animal Studies Randomized Controlled Clinical Trial Meta-analysis Transplantation of allograft, autograft or cultivated LSC In a LSC deficient rabbit model, LSC with limbal stromal stem cell transplantation was significantly more effective than LSC transplantation at CoNV regression
Comparative studies on the effectiveness of different graft types and transplant procedures remain inconclusive
Animal: Zhu et al., 2022[146]
Human: Zakaria et al., 2014,[147] Le et al., 2020[148]
MICE Case Series Ablation of CNV with endothelial cell proliferation inhibition by MMC Effective in reducing corneal neovascularization in the described clinical case series
Further assessment is required for the safety and efficacy of MMC
Human: Mimouni et al., 2022,[93]
Addeen et al., 2023[94]
Corneal Cross- Linking Animal Studies
Case Series
Riboflavin inhibition of TNF-α, IL-1, and IL-6
synthesis
ROS generation associated with riboflavin activation by UV-A irradiation opposes angiogenesis and lymph angiogenesis
Effectiveness of CoNV inhibition observed in animal models and even in patients with progressive CoNV in need of high risk-keratoplasty
Further studies on optimal dosage and regimen required
Animal:
Xu et al., 2018,[149]
Kesim et al., 2021,[150]
Colombo- Barboza et al., 2014[103]
Human: Schaub et al., 2021[104]
Gene Therapy
Aganirsen Randomized Controlled Clinical Trial IRS-1 mRNA expression inhibiting antisense oligonucleotide Phase III study reported safety and efficacy of aganirsen
Reduced need for transplantation in viral keratitis and central corneal neovascularization
Human: Cursiefen et al., 2014[118]
Nano-based Delivery Therapy
Nanocomposite Hydrogels Animal Studies Different NPs embedded in hydrogels to capitalize on biomaterial properties that improve drug solubility, penetration and inhibitory effect on CoNV Subconjunctival injection of mesoporous silica nanoparticles integrated thermogel loaded with bevacizumab and cyclosporin showed significant inhibition of corneal neovascularization as compared to individual bevacizumab and cyclosporin therapies Animal: Lyu et al., 2021[151]
Liposomal NP Animal Studies Phospholipid bilayer membranes capable of carrying both hydrophilic and hydrophobic molecules
Enhances drug penetration and retention to improve therapeutic efficacy of vehicle drug
Therapeutic effect and drug retention time of cationic liposomes encapsulating tacrolimus were better than commercial tacrolimus eyedrops and the free drug at CoNV inhibition Animal: Lin et al., 2021[32]
Polymeric Micelles Animal Studies Hydrophobic fatty acid core with a hydrophilic outer layer
Smaller volume than liposomes may enhance corneal drug penetrance
In a corneal transplantation rat model, CoNV inhibitory effect of topical cyclosporine A micelle formulation was comparable to systemic cyclosporine A and significantly greater than the control group Animal: Di Tommaso et al., 2012[152]
Natural/Synthetic Polymeric NP Animal Studies Sub-micro colloidal particles formulated for enhanced drug penetration, bioavailability, biodegradability, and non-toxicity Kaempferol-loaded gelatin NPs was significantly more effective than kaempferol solution (poorly soluble flavonoid) at inhibiting corneal vessels formation in a mouse model
In chemically induced cornea neovascularization, mice corneas treated with gp91 peptide-encapsulated gelatin nanoparticles –an anti- angiogenic nicotinamide adenine dinucleotide phosphate oxidases
Animal: Chuang et al., 2019,[153]
Chu et al., 2023[154]
Zhang et al., 2018[155]
inhibitor, had a significant reduction in neovascular vessel area (8%) when compared to phosphate-buffered solution (34%) and gp91 (20%)
Subconjunctival bevacizumab- encapsulated polylactide-co-glycolide had significantly greater inhibitory effect on corneal neovascularization than
bevacizumab in an alkali burn mouse model
Inorganic Nanoparticles Animal Studies Submicron inorganic particles utilized for drug delivery
Downregulation of VEGFR-2 expression in Gold NPs Anti-inflammatory action by the ROS scavenging properties of Ce NPs
Gold NP resulted in significant reduction in corneal neovascularization in a mouse model compared to control
Ce NPs had comparable effectiveness in corneal neovascularization reduction to dexamethasone in an alkali mouse model
Potential toxicity due to non- biodegradable nature
Animal: Cho et al., 2015,[156]
Zheng et al., 2019[157]
Nanoemulsions Animal Studies Immiscible oil and water layers with accompanying active surfactants that allow for enhanced stability and sustained delivery of hydrophobic drugs In an alkali-burn mouse model, topical application of eNano-Ro5 significantly reduced corneal neovascularization as compared to the control group, but was slightly less effective than topical dexamethasone
Multicomponent nature requires extensive research and complicates manufacturing processes
Animal: Delgado- Tirado et al., 2022[158]
Dendrimers Animal Studies Polymeric NPs with a regular branched structure where surface groups can increase drug solubility and stability
Modifiable chemical parameters by altering surface groups allow for further drug development
Subconjunctival injectable dendrimer- dexamethasone gel significantly limited the extent of corneal neovascularization as compared to free dexamethasone in an alkali burn rat model Animal: Soiberman et al., 2017[159]
Nanowafers Animal Studies Nano-sized polymer with arrays of drug-filled nano reservoir to enable sustained drug release and absorption Axitinib nanowafer was more effective than topical axitinib in inhibiting corneal neovascularization in a murine ocular burn model Animal: Yuan et al., 2015[160]

LSC, Limbal Stem Cell; MMC, Mitomycin Intravascular Chemoembolization; MMC, Mitomycin; TNF-α, Tumour Necrosis Factor α; IL, Interleukin; ROS, Reactive; UV-A, Ultraviolet A; IRS-1, Insulin Receptor Substrate 1; mRNA, messenger Ribonucleic Acid; NP, Nanoparticle; VEGFR, Vascular Endothelial Growth Factor Receptor; Ce, Cerium oxide; eNano-Ro5, Nanoemulsion containing Ro5-3335

Table 3.

Summary of other treatment modalities studied for corneal neovascularization

Agent Level of Evidence Mechanism of Action Efficacy and Remarks Studies
Combination Therapy
Corticosteroids and Heparin Animal Studies
Case Reports
Corticosteroid inhibition of phospholipase A2 and subsequent inflammatory response Effectiveness of combination therapy remains to be further evaluated in comparative clinical studies Animal: Nikolic et al., 1986,[161]
Aydin et al., 2008[162]
Mechanism of heparin in reducing neovascularization not well understood; possibly via direct modulatory effect on angiogenic growth factors, or enhancing uptake of Human: Tommila et al., 1987[163]
corticosteroids into endothelial cells
Corticosteroids and Doxycycline Animal Studies Corticosteroid inhibits phospholipase A2 and subsequent inflammatory response
Doxycycline inhibits MMP
Combination of triamcinolone acetonide and doxycycline was more effective at CoNV inhibition than the individual therapies Animal: Aydin et al., 2008[162]
Corticosteroids and anti- VEGF Animal Studies Inhibition of angiogenic factors and proinflammatory cytokines like VEGF and IL-6 Effectiveness of the combination of corticosteroids and anti- VEGF did not differ significantly from individual therapeutics
Remains to be further evaluated in studies with
larger sample sizes and longer follow-up time
Animal: Kang et al., 2010,[164]
Hoffart et al., 2010[22]
NSAID and anti-VEGF (Diclofenac and Bevacizumab via thermosensitive hydrogel) Animal Studies Dual delivery of diclofenac and bevacizumab by the thermosensitive hydrogel (Poly (dl-lactide)-poly (ethylene glycol)-poly (dl-lactide) Proposed hydrogel caused minimal toxicity to tested cell lines at concentrations ranging from 0.05 mg/ml to 0.8 mg/ml with good ocular biocompatibility after a single subconjunctival injection Animal: Shi et al., 2022[165]
Superior anti-angiogenic effects as compared to
subconjunctival bevacizumab injection alone
TNF-α inhibitor and anti- VEGF Animal Studies Inhibition of TNF-α, a pro- inflammatory cytokine responsible for corneal lymphangiogenesis through the induction of VEGF-C production by macrophages Simultaneous inhibition of TNF-α and VEGF was more effective in CoNV inhibition than monotherapies Animal: Ozdemir et al.,2013,[34]
Zhou et al., 2023[166]
TNF-α is also responsible for the expression of angiogenic factors
Laser Photocoagulation and anti-VEGF Case Reports Heat-induced photocoagulation of corneal vasculature
Inhibition of VEGF
Clinical effectiveness of the combination therapy exhibited in multiple case reports
Effectiveness yet to be established in a comparative clinical study
Human: Gerten et al., 2008,[167]
Anand et al., 2018,[168]
Donato et al., 2021[169]
PDT and anti-VEGF Animal Studies
Case Series Studies
Excitation of photosensitizers to occlude corneal vessels through thrombus formation
Inhibition of VEGF
Combination therapy was more effective than Individual monotherapies in a suture- induced rabbit model and preliminary clinical studies Anti-VEGF agents suppress VEGF release post-PDT, reducing the risk of angiogenesis and recurrence Animal:
Kim et al., 2016[170]
Human: Hamdan et al., 2015,[171] Yoon et al., 2019[172]
FND and anti-VEGF Animal Coagulating current used to FND and bevacizumab Animal:
Studies occlude corneal vessels combination successfully Le et al.,
regressed mature CoNV in 2020[82]
Case Inhibition of VEGF patients previously
Series unresponsive to Human:
Studies corticosteroids Koenig et al.,
2012,[173]
Elbaz et al.,
2015,[84]
Mestanoglu et al.,
2022[174]
Other Therapies
Descemet Membrane Case Selective removal and Single study revealed a Human:
Endothelial Keratoplasty Series transplantation of Descemet significant regression in Hayashi et al.,
Studies membrane and endothelium corneal neovascularization 2021[175]
postoperatively
Mesenchymal Stem Cell Therapy Animal Source of progenitor cells Efficacy, bio-variability, and Animal:
Studies Anti-inflammatory properties safety to be further evaluated Yao et al.,
in clinical studies 2012,[176]
Song et al.,
2018[177]
Tetracyclines (Doxycycline, Minocycline) Animal Studies
Case
Series Studies
MMP and PI3K/Akt-eNOS pathway inhibition
MMP, ERK1/2 and Akt pathways inhibition
Lesser side effects than Anti- VEGF and corticosteroids Animal:
Aydin et al., 2008,[162] Xiao
et al., 2012[178]
Other Antimicrobials (Tigecycline, Itraconazole, Dihydroartemisinin) Animal Studies Tigecycline inhibits MMP Itraconazole inhibits cholesterol biosynthesis
Dihydroartemisinin inhibits VEGF, ERK1/2 and p38 expression
Effectiveness in CoNV inhibition displayed in experimental models
Requires further evaluation of therapeutic effect in clinical studies
Animal: Zhong et al., 2011,[179]
Goktas et al., 2014[180]
Flavonoids Animal Anti-angiogenic properties: Variable effectiveness of Animal:
(Epigallocatechin Studies Mechanism not well individual flavonoids in Li et al,
Gallate, Kaempferol, understood, may be different experimental 2018,[181]
Fisetin, Luteolin, associated to corticosteroid- models of CoNV Chuang et al.,
Genistein, Naringenin, like molecular structure 2019[153]
Quercetin, Coumarin
Esculetin)
Non-Flavonoid Animal Anti-angiogenic properties Variable bioavailability may Animal:
Phytochemicals Studies Mechanism not well limit the effectiveness Bian et al.,
(Curcumin, Resveratrol, understood 2008,[182] Kim
Withaferin A, Xanthatin, et al. 2010,[183]
Triptolide,
Thymoquinone,
Glycyrrhizin)
Vitamins Animal Suppresses angiogenic factors such as VEGF and MMP Requires further evaluation of therapeutic effect in clinical studies Animal:
(Ascorbic Acid, Studies Suzuki et al.,
Riboflavin, 1α,25- 2000,[184] Lee
dihydroxyvitamin D3) Mechanism not well et al., 2012[185]
understood

MMP, Matrix Metalloproteinase; VEGF, Vascular Endothelial Growth Factor; NSAID, Non-Steroidal Anti-Inflammatory Drug; IL, Interleukin; TNF-α, Tumor Necrosis Factor α; PDT, Photodynamic Therapy; FND, Fine Needle Diathermy; PI3K/Akt-eNOS, Phosphatidylinositol 3-Kinase/Akt-endothelial Nitric Oxide Synthase; ERK1/2, Extracellular Signal-Regulated Kinase 1/2

Conclusion

CoNV is a common complication of severe corneal pathologies that can result in visually significant disabilities. Recent advances in our understanding of its pathogenesis, aided by developments in imaging and pharmacological approaches, have led to the development of a range of treatment strategies. While conventional therapy has focused on reducing inflammation and targeting the VEGF pathway, an expanding array of pharmacological agents, such as tyrosine kinase inhibitors, biologic immunomodulators, and gene therapy, may be useful in improving treatment efficacy. Moreover, targeting multiple angiogenic pathways in the form of combination therapy could produce a synergistic effect and should be considered in future drug and clinical trials. However, further research, rigorous clinical trials, and long-term follow up studies are essential to refine these protocols and ensure their safety and efficacy. As our knowledge continues to expand and innovative therapies emerge, the outlook for patients with CoNV is increasingly promising.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

Acknowledgements

We thank Yishan Wu for her assistance with the illustrative material.

References

  • 1.Lee P, Wang CC, Adamis AP. Ocular neovascularization: An epidemiologic review. Surv Ophthalmol. 1998;43:245–69. doi: 10.1016/s0039-6257(98)00035-6. [DOI] [PubMed] [Google Scholar]
  • 2.Sridhar MS. Anatomy of cornea and ocular surface. Indian J Ophthalmol. 2018;66:190–4. doi: 10.4103/ijo.IJO_646_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dua HS, Faraj LA, Said DG, Gray T, Lowe J. Human corneal anatomy redefined: A novel pre- Descemet’s layer (Dua’s layer) Ophthalmology. 2013;120:1778–85. doi: 10.1016/j.ophtha.2013.01.018. [DOI] [PubMed] [Google Scholar]
  • 4.Qazi Y, Maddula S, Ambati BK. Mediators of ocular angiogenesis. J Genet. 2009;88:495–515. doi: 10.1007/s12041-009-0068-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: Beyond discovery and development. Cell. 2019;176:1248–64. doi: 10.1016/j.cell.2019.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ahmad A, Nawaz MI. Molecular mechanism of VEGF and its role in pathological angiogenesis. J Cell Biochem. 2022;123:1938–65. doi: 10.1002/jcb.30344. [DOI] [PubMed] [Google Scholar]
  • 7.Alitalo K, Tammela T, Petrova TV. Lymphangiogenesis in development and human disease. Nature. 2005;438:946–53. doi: 10.1038/nature04480. [DOI] [PubMed] [Google Scholar]
  • 8.Han KY, Chang JH, Lee H, Azar DT. Proangiogenic interactions of vascular endothelial MMP14 with VEGF receptor 1 in VEGFA-mediated corneal angiogenesis. Invest Ophthalmol Vis Sci. 2016;57:3313–22. doi: 10.1167/iovs.16-19420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Webb AH, Gao BT, Goldsmith ZK, Irvine AS, Saleh N, Lee RP, et al. Inhibition of MMP-2 and MMP-9 decreases cellular migration, and angiogenesis in in vitro models of retinoblastoma. BMC Cancer. 2017;17:434. doi: 10.1186/s12885-017-3418-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ebihara N, Matsuda A, Nakamura S, Matsuda H, Murakami A. Role of the IL-6 classic- and trans-signaling pathways in corneal sterile inflammation and wound healing. Invest Ophthalmol Vis Sci. 2011;52:8549–57. doi: 10.1167/iovs.11-7956. [DOI] [PubMed] [Google Scholar]
  • 11.Lu P, Li L, Liu G, Zhang X, Mukaida N. Enhanced experimental corneal neovascularization along with aberrant angiogenic factor expression in the absence of IL-1 receptor antagonist. Invest Ophthalmol Vis Sci. 2009;50:4761–8. doi: 10.1167/iovs.08-2732. [DOI] [PubMed] [Google Scholar]
  • 12.Feizi S, Azari AA, Safapour S. Therapeutic approaches for corneal neovascularization. Eye Vis (Lond) 2017;4:28. doi: 10.1186/s40662-017-0094-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Troyanovsky B, Levchenko T, Månsson G, Matvijenko O, Holmgren L. Angiomotin: An angiostatin binding protein that regulates endothelial cell migration and tube formation. J Cell Biol. 2001;152:1247–54. doi: 10.1083/jcb.152.6.1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ambati BK, Nozaki M, Singh N, Takeda A, Jani PD, Suthar T, et al. Corneal avascularity is due to soluble VEGF receptor-1. Nature. 2006;443:993–7. doi: 10.1038/nature05249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ahmad S. Concise review: Limbal stem cell deficiency, dysfunction, and distress. Stem Cells Transl Med. 2012;1:110–5. doi: 10.5966/sctm.2011-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lim P, Fuchsluger TA, Jurkunas UV. Limbal stem cell deficiency and corneal neovascularization. Semin Ophthalmol. 2009;24:139–48. doi: 10.1080/08820530902801478. [DOI] [PubMed] [Google Scholar]
  • 17.Notara M, Alatza A, Gilfillan J, Harris AR, Levis HJ, Schrader S, et al. In sickness and in health: Corneal epithelial stem cell biology, pathology and therapy. Exp Eye Res. 2010;90:188–95. doi: 10.1016/j.exer.2009.09.023. [DOI] [PubMed] [Google Scholar]
  • 18.Bock F, Maruyama K, Regenfuss B, Hos D, Steven P, Heindl LM, et al. Novel anti(lymph)angiogenic treatment strategies for corneal and ocular surface diseases. Prog Retin Eye Res. 2013;34:89–124. doi: 10.1016/j.preteyeres.2013.01.001. [DOI] [PubMed] [Google Scholar]
  • 19.Nauck M, Karakiulakis G, Perruchoud AP, Papakonstantinou E, Roth M. Corticosteroids inhibit the expression of the vascular endothelial growth factor gene in human vascular smooth muscle cells. Eur J Pharmacol. 1998;341:309–15. doi: 10.1016/s0014-2999(97)01464-7. [DOI] [PubMed] [Google Scholar]
  • 20.Cursiefen C, Wenkel H, Martus P, Langenbucher A, Nguyen NX, Seitz B, et al. Impact of short- term versus long-term topical steroids on corneal neovascularization after non-high-risk keratoplasty. Graefes Arch Clin Exp Ophthalmol. 2001;239:514–21. doi: 10.1007/s004170100313. [DOI] [PubMed] [Google Scholar]
  • 21.Sharif Z, Sharif W. Corneal neovascularization: Updates on pathophysiology, investigations & management. Rom J Ophthalmol. 2019;63:15–22. [PMC free article] [PubMed] [Google Scholar]
  • 22.Hoffart L, Matonti F, Conrath J, Daniel L, Ridings B, Masson GS, et al. Inhibition of corneal neovascularization after alkali burn: Comparison of different doses of bevacizumab in monotherapy or associated with dexamethasone. Clin Exp Ophthalmol. 2010;38:346–52. doi: 10.1111/j.1442-9071.2010.02252.x. [DOI] [PubMed] [Google Scholar]
  • 23.Roshandel D, Eslani M, Baradaran-Rafii A, Cheung AY, Kurji K, Jabbehdari S, et al. Current and emerging therapies for corneal neovascularization. Ocul Surf. 2018;16:398–414. doi: 10.1016/j.jtos.2018.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Oray M, Abu Samra K, Ebrahimiadib N, Meese H, Foster CS. Long-term side effects of glucocorticoids. Expert Opin Drug Saf. 2016;15:457–65. doi: 10.1517/14740338.2016.1140743. [DOI] [PubMed] [Google Scholar]
  • 25.Shah G, Middleton FA, Gentile KL, Tripathi S, Bruch D, Maier KG, et al. Cyclosporine inhibition of angiogenesis involves the transcription factor HESR1. J Surg Res. 2008;149:171–6. doi: 10.1016/j.jss.2008.03.016. [DOI] [PubMed] [Google Scholar]
  • 26.Bucak YY, Erdurmus M, Terzi EH, Kükner A, Çelebi S. Inhibitory effects of topical cyclosporine A 0.05% on immune-mediated corneal neovascularization in rabbits. Graefes Arch Clin Exp Ophthalmol. 2013;251:2555–61. doi: 10.1007/s00417-013-2467-4. [DOI] [PubMed] [Google Scholar]
  • 27.Bock F, Matthaei M, Reinhard T, Böhringer D, Christoph J, Ganslandt T, et al. High-dose subconjunctival cyclosporine a implants do not affect corneal neovascularization after high-risk keratoplasty. Ophthalmology. 2014;121:1677–82. doi: 10.1016/j.ophtha.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 28.Althaus C, Dagres E, Reinhard T, Christians U, Sundmacher R. Cyclosporin-A and its metabolites in the anterior chamber after topical and systemic application as determined with high- performance liquid chromatography-electrospray mass spectrometry. Ger J Ophthalmol. 1996;5:189–94. [PubMed] [Google Scholar]
  • 29.Kino T, Hatanaka H, Miyata S, Inamura N, Nishiyama M, Yajima T, et al. FK-506, a novel immunosuppressant isolated from a Streptomyces. II. Immunosuppressive effect of FK-506 in vitro. J Antibiot (Tokyo) 1987;40:1256–65. doi: 10.7164/antibiotics.40.1256. [DOI] [PubMed] [Google Scholar]
  • 30.Turgut B, Guler M, Akpolat N, Demır T, Celıker U. The impact of tacrolimus on vascular endothelial growth factor in experimental corneal neovascularization. Curr Eye Res. 2011;36:34–40. doi: 10.3109/02713683.2010.516620. [DOI] [PubMed] [Google Scholar]
  • 31.Chen L, Zhong J, Li S, Li W, Wang B, Deng Y, et al. The long-term effect of tacrolimus on alkali burn-induced corneal neovascularization and inflammation surpasses that of anti-vascular endothelial growth factor. Drug Des Devel Ther. 2018;12:2959–69. doi: 10.2147/DDDT.S175297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lin X, Yu X, Chen X, Sheng S, Wang J, Wang B, et al. Inhibition of neovascularization and inflammation in a mouse model of corneal alkali burns using cationic liposomal tacrolimus. Front Bioeng Biotechnol. 2021;9:791954. doi: 10.3389/fbioe.2021.791954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Zhai LY, Zhang XR, Liu H, Ma Y, Xu HC. Observation of topical tacrolimus on high-risk penetrating keratoplasty patients: A randomized clinical trial study. Eye (Lond) 2020;34:1600–7. doi: 10.1038/s41433-019-0717-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ozdemir O, Altintas O, Altintas L, Yildiz DK, Sener E, Caglar Y. Effects of subconjunctivally injected bevacizumab, etanercept, and the combination of both drugs on experimental corneal neovascularization. Can J Ophthalmol. 2013;48:115–20. doi: 10.1016/j.jcjo.2012.12.003. [DOI] [PubMed] [Google Scholar]
  • 35.Yoo AR, Chung SK. Effects of subconjunctival tocilizumab versus bevacizumab in treatment of corneal neovascularization in rabbits. Cornea. 2014;33:1088–94. doi: 10.1097/ICO.0000000000000220. [DOI] [PubMed] [Google Scholar]
  • 36.Onder HI, Erdurmus M, Bucak YY, Simavli H, Oktay M, Kukner AS. Inhibitory effects of regorafenib, a multiple tyrosine kinase inhibitor, on corneal neovascularization. Int J Ophthalmol. 2014;7:220–5. doi: 10.3980/j.issn.2222-3959.2014.02.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hashemian MN, Mahrjerdi HZ, Mazloumi M, Safizadeh MS, Shakiba Y, Rahimi F, et al. Comparison of different doses of subconjunctival sunitinib with bevacizumab in the treatment of corneal neovascularization in experimental rats. J Res Med Sci. 2017;22:16. doi: 10.4103/1735-1995.200266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sener E, Yuksel N, Yildiz DK, Yilmaz B, Ozdemir O, Caglar Y, et al. The impact of subconjuctivally injected EGF and VEGF inhibitors on experimental corneal neovascularization in rat model. Curr Eye Res. 2011;36:1005–13. doi: 10.3109/02713683.2011.601840. [DOI] [PubMed] [Google Scholar]
  • 39.Akar EE, Oner V, Küçükerdönmez C, Aydın Akova Y. Comparison of subconjunctivally injected bevacizumab, ranibizumab, and pegaptanib for inhibition of corneal neovascularization in a rat model. Int J Ophthalmol. 2013;6:136–40. doi: 10.3980/j.issn.2222-3959.2013.02.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Park YR, Chung SK. Inhibitory effect of topical aflibercept on corneal neovascularization in rabbits. Cornea. 2015;34:1303–7. doi: 10.1097/ICO.0000000000000507. [DOI] [PubMed] [Google Scholar]
  • 41.Ucgul RK, Celebi S, Yilmaz NS, Bukan N, Ucgul AY. Intrastromal versus subconjunctival anti- VEGF agents for treatment of corneal neovascularization: A rabbit study. Eye (Lond) 2021;35:3123–30. doi: 10.1038/s41433-020-01347-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dastjerdi MH, Sadrai Z, Saban DR, Zhang Q, Dana R. Corneal penetration of topical and subconjunctival bevacizumab. Invest Ophthalmol Vis Sci. 2011;52:8718–23. doi: 10.1167/iovs.11-7871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Manzano RP, Peyman GA, Khan P, Carvounis PE, Kivilcim M, Ren M, et al. Inhibition of experimental corneal neovascularisation by bevacizumab (Avastin) Br J Ophthalmol. 2007;91:804–7. doi: 10.1136/bjo.2006.107912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cheng SF, Dastjerdi MH, Ferrari G, Okanobo A, Bower KS, Ryan DS, et al. Short-term topical bevacizumab in the treatment of stable corneal neovascularization. Am J Ophthalmol. 2012;154:940–8.e1. doi: 10.1016/j.ajo.2012.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kim SW, Ha BJ, Kim EK, Tchah H, Kim TI. The effect of topical bevacizumab on corneal neovascularization. Ophthalmology. 2008;115:e33–8. doi: 10.1016/j.ophtha.2008.02.013. [DOI] [PubMed] [Google Scholar]
  • 46.Koenig Y, Bock F, Horn F, Kruse F, Straub K, Cursiefen C. Short- and long-term safety profile and efficacy of topical bevacizumab (Avastin) eye drops against corneal neovascularization. Graefes Arch Clin Exp Ophthalmol. 2009;247:1375–82. doi: 10.1007/s00417-009-1099-1. [DOI] [PubMed] [Google Scholar]
  • 47.Bahar I, Kaiserman I, McAllum P, Rootman D, Slomovic A. Subconjunctival bevacizumab injection for corneal neovascularization. Cornea. 2008;27:142–7. doi: 10.1097/ICO.0b013e318159019f. [DOI] [PubMed] [Google Scholar]
  • 48.Papathanassiou M, Theodoropoulou S, Analitis A, Tzonou A, Theodossiadis PG. Vascular endothelial growth factor inhibitors for treatment of corneal neovascularization: A meta-analysis. Cornea. 2013;32:435–44. doi: 10.1097/ICO.0b013e3182542613. [DOI] [PubMed] [Google Scholar]
  • 49.Ferrari G, Dastjerdi MH, Okanobo A, Cheng SF, Amparo F, Nallasamy N, et al. Topical ranibizumab as a treatment of corneal neovascularization. Cornea. 2013;32:992–7. doi: 10.1097/ICO.0b013e3182775f8d. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Stevenson W, Cheng SF, Dastjerdi MH, Ferrari G, Dana R. Corneal neovascularization and the utility of topical VEGF inhibition: Ranibizumab (Lucentis) vs bevacizumab (Avastin) Ocul Surf. 2012;10:67–83. doi: 10.1016/j.jtos.2012.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Liarakos VS, Papaconstantinou D, Vergados I, Douvali M, Theodossiadis PG. The effect of subconjunctival ranibizumab on corneal and anterior segment neovascularization: Study on an animal model. Eur J Ophthalmol. 2014;24:299–308. doi: 10.5301/ejo.5000391. [DOI] [PubMed] [Google Scholar]
  • 52.Kim JH, Seo HW, Han HC, Lee JH, Choi SK, Lee D. The effect of bevacizumab versus ranibizumab in the treatment of corneal neovascularization: A preliminary study. Korean J Ophthalmol. 2013;27:235–42. doi: 10.3341/kjo.2013.27.4.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Andrade N, Crispim Ribeiro J, Araújo ESPM, Savio D, Fechine FV, de Moraes MO. Effect of topical instillation of pegaptanib sodium upon inflammatory corneal neovascularization in rabbits. J Ocul Pharmacol Ther. 2021;37:277–83. doi: 10.1089/jop.2020.0010. [DOI] [PubMed] [Google Scholar]
  • 54.de Oliveira Dias JR, de Andrade GC, Novais EA, Farah ME, Rodrigues EB. Fusion proteins for treatment of retinal diseases: Aflibercept, ziv-aflibercept, and conbercept. Int J Retina Vitreous. 2016;2:3. doi: 10.1186/s40942-016-0026-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Cai S, Yang Q, Li X, Zhang Y. The efficacy and safety of aflibercept and conbercept in diabetic macular edema. Drug Des Devel Ther. 2018;12:3471–83. doi: 10.2147/dddt.s177192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Eski MT, Teberik K, Oltulu P, Ankaralı H, Kaya M, Alpay M. The effects of subconjunctival bevacizumab, ranibizumab, and aflibercept on corneal neovascularization. Hum Exp Toxicol. 2022;41:9603271221084674. doi: 10.1177/09603271221084674. [DOI] [PubMed] [Google Scholar]
  • 57.Sella R, Gal-Or O, Livny E, Dachbash M, Nisgav Y, Weinberger D, et al. Efficacy of topical aflibercept versus topical bevacizumab for the prevention of corneal neovascularization in a rat model. Exp Eye Res. 2016;146:224–32. doi: 10.1016/j.exer.2016.03.021. [DOI] [PubMed] [Google Scholar]
  • 58.Sella R, Ben Ishai M, Livny E, Nahum Y, Bahar I. Subconjunctival aflibercept for the treatment of formed corneal neovascularization. Eye Contact Lens. 2021;47:180–4. doi: 10.1097/ICL.0000000000000709. [DOI] [PubMed] [Google Scholar]
  • 59.Sarah B, Ibtissam H, Mohammed B, Hasna S, Abdeljalil M. Intrastromal injection of bevacizumab in the management of corneal neovascularization: About 25 eyes. J Ophthalmol. 2016;2016:6084270. doi: 10.1155/2016/6084270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yeung SN, Lichtinger A, Kim P, Amiran MD, Slomovic AR. Combined use of subconjunctival and intracorneal bevacizumab injection for corneal neovascularization. Cornea. 2011;30:1110–4. doi: 10.1097/ICO.0b013e31821379aa. [DOI] [PubMed] [Google Scholar]
  • 61.Kim YC, Grossniklaus HE, Edelhauser HF, Prausnitz MR. Intrastromal delivery of bevacizumab using microneedles to treat corneal neovascularization. Invest Ophthalmol Vis Sci. 2014;55:7376–86. doi: 10.1167/iovs.14-15257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gupta AA, Mammo DA, Page MA. Intrastromal bevacizumab in the management of corneal neovascularization: A retrospective review. Graefes Arch Clin Exp Ophthalmol. 2020;258:167–73. doi: 10.1007/s00417-019-04519-4. [DOI] [PubMed] [Google Scholar]
  • 63.Bodnar RJ. Anti-angiogenic drugs: Involvement in cutaneous side effects and wound-healing complication. Adv Wound Care (New Rochelle) 2014;3:635–46. doi: 10.1089/wound.2013.0496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Bock F, Onderka J, Rummelt C, Dietrich T, Bachmann B, Kruse FE, et al. Safety profile of topical VEGF neutralization at the cornea. Invest Ophthalmol Vis Sci. 2009;50:2095–102. doi: 10.1167/iovs.07-1129. [DOI] [PubMed] [Google Scholar]
  • 65.Oh JY, Kim MK, Wee WR. Subconjunctival and intracorneal bevacizumab injection for corneal neovascularization in lipid keratopathy. Cornea. 2009;28:1070–3. doi: 10.1097/ICO.0b013e31819839f9. [DOI] [PubMed] [Google Scholar]
  • 66.Pérez-Santonja JJ, Campos-Mollo E, Lledó-Riquelme M, Javaloy J, Alió JL. Inhibition of corneal neovascularization by topical bevacizumab (Anti-VEGF) and Sunitinib (Anti-VEGF and Anti-PDGF) in an animal model. Am J Ophthalmol. 2010;150:519–28.e1. doi: 10.1016/j.ajo.2010.04.024. [DOI] [PubMed] [Google Scholar]
  • 67.You IC, Kang IS, Lee SH, Yoon KC. Therapeutic effect of subconjunctival injection of bevacizumab in the treatment of corneal neovascularization. Acta Ophthalmol. 2009;87:653–8. doi: 10.1111/j.1755-3768.2008.01399.x. [DOI] [PubMed] [Google Scholar]
  • 68.Benayoun Y, Adenis JP, Casse G, Forte R, Robert PY. Effects of subconjunctival bevacizumab on corneal neovascularization: Results of a prospective study. Cornea. 2012;31:937–44. doi: 10.1097/ICO.0b013e31823f8d71. [DOI] [PubMed] [Google Scholar]
  • 69.Chu HS, Chen TC, Hu FR, Chen WL. Recurrence of corneal neovascularization associated with lipid deposition after subconjunctival injection of bevacizumab. Cornea. 2013;32:1446–53. doi: 10.1097/ICO.0b013e31825ec407. [DOI] [PubMed] [Google Scholar]
  • 70.Britton AK, Crayford BB. Resolution of refractory corneal neovascularization with subconjunctival bevacizumab. Case Rep Ophthalmol. 2020;11:652–7. doi: 10.1159/000510114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Cavallini GM, Pellegrini G, Volante V, Ducange P, De Maria M, Torlai G, et al. Chemical injury treated with autologous limbal epithelial stem cell transplantation and subconjunctival bevacizumab. Clin Ophthalmol. 2014;8:1671–3. doi: 10.2147/OPTH.S65298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Iannetti L, Abbouda A, Fabiani C, Zito R, Campanella M. Treatment of corneal neovascularization in ocular chemical injury with an off-label use of subconjunctival bevacizumab: A case report. J Med Case Rep. 2013;7:199. doi: 10.1186/1752-1947-7-199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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;11:45–9. doi: 10.3928/1081-597X-19950101-12. [DOI] [PubMed] [Google Scholar]
  • 74.Huang AJ, Watson BD, Hernandez E, Tseng SC. Photothrombosis of corneal neovascularization by intravenous rose bengal and argon laser irradiation. Arch Ophthalmol. 1988;106:680–5. doi: 10.1001/archopht.1988.01060130742036. [DOI] [PubMed] [Google Scholar]
  • 75.Nirankari VS, Dandona L, Rodrigues MM. Laser photocoagulation of experimental corneal stromal vascularization. Efficacy and histopathology. Ophthalmology. 1993;100:111–8. doi: 10.1016/s0161-6420(93)31706-9. [DOI] [PubMed] [Google Scholar]
  • 76.Sharma A, Samal A, Narang S, Gutpa A, Ram J, Gupta A. Frequency doubled Nd:YAG (532 nm) laser photocoagulation in corneal vascularisation: Efficacy and time sequenced changes. Indian J Ophthalmol. 2001;49:235–40. [PubMed] [Google Scholar]
  • 77.Kumar J, Gehra A, Sirohi N. Role of frequency doubled Nd: Yag Laser in treatment of corneal neovascularisation. J Clin Diagn Res. 2016;10:Nc01–4. doi: 10.7860/JCDR/2016/17502.7543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hou Y, Le VNH, Clahsen T, Schneider AC, Bock F, Cursiefen C. Photodynamic therapy leads to time-dependent regression of pathologic corneal (Lymph) angiogenesis and promotes high-risk corneal allograft survival. Invest Ophthalmol Vis Sci. 2017;58:5862–9. doi: 10.1167/iovs.17-22904. [DOI] [PubMed] [Google Scholar]
  • 79.Bucher F, Bi Y, Gehlsen U, Hos D, Cursiefen C, Bock F. Regression of mature lymphatic vessels in the cornea by photodynamic therapy. Br J Ophthalmol. 2014;98:391–5. doi: 10.1136/bjophthalmol-2013-303887. [DOI] [PubMed] [Google Scholar]
  • 80.Pillai CT, Dua HS, Hossain P. Fine needle diathermy occlusion of corneal vessels. Invest Ophthalmol Vis Sci. 2000;41:2148–53. [PubMed] [Google Scholar]
  • 81.Faraj LA, Elalfy MS, Said DG, Dua HS. Fine needle diathermy occlusion of corneal vessels. Br J Ophthalmol. 2014;98:1287–90. doi: 10.1136/bjophthalmol-2014-304891. [DOI] [PubMed] [Google Scholar]
  • 82.Le VNH, Hou Y, Bock F, Cursiefen C. Supplemental Anti Vegf A-therapy prevents rebound neovascularisation after fine needle diathermy treatment to regress pathological corneal (LYMPH) angiogenesis. Sci Rep. 2020;10:3908. doi: 10.1038/s41598-020-60705-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hos D, Le VNH, Hellmich M, Siebelmann S, Roters S, Bachmann BO, et al. Risk of corneal graft rejection after high-risk keratoplasty following fine-needle vessel coagulation of corneal neovascularization combined with bevacizumab: A pilot study. Transplant Direct. 2019;5:e452. doi: 10.1097/TXD.0000000000000894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Elbaz U, Mireskandari K, Shen C, Ali A. Corneal fine needle diathermy with adjuvant bevacizumab to treat corneal neovascularization in children. Cornea. 2015;34:773–7. doi: 10.1097/ICO.0000000000000419. [DOI] [PubMed] [Google Scholar]
  • 85.Le VNH, Schneider AC, Scholz R, Bock F, Cursiefen C. Fine needle-diathermy regresses pathological corneal (Lymph) angiogenesis and promotes high-risk corneal transplant survival. Sci Rep. 2018;8:5707. doi: 10.1038/s41598-018-24037-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Cursiefen C, Hofmann-Rummelt C, Küchle M, Schlötzer-Schrehardt U. Pericyte recruitment in human corneal angiogenesis: An ultrastructural study with clinicopathological correlation. Br J Ophthalmol. 2003;87:101–6. doi: 10.1136/bjo.87.1.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Spiteri N, Romano V, Zheng Y, Yadav S, Dwivedi R, Chen J, et al. Corneal angiography for guiding and evaluating fine-needle diathermy treatment of corneal neovascularization. Ophthalmology. 2015;122:1079–84. doi: 10.1016/j.ophtha.2015.02.012. [DOI] [PubMed] [Google Scholar]
  • 88.Kim BY, Riaz KM, Bakhtiari P, Chan CC, Welder JD, Holland EJ, et al. Medically reversible limbal stem cell disease: Clinical features and management strategies. Ophthalmology. 2014;121:2053–8. doi: 10.1016/j.ophtha.2014.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Le Q, Chauhan T, Yung M, Tseng CH, Deng SX. Outcomes of limbal stem cell transplant: A meta-analysis. JAMA Ophthalmol. 2020;138:660–70. doi: 10.1001/jamaophthalmol.2020.1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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;96:931–4. doi: 10.1136/bjophthalmol-2011-301164. [DOI] [PubMed] [Google Scholar]
  • 91.Jackson CJ, Myklebust Ernø IT, Ringstad H, Tønseth KA, Dartt DA, Utheim TP. Simple limbal epithelial transplantation: Current status and future perspectives. Stem Cells Transl Med. 2020;9:316–27. doi: 10.1002/sctm.19-0203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Seki Y, Toba K, Fuse I, Sato N, Niwano H, Takahashi H, et al. In vitro effect of cyclosporin A, mitomycin C and prednisolone on cell kinetics in cultured human umbilical vein endothelial cells. Thromb Res. 2005;115:219–28. doi: 10.1016/j.thromres.2004.09.001. [DOI] [PubMed] [Google Scholar]
  • 93.Mimouni M, Ouano D. Initial outcomes of mitomycin intravascular chemoembolization (MICE) for corneal neovascularization. Int Ophthalmol. 2022;42:2407–16. doi: 10.1007/s10792-022-02240-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Addeen SZ, Oyoun Z, Alfhaily H, Anbari A. Outcomes of mitomycin C intravascular chemoembolization (MICE) in refractory corneal neovascularization after failed keratoplasty. Digit J Ophthalmol. 2023;29:9–13. doi: 10.5693/djo.02.2023.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wu D, Lim DK, Lim BXH, Wong N, Hafezi F, Manotosh R, et al. Corneal cross-linking: The evolution of treatment for corneal diseases. Front Pharmacol. 2021;12:686630. doi: 10.3389/fphar.2021.686630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhu Y, Li L, Reinach PS, Li Y, Ge C, Qu J, et al. Corneal collagen cross-linking with Riboflavin and UVA regulates hemangiogenesis and lymphangiogenesis in rats. Invest Ophthalmol Vis Sci. 2018;59:3702–12. doi: 10.1167/iovs.17-23036. [DOI] [PubMed] [Google Scholar]
  • 97.Bertollo CM, Oliveira AC, Rocha LT, Costa KA, Nascimento EB, Jr, Coelho MM. Characterization of the antinociceptive and anti-inflammatory activities of riboflavin in different experimental models. Eur J Pharmacol. 2006;547:184–91. doi: 10.1016/j.ejphar.2006.07.045. [DOI] [PubMed] [Google Scholar]
  • 98.Notara M, Refaian N, Braun G, Steven P, Bock F, Cursiefen C. Short-term ultraviolet A irradiation leads to dysfunction of the limbal niche cells and an antilymphangiogenic and anti- inflammatory micromilieu. Invest Ophthalmol Vis Sci. 2016;57:928–39. doi: 10.1167/iovs.15-18343. [DOI] [PubMed] [Google Scholar]
  • 99.Hou Y, Le VNH, Tóth G, Siebelmann S, Horstmann J, Gabriel T, et al. UV light crosslinking regresses mature corneal blood and lymphatic vessels and promotes subsequent high-risk corneal transplant survival. Am J Transplant. 2018;18:2873–84. doi: 10.1111/ajt.14874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Xu X, Liu T, Li H. Effect of collagen cross-linking on alkali burn-induced corneal neovascularization in rabbits. J Ophthalmol. 2018;2018:7325483. doi: 10.1155/2018/7325483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kesim E, Pirhan D, Yardimoglu Yilmaz M, Yuksel N, Yazir Y, Bicaklioglu G, et al. Comparative analysis of matrix-regenerating agent and corneal cross-linking in an experimental alkali burn rabbit model. Curr Eye Res. 2022;47:187–95. doi: 10.1080/02713683.2021.1971722. [DOI] [PubMed] [Google Scholar]
  • 102.Subasi S, Altintas O, Yardimoglu M, Yazir Y, Karaman S, Rencber SF, et al. Comparison of collagen cross-linking and amniotic membrane transplantation in an experimental alkali burn rabbit model. Cornea. 2017;36:1106–15. doi: 10.1097/ICO.0000000000001276. [DOI] [PubMed] [Google Scholar]
  • 103.Colombo-Barboza M, Colombo-Barboza G, Felberg S, Dantas PE, Sato EH. Induction of corneal collagen cross-linking in experimental corneal alkali burns in rabbits. Arq Bras Oftalmol. 2014;77:310–4. doi: 10.5935/0004-2749.20140078. [DOI] [PubMed] [Google Scholar]
  • 104.Schaub F, Hou Y, Zhang W, Bock F, Hos D, Cursiefen C. Corneal crosslinking to regress pathologic corneal neovascularization before high-risk keratoplasty. Cornea. 2021;40:147–55. doi: 10.1097/ICO.0000000000002406. [DOI] [PubMed] [Google Scholar]
  • 105.Anguela XM, High KA. Entering the modern era of gene therapy. Annu Rev Med. 2019;70:273–88. doi: 10.1146/annurev-med-012017-043332. [DOI] [PubMed] [Google Scholar]
  • 106.Amador C, Shah R, Ghiam S, Kramerov AA, Ljubimov AV. Gene therapy in the anterior eye segment. Curr Gene Ther. 2022;22:104–31. doi: 10.2174/1566523221666210423084233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Lai YK, Shen WY, Brankov M, Lai CM, Constable IJ, Rakoczy PE. Potential long-term inhibition of ocular neovascularisation by recombinant adeno-associated virus-mediated secretion gene therapy. Gene Ther. 2002;9:804–13. doi: 10.1038/sj.gt.3301695. [DOI] [PubMed] [Google Scholar]
  • 108.Iriyama A, Usui T, Yanagi Y, Amano S, Oba M, Miyata K, et al. Gene transfer using micellar nanovectors inhibits corneal neovascularization in vivo. Cornea. 2011;30:1423–7. doi: 10.1097/ICO.0b013e318206c893. [DOI] [PubMed] [Google Scholar]
  • 109.Yu H, Wu J, Li H, Wang Z, Chen X, Tian Y, et al. Inhibition of corneal neovascularization by recombinant adenovirus-mediated sFlk-1 expression. Biochem Biophys Res Commun. 2007;361:946–52. doi: 10.1016/j.bbrc.2007.07.114. [DOI] [PubMed] [Google Scholar]
  • 110.Yoon KC, Bae JA, Park HJ, Im SK, Oh HJ, Lin XH, et al. Subconjunctival gene delivery of the transcription factor GA-binding protein delays corneal neovascularization in a mouse model. Gene Ther. 2009;16:973–81. doi: 10.1038/gt.2009.50. [DOI] [PubMed] [Google Scholar]
  • 111.Lai CM, Spilsbury K, Brankov M, Zaknich T, Rakoczy PE. Inhibition of corneal neovascularization by recombinant adenovirus mediated antisense VEGF RNA. Exp Eye Res. 2002;75:625–34. doi: 10.1006/exer.2002.2075. [DOI] [PubMed] [Google Scholar]
  • 112.Qazi Y, Stagg B, Singh N, Singh S, Zhang X, Luo L, et al. Nanoparticle-mediated delivery of shRNA.VEGF-a plasmids regresses corneal neovascularization. Invest Ophthalmol Vis Sci. 2012;53:2837–44. doi: 10.1167/iovs.11-9139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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]
  • 114.Parker M, Bellec J, McFarland T, Scripps V, Appukuttan B, Hartzell M, et al. Suppression of neovascularization of donor corneas by transduction with equine infectious anemia virus-based lentiviral vectors expressing endostatin and angiostatin. Hum Gene Ther. 2014;25:408–18. doi: 10.1089/hum.2013.079. [DOI] [PubMed] [Google Scholar]
  • 115.Torrecilla J, Gómez-Aguado I, Vicente-Pascual M, Del Pozo-Rodríguez A, Solinís M, Rodríguez- Gascón A. MMP-9 downregulation with lipid nanoparticles for inhibiting corneal neovascularization by gene silencing. Nanomaterials (Basel) 2019;9:631. doi: 10.3390/nano9040631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Chen J, Li F, Xu Y, Zhang W, Hu Y, Fu Y, et al. Cholesterol modification of SDF-1-specific siRNA enables therapeutic targeting of angiogenesis through Akt pathway inhibition. Exp Eye Res. 2019;184:64–71. doi: 10.1016/j.exer.2019.03.006. [DOI] [PubMed] [Google Scholar]
  • 117.Cao X, Wang C, Deng Z, Zhong Y, Chen H. Efficient ocular delivery of siRNA via pH-sensitive vehicles for corneal neovascularization inhibition. Int J Pharm X. 2023;5:100183. doi: 10.1016/j.ijpx.2023.100183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cursiefen C, Viaud E, Bock F, Geudelin B, Ferry A, Kadlecová P, et al. Aganirsen antisense oligonucleotide eye drops inhibit keratitis-induced corneal neovascularization and reduce need for transplantation: The I-CAN study. Ophthalmology. 2014;121:1683–92. doi: 10.1016/j.ophtha.2014.03.038. [DOI] [PubMed] [Google Scholar]
  • 119.Zhang C, Yin Y, Zhao J, Li Y, Wang Y, Zhang Z, et al. An update on novel ocular nanosystems with possible benefits in the treatment of corneal neovascularization. Int J Nanomedicine. 2022;17:4911–31. doi: 10.2147/IJN.S375570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Hos D, Saban DR, Bock F, Regenfuss B, Onderka J, Masli S, et al. Suppression of Inflammatory corneal lymphangiogenesis by application of topical corticosteroids. Arch Ophthalmol. 2011;129:445–52. doi: 10.1001/archophthalmol.2011.42. [DOI] [PubMed] [Google Scholar]
  • 121.Cooper CA, Bergamini MVW, Leopold IH. Use of flurbiprofen to inhibit corneal neovascularization. Arch Ophthalmol. 1980;98:1102–5. doi: 10.1001/archopht.1980.01020031092017. [DOI] [PubMed] [Google Scholar]
  • 122.Pakneshan P, Birsner AE, Adini I, Becker CM, D’Amato RJ. Differential suppression of vascular permeability and corneal angiogenesis by nonsteroidal anti-inflammatory drugs. Invest Ophthalmol Vis Sci. 2008;49:3909–13. doi: 10.1167/iovs.07-1527. [DOI] [PubMed] [Google Scholar]
  • 123.Ulusoy DM, Kahraman N, Balcıoğlu E, Duru Z. Comparison of the inhibitory effect of topical cyclosporine A 0.1% and topical anti-VEGF application in an experimental model of corneal neovascularization. Arq Bras Oftalmol. 2022;85:19–24. doi: 10.5935/0004-2749.20220004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Byun YS, Chung SK. The effect of methotrexate on corneal neovascularization in rabbits. Cornea. 2011;30:442–6. doi: 10.1097/IAE.0b013e3181e46ad8. [DOI] [PubMed] [Google Scholar]
  • 125.Kruse FE, Joussen AM, Rohrschneider K, Becker MD, Völcker HE. Thalidomide inhibits corneal angiogenesis induced by vascular endothelial growth factor. Graefes Arch Clin Exp Ophthalmol. 1998;236:461–6. doi: 10.1007/s004170050106. [DOI] [PubMed] [Google Scholar]
  • 126.Abbas A, Khan B, Feroze AH, Hyman GF. Thalidomide prevents donor corneal graft neovascularization in an alkali burn model of corneal angiogenesis. J Pak Med Assoc. 2002;52:476–82. [PubMed] [Google Scholar]
  • 127.Lee YK, Chung SK. The inhibitory effect of thalidomide analogue on corneal neovascularization in rabbits. Cornea. 2013;32:1142–8. doi: 10.1097/ICO.0b013e318292a79d. [DOI] [PubMed] [Google Scholar]
  • 128.Dana MR, Zhu SN, Yamada J. Topical modulation of interleukin-1 activity in corneal neovascularization. Cornea. 1998;17:403–9. doi: 10.1097/00003226-199807000-00011. [DOI] [PubMed] [Google Scholar]
  • 129.Özkaya D, Karaca U, Usta Sofu G, Savran M, Özgöçmen M, Ertuğrul A. Effect of adalimumab on experimental corneal neovascularization model. Int Ophthalmol. 2023;43:2119–28. doi: 10.1007/s10792-023-02695-1. [DOI] [PubMed] [Google Scholar]
  • 130.Amparo F, Sadrai Z, Jin Y, Alfonso-Bartolozzi B, Wang H, Shikari H, et al. Safety and efficacy of the multitargeted receptor kinase inhibitor pazopanib in the treatment of corneal neovascularization. Invest Ophthalmol Vis Sci. 2013;54:537–44. doi: 10.1167/iovs.12-11032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wang JH, Tseng CL, Lin FL, Chen J, Hsieh EH, Lama S, et al. Topical application of TAK1 inhibitor encapsulated by gelatin particle alleviates corneal neovascularization. Theranostics. 2022;12:657–74. doi: 10.7150/thno.65098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Dastjerdi MH, Saban DR, Okanobo A, Nallasamy N, Sadrai Z, Chauhan SK, et al. Effects of topical and subconjunctival bevacizumab in high-risk corneal transplant survival. Invest Ophthalmol Vis Sci. 2010;51:2411–7. doi: 10.1167/iovs.09-3745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Petsoglou C, Balaggan KS, Dart JK, Bunce C, Xing W, Ali RR, et al. Subconjunctival bevacizumab induces regression of corneal neovascularisation: A pilot randomised placebo-controlled double- masked trial. Br J Ophthalmol. 2013;97:28–32. doi: 10.1136/bjophthalmol-2012-302137. [DOI] [PubMed] [Google Scholar]
  • 134.Kim EK, Kong SJ, Chung SK. Comparative study of ranibizumab and bevacizumab on corneal neovascularization in rabbits. Cornea. 2014;33:60–4. doi: 10.1097/ICO.0000000000000007. [DOI] [PubMed] [Google Scholar]
  • 135.Oliveira HB, Sakimoto T, Javier JA, Azar DT, Wiegand SJ, Jain S, et al. VEGF Trap(R1R2) suppresses experimental corneal angiogenesis. Eur J Ophthalmol. 2010;20:48–54. doi: 10.1177/112067211002000106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Eski M, Teberik K, Oltulu P, Ankaralı H, Kaya M, Alpay M. The effects of subconjunctival bevacizumab, ranibizumab, and aflibercept on corneal neovascularization. Hum Exp Toxicol. 2022;41:09603271221084674. doi: 10.1177/09603271221084674. [DOI] [PubMed] [Google Scholar]
  • 137.Liu H, Zhang XR, Xu HC, Ma Y, Huang LY, Zhai LY, et al. Effects of VEGF inhibitor conbercept on corneal neovascularization following penetrating keratoplasty in rabbit model. Clin Ophthalmol. 2020;14:2185–93. doi: 10.2147/OPTH.S260302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Du L, Sun J, Liu J, Xu N, Liu M, Wu X. Effect of conbercept on corneal neovascularization in a rabbit model. Semin Ophthalmol. 2023;38:670–8. doi: 10.1080/08820538.2023.2201652. [DOI] [PubMed] [Google Scholar]
  • 139.Sun C, Ruan F, Li S, Zhang JQ, Jie Y. Subconjunctival conbercept for the treatment of corneal neovascularization. Int J Ophthalmol. 2023;16:871–5. doi: 10.18240/ijo.2023.06.06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Cherry PM, Garner A. Corneal neovascularization treated with argon laser. Br J Ophthalmol. 1976;60:464–72. doi: 10.1136/bjo.60.6.464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gordon YJ, Mann RK, Mah TS, Gorin MB. Fluorescein-potentiated argon laser therapy improves symptoms and appearance of corneal neovascularization. Cornea. 2002;21:770–3. doi: 10.1097/00003226-200211000-00007. [DOI] [PubMed] [Google Scholar]
  • 142.Primbs GB, Casey R, Wamser K, Snyder WJ, Crean DH. Photodynamic therapy for corneal neovascularization. Ophthalmic Surg Lasers. 1998;29:832–8. [PubMed] [Google Scholar]
  • 143.Al-Torbak AA. Photodynamic therapy with verteporfin for corneal neovascularization. Middle East Afr J Ophthalmol. 2012;19:185–9. doi: 10.4103/0974-9233.95246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Yoon KC, You IC, Kang IS, Im SK, Ahn JK, Park YG, et al. Photodynamic therapy with verteporfin for corneal neovascularization. Am J Ophthalmol. 2007;144:390–5. doi: 10.1016/j.ajo.2007.05.028. [DOI] [PubMed] [Google Scholar]
  • 145.Romano V, Steger B, Brunner M, Ahmad S, Willoughby CE, Kaye SB. Method for angiographically guided fine-needle diathermy in the treatment of corneal neovascularization. Cornea. 2016;35:1029–32. doi: 10.1097/ICO.0000000000000865. [DOI] [PubMed] [Google Scholar]
  • 146.Zhu L, Zhang W, Zhu J, Chen C, Mo K, Guo H, et al. Cotransplantation of limbal epithelial and stromal cells for ocular surface reconstruction. Ophthalmol Sci. 2022;2:100148. doi: 10.1016/j.xops.2022.100148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Zakaria N, Possemiers T, Dhubhghaill SN, Leysen I, Rozema J, Koppen C, et al. Results of a phase I/II clinical trial: Standardized, non-xenogenic, cultivated limbal stem cell transplantation. J Transl Med. 2014;12:58. doi: 10.1186/1479-5876-12-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Le Q, Chauhan T, Yung M, Tseng C-H, Deng SX. Outcomes of limbal stem cell transplant: A meta-analysis. JAMA Ophthalmol. 2020;138:660–70. doi: 10.1001/jamaophthalmol.2020.1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Xu X, Liu T, Li H. Effect of collagen cross-linking on alkali burn-induced corneal neovascularization in rabbits. J Ophthalmol. 2018;2018:7325483. doi: 10.1155/2018/7325483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kesim E, Pirhan D, Yardimoglu Yilmaz M, Yuksel N, Yazir Y, Bicaklioglu G, et al. Comparative analysis of matrix-regenerating agent and corneal cross-linking in an experimental alkali burn rabbit model. Curr Eye Res. 2022;47:187–95. doi: 10.1080/02713683.2021.1971722. [DOI] [PubMed] [Google Scholar]
  • 151.Lyu N, Zhao Y, Xiang J, Fan X, Huang C, Sun X, et al. Inhibiting corneal neovascularization by sustainably releasing anti-VEGF and anti-inflammation drugs from silica-thermogel nanohybrids. Mater Sci Eng C Mater Biol Appl. 2021;128:112274. doi: 10.1016/j.msec.2021.112274. [DOI] [PubMed] [Google Scholar]
  • 152.Di Tommaso C, Bourges JL, Valamanesh F, Trubitsyn G, Torriglia A, Jeanny JC, et al. Novel micelle carriers for cyclosporin A topical ocular delivery: In vivo cornea penetration, ocular distribution and efficacy studies. Eur J Pharm Biopharm. 2012;81:257–64. doi: 10.1016/j.ejpb.2012.02.014. [DOI] [PubMed] [Google Scholar]
  • 153.Chuang YL, Fang HW, Ajitsaria A, Chen KH, Su CY, Liu GS, et al. Development of Kaempferol- loaded gelatin nanoparticles for the treatment of corneal neovascularization in mice. Pharmaceutics. 2019;11:635. doi: 10.3390/pharmaceutics11120635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Chu YC, Fang HW, Wu YY, Tang YJ, Hsieh EH, She Y, et al. Functional peptide-loaded gelatin nanoparticles as eyedrops for cornea neovascularization treatment. Int J Nanomedicine. 2023;18:1413–31. doi: 10.2147/IJN.S398769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Zhang X-P, Sun J-G, Yao J, Shan K, Liu B-H, Yao M-D, et al. Effect of nanoencapsulation using poly (lactide-co-glycolide) (PLGA) on anti-angiogenic activity of bevacizumab for ocular angiogenesis therapy. Biomed Pharmacother. 2018;107:1056–63. doi: 10.1016/j.biopha.2018.08.092. [DOI] [PubMed] [Google Scholar]
  • 156.Cho WK, Kang S, Choi H, Rho CR. Topically administered gold nanoparticles inhibit experimental corneal neovascularization in mice. Cornea. 2015;34:456–9. doi: 10.1097/ICO.0000000000000343. [DOI] [PubMed] [Google Scholar]
  • 157.Zheng Q, Fang Y, Zeng L, Li X, Chen H, Song H, et al. Cytocompatible cerium oxide-mediated antioxidative stress in inhibiting ocular inflammation-associated corneal neovascularization. J Mater Chem B. 2019;7:6759–69. doi: 10.1039/c9tb01066a. [DOI] [PubMed] [Google Scholar]
  • 158.Delgado-Tirado S, Gonzalez-Buendia L, An M, Amarnani D, Isaacs-Bernal D, Whitmore H, et al. Topical nanoemulsion of a runt-related transcription factor 1 inhibitor for the treatment of pathologic ocular angiogenesis. Ophthalmol Sci. 2022;2:100163. doi: 10.1016/j.xops.2022.100163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Soiberman U, Kambhampati SP, Wu T, Mishra MK, Oh Y, Sharma R, et al. Subconjunctival injectable dendrimer-dexamethasone gel for the treatment of corneal inflammation. Biomaterials. 2017;125:38–53. doi: 10.1016/j.biomaterials.2017.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Yuan X, Marcano DC, Shin CS, Hua X, Isenhart LC, Pflugfelder SC, et al. Ocular drug delivery nanowafer with enhanced therapeutic efficacy. ACS Nano. 2015;9:1749–58. doi: 10.1021/nn506599f. [DOI] [PubMed] [Google Scholar]
  • 161.Nikolic L, Friend J, Taylor S, Thoft R. Inhibition of vascularization in rabbit corneas by heparin: Cortisone pellets. Invest Ophthalmol Vis Sci. 1986;27:449–56. [PubMed] [Google Scholar]
  • 162.Aydin E, Kivilcim M, Peyman GA, Esfahani MR, Kazi AA, Sanders DR. Inhibition of experimental angiogenesis of cornea by various doses of doxycycline and combination of triamcinolone acetonide with low-molecular-weight heparin and doxycycline. Cornea. 2008;27:446–53. doi: 10.1097/ICO.0b013e3181605ff9. [DOI] [PubMed] [Google Scholar]
  • 163.Tommila P, Summanen P, Tervo T. Cortisone, heparin and argon laser in the treatment of corneal neovascularization. Acta Ophthalmol Suppl (1985) 1987;182:89–92. doi: 10.1111/j.1755-3768.1987.tb02600.x. [DOI] [PubMed] [Google Scholar]
  • 164.Kang S, Chung SK. The effect of subconjuctival combined treatment of bevacizumab and triamcinolone acetonide on corneal neovascularization in rabbits. Cornea. 2010;29:192–6. doi: 10.1097/ICO.0b013e3181b1c82f. [DOI] [PubMed] [Google Scholar]
  • 165.Shi H, Zhu Y, Xing C, Li S, Bao Z, Lei L, et al. An injectable thermosensitive hydrogel for dual delivery of diclofenac and Avastin® to effectively suppress inflammatory corneal neovascularization. Int J Pharm. 2022;625:122081. doi: 10.1016/j.ijpharm.2022.122081. [DOI] [PubMed] [Google Scholar]
  • 166.Zhou C, Lei F, Sharma J, Hui PC, Wolkow N, Dohlman CH, et al. Sustained Inhibition of VEGF and TNF-α achieves multi-ocular protection and prevents formation of blood vessels after severe ocular trauma. Pharmaceutics. 2023;15:2059. doi: 10.3390/pharmaceutics15082059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Gerten G. Bevacizumab (avastin) and argon laser to treat neovascularization in corneal transplant surgery. Cornea. 2008;27:1195–9. doi: 10.1097/ICO.0b013e318180e50f. [DOI] [PubMed] [Google Scholar]
  • 168.Anand N, Reidy JJ, Riaz KM. Short-term regression of corneal neovascularization with combination therapy of argon green laser photocoagulation and subconjunctival bevacizumab. Int Med Case Rep J. 2019;12:89–92. doi: 10.2147/IMCRJ.S195990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Donato MG, Donato E, Cordeiro MÁ C, Martins de Andrade M, Holanda de Freitas JA. Treating corneal neovascularization using a combination of anti-VEGF injection and argon laser photocoagulation application - case report. Rom J Ophthalmol. 2021;65:286–9. doi: 10.22336/rjo.2021.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Kim RY, Chung SK, Kim MS, Ra H. Effects of combined photodynamic therapy and topical bevacizumab treatment on corneal neovascularization in rabbits. Cornea. 2016;35:1615–20. doi: 10.1097/ICO.0000000000001016. [DOI] [PubMed] [Google Scholar]
  • 171.Hamdan J, Boulze M, Aziz A, Alessi G, Hoffart L. [Corneal neovascularisation treatments compared: Subconjunctival bevacizumab injections and/or photodynamic therapy] J Fr Ophtalmol. 2015;38:924–33. doi: 10.1016/j.jfo.2015.06.006. [DOI] [PubMed] [Google Scholar]
  • 172.Yoon HJ, Kim MK, Seo KY, Ueta M, Yoon KC. Effectiveness of photodynamic therapy with verteporfin combined with intrastromal bevacizumab for corneal neovascularization in Stevens- Johnson syndrome. Int Ophthalmol. 2019;39:55–62. doi: 10.1007/s10792-017-0786-x. [DOI] [PubMed] [Google Scholar]
  • 173.Koenig Y, Bock F, Kruse FE, Stock K, Cursiefen C. Angioregressive pretreatment of mature corneal blood vessels before keratoplasty: Fine-needle vessel coagulation combined with anti-VEGFs. Cornea. 2012;31:887–92. doi: 10.1097/ICO.0b013e31823f8f7a. [DOI] [PubMed] [Google Scholar]
  • 174.Mestanoglu M, Händel A, Cursiefen C, Hos D. Three-year follow-up of high-risk keratoplasty following fine-needle diathermy of corneal neovascularization combined with bevacizumab. Graefes Arch Clin Exp Ophthalmol. 2022;260:2383–5. doi: 10.1007/s00417-021-05546-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Hayashi T, Zhang W, Hos D, Schrittenlocher S, Nhat Hung Le V, Siebelmann S, et al. Descemet membrane endothelial keratoplasty in vascularized eyes: Outcome and effect on corneal neovascularization. Cornea. 2021;40:685–9. doi: 10.1097/ICO.0000000000002502. [DOI] [PubMed] [Google Scholar]
  • 176.Yao L, Li ZR, Su WR, Li YP, Lin ML, Zhang WX, et al. Role of mesenchymal stem cells on cornea wound healing induced by acute alkali burn. PLoS One. 2012;7:e30842. doi: 10.1371/journal.pone.0030842. doi: 10.1371/journal.pone.0030842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Song HB, Park SY, Ko JH, Park JW, Yoon CH, Kim DH, et al. Mesenchymal stromal cells inhibit Inflammatory lymphangiogenesis in the cornea by suppressing macrophage in a TSG-6-dependent manner. Mol Ther. 2018;26:162–72. doi: 10.1016/j.ymthe.2017.09.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Xiao O, Xie ZL, Lin BW, Yin XF, Pi RB, Zhou SY. Minocycline inhibits alkali burn-induced corneal neovascularization in mice. PLoS One. 2012;7:e41858. doi: 10.1371/journal.pone.0041858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Zhong YY, Zhang HF, Zhong JX, Bai L, Lu XH. Topical dihydroartemisinin inhibits suture-induced neovascularization in rat corneas through ERK1/2 and p38 pathways. Int J Ophthalmol. 2011;4:150–5. doi: 10.3980/j.issn.2222-3959.2011.02.08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Goktas S, Erdogan E, Sakarya R, Sakarya Y, Yılmaz M, Ozcimen M, et al. Inhibition of corneal neovascularization by topical and subconjunctival tigecycline. J Ophthalmol. 2014;2014:452685. doi: 10.1155/2014/452685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Li H, Ye B, Zhu L, Wu L. Epigallocatechin gallate inhibits corneal neovascularization in ratalkaline burn model. Pak J Pharm Sci. 2018;31(3(Supplementary)):1087–92. [PubMed] [Google Scholar]
  • 182.Bian F, Zhang M-C, Zhu Y. Inhibitory effect of curcumin on corneal neovascularization in vitro and in vivo. Ophthalmologica. 2008;222:178–86. doi: 10.1159/000126081. [DOI] [PubMed] [Google Scholar]
  • 183.Kim JS, Choi JS, Chung SK. The effect of curcumin on corneal neovascularization in rabbit eyes. Curr Eye Res. 2010;35:274–80. doi: 10.3109/02713680903528345. [DOI] [PubMed] [Google Scholar]
  • 184.Suzuki T, Sano Y, Kinoshita S. Effects of 1alpha,25-dihydroxyvitamin D3 on Langerhans cell migration and corneal neovascularization in mice. Invest Ophthalmol Vis Sci. 2000;41:154–8. [PubMed] [Google Scholar]
  • 185.Lee MY, Chung SK. Treatment of corneal neovascularization by topical application of ascorbic acid in the rabbit model. Cornea. 2012;31:1165–9. doi: 10.1097/ICO.0b013e318241433b. [DOI] [PubMed] [Google Scholar]

Articles from Indian Journal of Ophthalmology are provided here courtesy of Wolters Kluwer -- Medknow Publications

RESOURCES