Nano-based ocular drug delivery systems: an insight into the preclinical/clinical studies and their potential in the treatment of posterior ocular diseases

Yun Su ab, Xianqun Fan *ab and Yan Pang *ab
aDepartment of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China. E-mail: yanpang@sjtu.edu.cn; fanxq@sjtu.edu.cn; Tel: +8621-63135606
bShanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China

Received 23rd March 2023 , Accepted 8th May 2023

First published on 9th May 2023


Abstract

Numerous novel nano-based ocular drug delivery systems have been developed to overcome the limitations of conventional drug delivery systems, which have demonstrated promising results in ocular disease models and clinical practice. Of all the nano-based drug delivery systems approved or under clinical investigation, topical instillation of eye drops is the most common route for administering therapeutics to the eye. Although this pathway is a viable way of ocular drug delivery to treat many ocular diseases because of its potential to eliminate the risks of intravitreal injection and the toxicity of systemic drug delivery, it remains a major challenge to efficiently treat posterior ocular diseases through topical administration of eye drops. So far, relentless efforts have been dedicated to the development of novel nano-based drug delivery systems with the aim of possible clinical translation. They are designed or modified to facilitate drug delivery to the retina by increasing the retention time, promoting drug penetration across barriers, and targeting specific cells or tissues. In this paper, we provided a snapshot of nano-based drug delivery systems that are currently marketed and under investigation in clinical trials for the treatment of ocular diseases and highlighted some examples of recent preclinical research on novel nano-based systems as eye drops to the posterior segment of the eye.


1. Introduction

The eye is one of the most complicated organs in the human body with distinctive anatomical and physiological structures. Anatomically, the eye can be divided into anterior and posterior segments with the anterior segment including the cornea, conjunctiva, anterior chamber, aqueous humor, iris, and lens and the posterior segment consisting of the vitreous humor, retina, choroid, sclera, and optic nerve. The complexity of the eye's structure, such as the cornea, blood–aqueous barrier, and blood–retinal barrier, prevents the drug from penetrating deeply into the eye. Other major barriers, including tear dilution, tear turnover, and nasolacrimal drainage, also lead to a reduced residence time of the drug. Conventional drug delivery systems, such as eye drops, ointments, and suspensions, are mostly effective in the treatment of ocular diseases in the anterior segment of the eye due to limited ocular bioavailability. For the treatment of posterior segment diseases, invasive procedures are commonly used to achieve higher concentrations, but the eye is at risk of complications from repeated injection. Therefore, the introduction of an ocular drug delivery system is one of the most promising ways to deliver drugs to the target site in a therapeutic dose.

Nanomedicine is an emerging field that combines nanotechnology with pharmaceutical and biomedical sciences to improve medical interventions for the prevention, diagnosis, and treatment of diseases.1 It intends to develop drugs and imaging agents with increased clinical efficacy, decreased degradation or physiologic clearance rate, and enhanced toxicological profiles. Many of these properties are not inherent to specific particles. They are controllable under the precise design of drug delivery systems with the most favorable physical properties.2,3

Recently, a number of nano-based ocular drug delivery systems have been developed to overcome the limitations of conventional ones, which have demonstrated promising results in both in vitro and in vivo ocular disease models and clinical practice. They are designed or modified to facilitate drug delivery to the eye by increasing the retention time, promoting drug penetration across barriers, and targeting specific cells and tissues.4

In this review, we aim to provide a snapshot of the nano-based ocular drug delivery systems that have been approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in ocular diseases, along with a description of key drug delivery systems that are emerging in the clinical trial pipeline, and to highlight some examples for topical delivery to the posterior segment of the eye as potential pathways for translational development in recent preclinical studies.

2. Approved and investigational nano-based drug delivery systems

Over the last three decades, various nano-based drug delivery systems have already been approved by the FDA and EMA.5 To date, about nine nano-based ophthalmic drugs have been approved by the FDA and/or EMA (Table 1), while others remain under Phase I, II, and III clinical trials (Table 2).
Table 1 Clinically approved nanodrugs for ocular diseases
Formulation Drug name Active drug Route Approved application/indication Approval (year)
Anterior segment disease
Nano-emulsion Durezol Difluprednate Topical Postoperative ocular inflammation and pain FDA (2008)
Polymeric hydrogel Zirgan Ganciclovir Topical Acute herpetic keratitis FDA (2009)
Nano-emulsion Ikervis Cyclosporine Topical Dry eye disease EMA (2015)
Nanomicelles Cequa Cyclosporine Topical Dry eye disease FDA (2018)
Polymeric hydrogel Timoptic-XE Timolol maleate Topical Ocular hypertension or open-angle glaucoma FDA (2018)
Nanoparticles Inveltys Loteprednol etabonate Topical Postoperative ocular inflammation and pain FDA (2018)
Nanoparticles Eysuvis Loteprednol etabonate Topical Dry eye disease FDA (2020)
Posterior segment disease
Liposome Visudyne Verteporfin Intravenous Subfoveal choroidal neovascularization secondary to age-related macular degeneration FDA (2000) and EMA (2000)
Polymer Macugen Pegaptanib sodium Intravitreal Neovascular age-related macular degeneration FDA (2004) and EMA (2006)


Table 2 Nanodrugs still under investigation (not yet recruiting, recruiting, completed but not clinically approved)
Formulation Drug name Active drug Disease Route Clinical trial phase Clinical trials gov ID Status
Dry eye disease
Nanomicelles VOS (voclosporin ophthalmic solution) Voclosporin Dry eye disease Topical III NCT04147650 Completed
Nanoparticles KPI-121 Loteprednol etabonate Meibomian gland disease Topical II NCT02218489 Completed
Nanoparticles Haporine-S Cyclosporine Dry eye disease Topical III NCT01804361 Completed
Nano-emulsion NOVA22007 (Ikervis® approved by EMA) Cyclosporine Dry eye disease Topical III NCT00814515 Completed
Nano-emulsion TJCS eye drops Cyclosporine Dry eye disease Topical III NCT02461719 Completed
Nano-emulsion Nanodrop (PRO-176) Propylene glycol Dry eye disease Topical Iand II NCT04111965 Not yet recruiting
Nano-emulsion OCU-310 Brimonidine Tartrate Dry eye disease Topical III NCT03785340 Completed
Glaucoma
Liposome POLAT-001 Latanoprost Ocular hypertension, open-angle glaucoma Subconjunctival injection II NCT02466399 Completed
Nanoparticles Intracameral implant ENV515 Travoprost Ocular hypertension, open-angle glaucoma Topical II NCT02371746 Completed
Ocular inflammations
Nanocrystal NCX 4251 Fluticasone propionate Blepharitis Topical II NCT03926026 Completed
Nano-emulsion OCU300 Brimonidine tartrate Ocular graft versus host disease Topical III NCT03591874 Terminated
Nanoparticles DexNP (OCS-01) Dexamethasone Inflammation and pain following cataract surgery Topical II NCT04130802 Completed
Nano-emulsion SVT-15473 Clobetasol propionate Inflammation and pain following cataract surgery Topical III NCT04246801 Completed
Nanomicellar LX211 Voclosporin Non-infectious uveitis Oral III NCT00404612 Completed
Nano-emulsion NOVA22007 Cyclosporine Vernal keratoconjunctivitis Topical II and III NCT00328653 and NCT01751126 Completed
Retinal diseases
Nanoparticles Dexamethasone-cyclodextrin eye drops Dexamethasone Diabetic macular edema Topical II and III NCT01523314 Unknown
Nanoparticles DexNP (OCS-01) Dexamethasone Diabetic macular edema Topical II and III NCT05066997 Recruiting
Nanoparticles TLC399 (ProDex) Dexamethasone Retinal vein occlusion, macula edema Intravitreal II NCT03093701 Completed
Nanoparticles KPI-121 Loteprednol etabonate Intra- or subretinal fluid due to retinal vein occlusion, macula edema Topical II NCT02245516 Completed
Other diseases
Nanoparticles Pluronic®F-127 Urea Cataract Topical II NCT03001466 Completed
Liposomes Coenzyme Q10 Coenzyme Q10 Ataxia-oculomotor apraxia 1 Oral III NCT02333305 Completed
Liposomes Marqibo Vincristine sulfate Metastatic malignant uveal melanoma Intravenous II NCT00506142 Completed
Vincristine sulfate Intraocular retinoblastoma Intravenous III NCT00072384 Completed
Nanoparticles Nab-paclitaxel Paclitaxel Intraocular melanoma Intravenous II NCT00738361 Completed


2.1 Approved nano-based drug delivery systems

The first FDA-approved nano-based drug delivery system was Visudyne®, a verteporfin-loaded liposomal system, used as a photosensitizing agent along with laser light treatment for choroidal neovascularization (CNV) secondary to age-related macular degeneration (AMD). Visudyne® is administered intravenously, followed by directing a red laser through the pupil into the eye 10 minutes after injection. After absorbing the light, it is boosted into an excited state and transmits its energy to ambient oxygen to produce singlet oxygen. This reactive oxygen can stop and even reverse the progressive loss of vision by damaging the newly produced leaking blood vessels.6,7 The results of the “Treatment of Age-Related Macular Degeneration with Photodynamic Therapy (TAP)” study, composed of two multicenter, double-masked, randomized phase-III clinical trials, revealed that, as opposed to the placebo, photodynamic therapy with verteporfin significantly reduced the risk of moderate and severe vision loss in patients with neovascular AMD with 61% versus 46% at 12 months and 53% versus 38% at 24 months (p < 0.001).8 After 20 years of its approval, this drug continues to be widely used and remains the only intravenously injected nano-based drug delivery system, for the treatment of retinal diseases.9,10

In the real sense, Macugen®, a pegaptanib sodium-loaded polyethylene glycol (PEG) aptamer, is the first approved nano-based drug delivery system, for the treatment of neovascular AMD. It is the first in a new class of ophthalmic drugs to specifically target vascular endothelial growth factor (VEGF), a protein that acts as a signal in the case of abnormal blood vessel growth and leakage in neovascular AMD.11 In two pivotal phase II/III, randomized, multicenter, double-blinded clinical trials involving approximately 1190 patients with all subtypes of neovascular AMD, intravitreally injected with 0.3 mg of Macugen®, 70% of patients achieved less vision loss compared with 55% in the placebo group (p < 0.001) at 54 weeks.12 At year 2, patients receiving 0.3 mg of Macugen® experienced a change in vision that was approximately 50% better than those receiving placebo injections. Macugen®, therefore, has the notable distinction of being the first aptamer therapeutic approved for use in humans, shedding light on future aptamer applications.

Other nanocarriers, such as nanomicellar and nanoemulsion, have been widely explored for their potential to treat various ocular disorders. The treatment of anterior segment disease, including dry eye disease, glaucoma, and postoperative ocular inflammation, is the major principal therapeutic target for these delivery systems. Cequa® is a nanotechnology-derived ophthalmic delivery system approved by the FDA for dry eye disease. It is a transparent nanomicellar drug delivery system of a highly hydrophobic drug, cyclosporine A (CsA). In fact, as a highly hydrophobic compound, it is mostly administered as an oily emulsion, but oil-based preparations are poorly tolerated by patients and lead to low bioavailability because CsA is more attracted to the hydrophobic vehicle than the highly hydrophilic tissue. However, the unique CsA nanomicellar delivery system of Cequa® can offer improved stability, safety, and efficacy, as well as lower cost.13 Its safety can be confirmed by low toxicity from the preclinical results performed in human-derived corneal and retinal cells.14 In addition, the insignificant charge of the system helps to prevent rejection by negatively charged cell surfaces, which contributes to improved interaction with the ocular cells.15 In clinical practice, in phase III clinical trial with a total of 745 patients with dry eye disease, both the primary endpoints of the trial, Schirmer's test (a measure of tear production), and secondary endpoints indicated a statistically significant increase.16 It was also clarified in the trial that Cequa® was a highly efficient and safe ophthalmic solution resulting in a clinically meaningful increase in tear production and a large reduction in signs and symptoms of ocular surface inflammation compared to the vehicle in dry eye disease.17–19

2.2 Investigational nano-based drug delivery systems

There are a handful of novel nano-based eye drops currently being investigated in clinical trials to establish their safety and efficacy for the treatment of ocular disorders. A phase III clinical trial was conducted to examine the effects of clobetasol propionate nanoemulsion eye drops (SVT-15473) in patients on the reduction of inflammation and pain after cataract surgery between June 2020 and May 2021 (NCT04246801). SVT-15473 was developed using a patented nanoemulsion technology, IMPACT-SVT® nanoemulsion (emulsion with nanometric-sized droplets), to improve drug–mucus penetration and bioadhesion, and to reduce irritation to the eye.20 The trial involved 212 patients who had recently undergone cataract surgery from 22 hospitals in the United States. Recently, the company submitted a New Drug Application (NDA) to the FDA for approval with favorable results for this innovative nano-based delivery systems. In addition, a randomized, double-masked, vehicle-controlled phase II study of loteprednol etabonate nanoparticle eye drop for the treatment of meibomian gland disease was completed at approximately 8 centers in the United States (NCT02218489).

Although nano-based drug delivery systems intended for topical instillation exhibit the ability to penetrate through the biological membrane and accumulate in the ocular tissues, few have received clinical use approval for the market. There are limited clinical trials that have been conducted for the clinical transition of topical instillation in the management of posterior segment diseases. One is a phase II/III clinical trial under investigation for dexamethasone nanoparticle eye drops (OCS-01) in the treatment of diabetic macular edema (DME) (NCT05066997). The OCS-01 ophthalmic suspension is 1.5% dexamethasone formulated in OPTIREACH technology, which leverages unique characteristics of drug/cyclodextrin nanoparticles to enhance drug permeability and bioavailability in eye tissues. Animal studies and preliminary clinical trials have proven that this cyclodextrin nanoparticle technology has the potential to increase drug concentrations in ocular tissues, particularly the retina, for the treatment of retinal diseases such as DME. It has successfully completed a phase II trial (NCT05343156). The trial achieved its pre-defined efficacy endpoints and demonstrated that OCS-01 eye drops were superior to vehicle eye drops (identical to the active treatment but without dexamethasone) in reducing the central macular thickness and improving visual acuity in 133 patients with DME.21

2.3 Complications in clinical trials

Safety is one of the most important issues relevant to the clinical development of nano-based drug delivery systems intended for ocular application. The significance of eye safety is highlighted by the FDA's guidance for the industry: safety of nanomaterials in cosmetic products (Docket number: FDA-2011-D-0489). The standard eye irritation test for measuring the ocular toxicity of nano-based drug delivery systems is established by The Organization for Economic Co-operation and Development. Although there has been certain attention paid to the safe use of these delivery systems to the eye, research on their toxicity effect on eyes is still in the early stage.

Even though the ocular surface is the first layer that comes in contact with nanomaterials, once they are absorbed into the eyes, the toxic effect may also be caused within the eye or on the inner surface of the retina. Specific literature on the complications of nanoparticles in ocular application remains scarce. The main toxicity caused by nanoparticles is the generation of oxidative stress, inflammation, and interaction with the cell membrane.22

KPI-121 is a nano-based drug delivery system using mucus-penetrating particles to deliver a custom-engineered ocular corticosteroid, loteprednol etabonate, to the ocular surface tissues.23–26 In the clinical trial of its application in dry eye disease, there was a low incidence of treatment-related adverse events (AEs), severe AEs, and serious AEs. The only AE reported with an incidence of more than 1% was pain at the site of instillation, which was reported by 5.2% of subjects in the KPI-121 group and 4.4% of subjects in the placebo group.24 But the pain was transient and mild or moderate in severity.25,26

NOVA22007 is a proprietary cationic emulsion that enables an optimal penetration of CsA through the ocular surface by Novasorb cationic emulsion technology.27 In a multicenter phase III trial of NOVA22007 for the treatment of vernal keratoconjunctivitis in children, instillation site pain was also the most common adverse event, reported by six patients (10.5%) in the four-times-daily treatment regimen group, three patients (5.6%) in the twice-daily treatment regimen group, and two patients (3.4%) in the placebo group.28 In the phase III study for its application in dry eye disease, 31 (4%) patients had a serious adverse event during the initial 6 months period including 15 (3.8%) in the NOVA22007 group and 16 (4.7%) in the vehicle group (cationic emulsion without CsA). Two patients were considered to be drug-related with one related to NOVA22007 (epithelial erosion of the cornea) and the other related to the vehicle (reduced visual acuity).29

3. Topical instillation of eye drops to the posterior segment of the eye

Of all the nano-based drug delivery systems approved and under investigation, topical instillation of eye drops is the most common route for administering therapeutics to the eye. Although this pathway is a viable way of drug delivery for the treatment of anterior segment diseases, such as dry eye disease, glaucoma, inflammation, and pain following ocular surgery, it remains a major challenge to efficiently deliver drugs topically to treat diseases of the posterior segment of the eye.

At present, all commercially available ophthalmic products used for the management of posterior segment diseases are invasive. It is true that intravitreal injection is the most efficient route for the administration of drugs to the posterior segment of the eye. However, this route is associated with various complications, including endophthalmitis, retinal detachment, vitreous hemorrhage, and cataract.30 Even though intravitreal injection is widely used in the clinic, it still demands high patient compliance. In fact, patients may receive fewer injections than those taking part in clinical trials, and thus their treatment outcomes may be worse than anticipated.31 Therefore, patients would benefit greatly if effective delivery could be achieved by a less invasive route of administration.

The absorption of drugs on the ocular surface is limited by various barriers, such as static (cornea, conjunctiva, sclera), dynamic (tear turnover, reflex blinking, conjunctival blood flow, and nasolacrimal drainage), metabolic barriers (phase I & II enzymes, efflux pumps) and intraocular environment (blood–retinal and blood–aqueous barriers), resulting in extremely low drug bioavailability which is usually less than 5% and difficult to be delivered to the posterior segment of the eye.32–35 After topical instillation, drugs may penetrate into the posterior segment via the corneal or/and conjunctival route. Drugs can penetrate through the cornea to the anterior chamber, then pass through the lens/iris to reach the vitreous and retina. Drugs can also diffuse through the sclera, choroid and eventually reach the retina following absorption into the conjunctiva, or they can diffuse from the cornea to the conjunctiva. In addition, a small number of drugs can reach the retina by overcoming the blood–retinal barrier after systemic absorption via nasolacrimal drainage, conjunctival blood vessels, or choroidal circulation (Fig. 1).36


image file: d3bm00505d-f1.tif
Fig. 1 Drug absorption pathways through the cornea/conjunctiva–sclera pathways following topical administration. (1) Corneal pathway is marked in yellow. (2) Conjunctival–scleral pathway is marked in red. (3) Systemic absorption is marked in rose.

Recently, a lot of nano-based drug delivery systems have been developed to overcome the limitations of conventional eye drops and deliver drugs to the retina.37–63 These advanced delivery systems have shown promising outcomes in the ocular disease models both in vitro and in vivo by increasing the retention time of drugs on the cornea, enhancing drug penetration, or promoting drug delivery to the posterior segment of the eye through the corneal or conjunctival–scleral pathway. Herein, we provide a summary of preclinical research on novel nano-based drug delivery systems for topical delivery to the posterior segment of the eye in the recent five years (Table 3).

Table 3 Representative topical nano-formulations in preclinical study in recent 5 years
Formulation Drug Function Pathway Disease Ref.
Liposomes Diclofenac Enhance penetration Corneal Choroidal neovascularization Shimazawa et al.37
Triamcinolone acetonide Enhance penetration, increase retention time Corneal Retinal edema Chen et al.38 and Li et al.39
Triamcinolone acetonide Enhance penetration Corneal Diabetic macular edema Altamirano-Vallejo et al.40 and Navarro-Partida et al.41
Triamcinolone acetonide Increase retention time Corneal Choroidal neovascularization Khalil et al.42
Dexamethasone Enhance penetration, increase retention time Non-corneal Posterior ocular tissues Gu et al.43
TGFβ1 Enhance penetration Corneal Age-related macular degeneration Platania et al.44
Lipid nanoparticles Myriocin Enhance penetration Conjunctival/scleral Retinitis pigmentosa Platania et al.45
Atorvastatin Enhance penetration, increase retention time Corneal, conjunctival/scleral Age-related macular degeneration Yadav et al.46
Triamcinolone acetonide Enhance penetration, increase retention time Corneal Posterior ocular tissues Tatke et al.47
Melatonin Enhance penetration, increase retention time Corneal Diabetic retinopathy Romeo et al.48
Verteporfin Enhance penetration Conjunctival/scleral Age-related macular degeneration Ran et al.49
Polymer nanoparticles Apatinib Enhance penetration, increase retention time Corneal Diabetic retinopathy Radwan et al.50
siRNA/antisense oligonucleotides Enhance penetration Non-corneal Retinoblastoma Jiang et al.51
Inorganic nanoparticles Cerium oxide nanoparticles Enhance penetration Conjunctival/Scleral Age-related macular degeneration Badia et al.52
Gold nanoparticles Enhance penetration Corneal Diabetic retinopathy Apaolaza et al.53
Guanabenz and valproic acid (magnetic nanoparticles) Enhance penetration by MRI Corneal Barded–Biedl syndrome related retinal degeneration Bassetto et al.54
Nanomicelles Aflibercept Enhance penetration, increase retention time Corneal, conjunctival/scleral Choroidal neovascularization Zhao et al.55
Doxorubicin Enhance penetration, increase retention time Conjunctival/scleral Oxygen-induced retinopathy Li et al.56
Anti-PDL1, anti-VEGFA Enhance penetration Conjunctival/scleral Choroidal neovascularization, choroidal melanoma Shen et al.57
Nanoemulsion Lutein Enhance penetration, increase retention time Non-corneal Age-related macular degeneration Ge et al.58
Ro5-3335 Increase retention time Not mentioned Ocular neovascularization, proliferative vitreoretinopathy Delgado-Tirado et al.59 and Delgado-Tirado et al.60
Dendrimer Not mentioned Enhance penetration Conjunctival/scleral Choroidal neovascularization Yang et al.61
Antisense oligonucleotides Enhance penetration Corneal Posterior ocular tissues Tai et al.62
Berberine hydrochloride, chrysophanol Enhance penetration Conjunctival/scleral Age-related macular degeneration Lai et al.63


3.1 Liposomes

Liposomes are spherical vesicles composed of one or more lipid bilayers and an aqueous core. Both hydrophilic and lipophilic drugs can be encapsulated in liposomes, with hydrophilic drugs encapsulated in the core and lipophilic drugs in the bilayer. Liposomes have been widely used in the development of topical therapeutics with increased residence time for drug absorption, protection of the encapsulated drug from the external environment, and enhanced drug penetration to ocular tissues.63,64 A recent study developed and evaluated the potential of triamcinolone acetonide-loaded liposomes, formed by soybean phosphatidylcholine and cholesterol, coated with chitosan by the calcium acetate gradient method as a novel drug delivery system enhancing the efficacy of triamcinolone acetonide as eye drops to the retina. Chitosan was chosen as the surface modification of liposomes due to its special properties of bioadhesion to the cornea and enhanced penetration. After modification, the cationic liposomes have a high binding affinity to the corneal surface, which interacts with the negative charge of the ocular surface to prolong drug retention and improve local drug concentration. This system was found to have a prolonged retention time on the ocular surface, which enhanced the absorption of cargo through the corneal pathway.37,38 Altamirano-Vallejo et al. also developed triamcinolone acetonide-loaded liposome formulations (TALF), self-forming synthetic PEGylated lipids (QuSomes®) containing TA, to enhance the delivery of TA into the posterior segment of the eye. TALF was able to cross the cornea and deliver TA to the vitreous body and retina, reaching the highest peak at 12 h.40 Moreover, in the phase I clinical study, TALF was able to reduce the thickness of the fovea in patients with adequate glycemic control but the presence of DME with good ocular tolerability.41

Liposomes modified by penetration-enhanced polymers can not only entrap drug molecules but also target the desired sites via binding to the cornea surface. Gu et al. prepared a novel nanocomposite eye drop by hybridizing dexamethasone-loaded liposomes, made by soybean phospholipids and cholesterol, with glycylsarcosine (GS)-anchored layered double hydroxides (LDH).43 GS is a classical substrate for PepT-1, which is used for targeting PepT-1 of the ocular surface and was modified on LDH. LDH, a kind of positive carrier, can increase precorneal retention through electrostatic adsorption. Combining the advantages of all compounds mentioned above, the formulation showed prolonged precorneal retention time by 2.0 times and superior in vitro permeability than commercial dexamethasone eye drops. Furthermore, the formulations also displayed higher drug concentrations in choroid-retina tissue compared with other formulations composed of single LDH (Fig. 2).


image file: d3bm00505d-f2.tif
Fig. 2 Dexamethasone (DEXP)-loaded liposomes with glycylsarcosine (GS)-anchored layered double hydroxides (LDH) can facilitate the noninvasive delivery of drugs to the posterior segment of the eye. (A) Synthesis of DEXP-HSPC@LDH-GS nanocomposites by an ion-exchange method. (B) Schematic illustration of topical delivery of DEXP to the posterior segment of the eye through a non-corneal pathway. Fluorescent images of frozen sections of cornea (C), conjunctiva (D), and sclera–choroid–retina (E) of rabbit eyes after topical administration of DOX-HSPC@LDH-GS nanocomposites. Scale bar, 50 μm.43 Copyright © 2019, American Chemical Society.

3.2 Lipid nanoparticles

Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) play a key role in a major shift in eye disease therapy. These lipid-based systems have compatible characteristics with biological membranes, which may promote mucus adhesion, thus improving drug uptake when compared to conventional drug delivery systems. In addition, they also allow modified drug release and a reduction in the required dose for topical instillation.65 Yadav et al. designed an SLN system loaded with atorvastatin (ATS-SLNs) as topical therapeutics for AMD. The ATS-SLNs, comprising atorvastatin, Compritol® 888 ATO, PEG400, Poloxamer 188 (P188) and Phospholipon 90H (P 90H), were prepared by a high-pressure homogenization method and were suitable to provide enhanced permeation through the cornea and attained higher drug bioavailability in both the aqueous and vitreous humor.

They found that ATS-SLNs were 8 and 12 times more bioavailable in the aqueous and vitreous humor, respectively, compared to free ATS. The AST-SLNs were observed on the corneal surface for up to 7 h, 4.7 times longer than the free solution after topical instillation. The prolonged residence on the eye may be attributed to their mucus penetration effects because the particles contain PEG400 and P188, and both these polymers have been established as effective mucopenetrating agents. The particles were able to penetrate through the mucus and reach the corneal surface with the help of P188, which coats the particle surfaces by adhering hydrophobic poly-propylene oxide segments and leaving a dense brush of uncharged, hydrophilic segments sticking out from the particle surface.46 Tatke et al. developed a similar TA-loaded SLN in situ gel (TA-SLN-IG), comprising TA, Compritol® 888 ATO, glyceryl monostearate, Tween® 80, P188 and glycerin, for enhanced topical ocular delivery to the posterior segment of the eye. The introduction of the in situ gelling agent with SLN will keep the formulation for a longer period of time, thus increasing the pre-corneal residence time. The rheological and trans-corneal permeability of TA-SLN and TA-SLN-IG were 10.2 and 9.3 fold higher, respectively, compared to the TA-control, with a higher tear concentration of 13.3 μg mL−1 at 2 hours, indicating an enhanced precorneal residence time. The enhanced retention time of TA-SLN-IG on the ocular surface was mainly due to cross-linking of the polymer chains mediated by the cations in the tear fluid, resulting in gel formation on the ocular surface, which contributes to an extended corneal contact time.47

In order to improve the encapsulation efficiency and achieve well-controlled release kinetics, a new generation of nanoparticles, lipid–polymer hybrid nanoparticles (LPHNs), has been designed.66 Romeo et al. developed and optimized melatonin-loaded LPHNs (mel-LPHNs) using the Design of Experiment as a safe hybrid platform suitable for topical instillation of diabetic retinopathy. With the aim of enhancing corneal retention time, the nanoparticles consisted of the PLGA-PEG polymer coated with a cationic lipid shell. PLGA-PEG was used as a mucopenetrating agent and cationic lipids were used to enhance mucoadhesion through electrostatic interaction with the anionic ocular mucosa. After topical instillation, mel-LPHNs displayed a high encapsulation efficiency of 79.8%, suitable pH and osmolarity values, good mucoadhesive properties, and a controlled release profile for over 8 days. Furthermore, biological evaluation in an in vitro model of diabetic retinopathy demonstrated enhanced neuroprotective and antioxidant activities of mel-LPHNs, when compared to melatonin aqueous solution at the same concentration.48

Ran et al. designed a low-density lipoprotein-inspired nanoparticle (PEN-rLDL-VP) with high verteporfin (VP) encapsulation efficacy and neovascularization recognizability for the targeted photodynamic (PDT) of wet AMD. VP was protected inside the hydrophobic core of reconstituted LDL (rLDL) vectors, and 5-carboxyfluorescein (FAM) conjugated penetratin (PEN) was anchored on the surface of the rLDL carrier, which allowed the nanoparticles (PEN-rLDL-VP) to pass across the blood–retina barrier to enable visual therapy. After topical instillation for only a single dose, PEN-rLDL-VP was able to deliver VP into the neovasculature to respond to PDT therapy, and decreased neovascularization and inflammation were observed afterwards (Fig. 3).49


image file: d3bm00505d-f3.tif
Fig. 3 Neovascularization-directed bionic eye drops for the treatment of age-related macular degeneration. (A) Schematic illustration of the composition of PEN-rLDL-VP nanoparticles and treatment of wet age-related macular degeneration after topical instillation. (B) Representative images of fluorescence fundus angiograms from the healthy, untreated, rLDL-VP, and PEN-rLDL-VP groups and quantification analyses of CNV areas on the 24th day of CNV induction. (C) Representative images for retinal sections of experimental groups stained with H&E on the 28th day (400×).49 Copyright © 2022, the author(s).

3.3 Polymer nanoparticles

Polymer nanoparticles are ideal for ocular drug delivery to the desired sites due to the diversity of polymers in their compositions. Recently, several hybrid polymer nanoparticle-based eye drop delivery systems have been prepared mainly focusing on the ability to penetrate deeper into the posterior segments of the eye, resulting in the localized delivery of drugs at high dosages and enriching the local drug concentration in the retina.

Radwan et al. investigated the feasibility of bovine serum albumin (BSA) nanoparticles coated with hyaluronic acid (HA), containing apatinib, a selective inhibitor of VEGF receptor 2, to the retina via topical instillation in diabetic retinopathy. The mucoadhesive nature of HA, as well as its interaction with hyaluronan receptors, on corneal epithelial cells may result in prolonged precorneal retention. The characteristics of BSA-NPs prepared in this work, including the highly negative surface charge (−29.5 ± 0.05 and −37.3 ± 1.8 mV) and the relatively small particle size (212 ± 0.35 and 222.2 ± 3.56 nm) of uncoated and HA-coated nanoparticles, respectively, allowed avoiding particle trapping within the vitreous meshwork, thus allowing them to freely diffuse towards the retina. In addition, the HA coating also worked to enhance apatinib delivery to actively target CD44 receptor positive retinal cells by receptor-mediated endocytosis.50

Polymer nanoparticles can also be used as carriers for gene delivery to the retina with the assistance of cell-penetrating peptides. An octopus-like 8-valent penetratin (VP) composed of a biocompatible multi-arm PEG core and several outspread penetratin tentacles was designed to effectively deliver siRNA or antisense oligonucleotides (ASOs) into retinoblastoma-bearing mice. The formation of a branched spatial structure and flexible cationic penetratin tentacles of 8VP significantly improved cancer cellular efficiency (approaching 100%) and transfection rate (over 75%). After topical instillation in the retinoblastoma-bearing mice model, 8VP enabled rapid (<10 min) and prolonged (>6 h) distribution of nucleic acids in the retina via a noncorneal pathway and efficiently inhibited the protein expression of intraocular tumor without toxicity (Fig. 4). This non-viral vector nanostructure provided a promising strategy for non-invasive gene delivery therapies for retinoblastoma treatment.51


image file: d3bm00505d-f4.tif
Fig. 4 Topical delivery of nucleic acids by octopus-like 8-valent penetratin polymer nanoparticles for the treatment of retinoblastoma. (A) Construction of multivalent penetratin (MVP) forms polyplexes with antisense oligonucleotides (ASO). (B) The polyplexes are formed as an octopus-like nanostructure that carry some cargo (nucleic acids) by binding cationic penetratin tentacles with anionic nucleic acids and moving forward by nonbinding tentacles to mediate the noninvasive intraocular delivery. (C and D) Intraocular distribution of aso in the whole eye, cornea, and retina treated with different polyplexes. Epi, epithelium; endo, endothelium; gcl, ganglion cell layer. (E) Semiquantitative inhibition efficiency on bioluminescence fluc expression of the tumor. Scale bars, 500 μm for (C) and 100 μm for (D).51 Copyright © 2019, American Chemical Society.

3.4 Inorganic nanoparticles

Inorganic nanoparticles, including silicone, iron oxide, zinc oxide, cerium oxide, gold, silver, and magnetic nanoparticles have been investigated for their potential in ocular drug delivery.67 These nanoparticles are gaining popularity as a result of their antioxidant, anti-inflammatory, antiangiogenic and magnetic properties. Cerium oxide nanoparticles (CeO2NPs) were found to have minimal toxicity to normal tissues while providing cellular protection from ROS-dependent oxidative damage. Badia et al. prepared a delivery system of 3 nm CeO2NPs which were monodispersed in an aqueous suspension stabilized by sodium citrate. The ultrasmall nanoparticles usually have a strong propensity to aggregate, making it difficult to create single CeO2NP colloidal dispersions in water. To overcome this problem, Badia et al. were able to make CeO2NPs stable in water by starting with sodium citrate complexed cerium ions rather than conventional cerium nitrate and using TMAOH as the base. The non-aggregation characteristics helped in the topical instillation of the nanoparticles to reach the retina and achieved a beneficial therapeutic effect. The system displayed the capacity to activate the expression of genes related to the antioxidant response, reduce the expression of inflammatory lesions, and decrease CNV both in vitro and in vivo (Fig. 5).52
image file: d3bm00505d-f5.tif
Fig. 5 Non-aggregated biocompatible cerium oxide nanoparticles (CeO2NPs) in the treatment of dry and wet forms of AMD as a beneficial antioxidant and a neuroprotective agent. (A) In vitro results indicated the biocompatible and protective character of 3 nm CeO2NPs in reducing oxidative stress in arpe19 cells and inhibiting neovascularization in both huvec and in vitro models of neovascular growth. (B) Schematic illustration of topical delivery of CeO2NPs to the retina through the conjunctival–scleral pathway. (C) Fundoscopy and optical coherence tomography (oct) images of DKOrd8 mice 2 months after treatment with CeO2NPs or vehicle, and C57BL/6N mice used as control. (D) Microglia quantification in RPE a–c and retinal flatmounts e–g of DKOrd8 mice stained with iba1 after the 2-month treatment. Scale bars, 500 μm for a–c and 100 μm for e–g. (E) Representative fundus, fluorescein angiography and oct images of laser induced CNV mouse eyes treated with CeO2NPs or vehicle for 7 days (F) Graphs comparing CNV area measured from RPE–choroid–sclera flatmounts.52 Copyright © 2023, American Chemical Society.

Gold nanoparticles possess several advantages, such as relatively small size, high biocompatibility, antioxidant and antiangiogenic features.68 Apaolaza et al. designed HA-coated gold nanoparticles for topical instillation to reach the retina. By modifying with HA, the gold nanoparticles were able to cross the physiological barrier of the eye and reach the retina. The HA coating helped to enhance the mobility of the nanoparticles in the vitreous and increase stability and distribution by a specific CD44 receptor interaction. As a result, protective and antiangiogenic effect was achieved by significant suppression of neovascularization and advanced glycation end mediated retinal pigment epithelial cell death after topical instillation.53

Magnetic nanoparticles are different from other nanocarriers due to the magnetic properties that make them unique for drug delivery. Recently, Bassetto et al. developed a topical, noninvasive, and magnetically aided delivery system that cargo guanabenz and valproic acid on the surface of Fe3O4 nanoparticles through anti-unfolded protein response towards the retina. Assisted by magnetic resonance imaging, the nanoparticles were successfully delivered to the photoreceptors after being topically applied onto the ocular surface of Bbs knockout and wild-type mice. Moreover, a therapeutic effect was seen in the mice models with significant amelioration of the photoreceptors’ functionality by electroretinogram.54

3.5 Nanomicelles

Nanomicelles are nanosized amphiphilic core–shell structures with a hydrophobic core, hydrophilic shell, and polymeric surfactants.69 Nanomicelles are emerging as a promising platform for the delivery of poorly water-soluble drugs to the eye due to their increased bioavailability, enhanced corneal permeation, and increased solubility and stability of drugs.70–73 Zhao et al. reported a nanomicelle drug delivery system made of a copolymer EPC (nEPC), comprising PEG, polypropylene glycol (PPG), and polycaprolactone (PCL) for the topical instillation of aflibercept to the posterior segment of the eye. nEPCs were made by concentrating EPC above the critical micelle concentration but below the concentration required for sol–gel transition. Aflibercept was then encapsulated by nEPCs through direct mixing. The systems of nEPC loaded with aflibercept could penetrate the cornea and deliver aflibercept to the retina with a therapeutic effect on CNV murine models. Interestingly, nEPCs exhibit intrinsic antiangiogenic properties by inhibiting VEGF-driven angiogenesis pathways responsible for endothelial cell proliferation and tube formation instead of haptotaxis, which is driven by extracellular matrix components such as collagen. The intrinsic antiangiogenic properties of nEPCs and their ability to deliver anti-VEGF drugs may result in a synergistic effect for the treatment of CNV.55

Li et al. designed a co-assembled glycopeptide nanotransforrs (GPNTs), consisting of glycopeptide, cationic peptide, and doxorubicin, which can effectively penetrate the cornea and sclera barriers, and then target M2 macrophages and release drugs to alleviate fundus neovascularization. The two peptide amphiphiles were modularly designed with hydrophobic, enzyme-responsive, and hydrophilic motifs. The appropriate ratio of these components enabled the legumain trigged transformation. This was not a typical delivery system of nanomicelles, because it was transformed from nanoparticles to a fiber-like structure induced by legumain cleavage intracellularly after mannose receptor-mediated phagocytosis. This dynamic transformation not only strengthened lysosome escape and prevented exocytosis, but also enhanced drug accumulation and M2 macrophage apoptosis. After topical instillation, pathological neovascularization branches and cell nuclei that broke through the inner limiting membrane were reduced by 55% and 72%, respectively, 25% and 20% less than those in the non-transformed controls (Fig. 6).56


image file: d3bm00505d-f6.tif
Fig. 6 The therapeutic effect of co-assembled glycopeptide nanotransforrs (GPNTs), MRP@DOX, on fundus neovascularization. (A) Construction of MRP@DOX, by the co-assembly of amphiphilic peptides of glycopeptide, cationic peptide, and doxorubicin. (B) Schematic illustration of topical delivery of MRP@DOX for the treatment of neovascularization in five steps: (i) penetration through the cornea and sclera barriers efficiently; (ii) mannose receptor (MR) targeting mediated phagocytosis into M2 macrophages; (iii) legumain triggered transformation instructed lysosome escape; (iv) DOX induced M2 macrophages apoptosis; (v) M2 macrophages elimination strengthened neovascularization. (C) Representative images of retinal flatmounts in the central area and enlarged view of peripheral neovascularization area treated by different agents. The white dashed line represented no perfusion area. The green arrow indicated the hemorrhagic point. The asterisk marked the branch site of the blood vessels in the unit area. Scale bar, 100 μm.56 Copyright © 2021, Elsevier.

Shen et al. prepared a fluorocarbon-modified chitosan (FCS) for macromolecular ophthalmic drug delivery, which was able to self-assemble with therapeutic proteins via electronic interactions to form stable nanocomplexes. The delivery system showed effective ocular penetration ability by temporally opening the tight junctions in both cornea and conjunctiva tissue barriers. Furthermore, the fluorocarbon chains in FCS are nonhydrophilic and nonlipophilic by nature, which would probably make such nanocomplexes less “sticky”, facilitating their intraocular penetration. In both choroidal melanoma-bearing mouse model and choroidal neovascularization-bearing mouse/rabbit models, FCS/anti-programmed cell death ligand 1 (PDL1) or FCS/anti-VEGFA eyedrops showed excellent therapeutic outcomes, comparable to those with intravenous or intravitreal injections.57

3.6 Nanoemulsions

Nanoemulsions are one of the most promising carriers applied topically onto the eye with a variety of properties, such as improved bioavailability, good in vivo biocompatibility, prolonged residence time, and enhanced corneal drug permeation.74 Ge et al. prepared a penetratin-modified lutein nanoemulsion for the treatment of AMD. Lutein was loaded into the nanoemulsion to improve water solubility. Stearyl penetratin was added to the nanoemulsion to enhance penetration via a noninvasive administration route. In order to extend corneal retention duration and enable penetratin to reach its full effect, the penetratin nanoemulsion was prepared as an ion-responsive in situ gel. With the aid of penetratin, the nanoemulsion was rapidly delivered to the posterior segment of the eye and distributed in the retinal region. This system showed good efficacy in the protection of the retinal cells from damage caused by ROS and apoptosis in the dry AMD mouse model.58 In addition, nanoemulsions can also be ideal nanocarriers when exploring the mechanism of small-molecule drugs. With the aim to deliver a hydrophobic small molecule, Ro5-3335, a runt-related transcription factor 1 (RUNX1) inhibitor, to the retina, Delgado-Tirado et al. designed a nanoemulsion delivery system utilizing surfactants as the encapsulation matrix within an aqueous phase. After topical instillation of this system, Ro5-3335 was detected to be 2.67 ng mL−1 within the vitreous cavity and effectively reduced the progression of proliferative vitreoretinopathy in the rabbit model.59 Such a system was also effective in the treatment of both corneal and choroidal neovascularization in animal models.60

3.7 Dendrimers

Dendrimers are spherical, tree-like nanostructures consisting of one core and many side-chain molecules that branch out to form a three-dimensional structure.75 With plenty of peripheral functional groups, dendrimers can receive surface modifications, which endow them with various properties for ocular drug delivery, such as enhanced permeation, and targeted delivery to specific sites.76

Recent studies on dendrimers for topical instillation and delivering the drug to the posterior segment of the eye mainly focused on enhancing their ability of penetration. As a powerful permeation enhancer in ophthalmic administration, modification of penetratin on dendrimers is an effective strategy.77 Yang et al. designed a novel penetratin and cyclic arginine–glycine–aspartate (RGD) co-modified PEGylation polyamidoamine (PAMAM) G4.0 as a nanocarrier with a significantly improved permeability of 1.5 times and extended retinal retention time of more than 12 h, as compared to PAMAM without modifications. At the same time, the RGD peptide worked to enhance the affinity toward integrin αγβ3, which validated the targeting of neovascularization.61 Penetratin also plays an important role in ocular gene delivery to the posterior segment of the eye. In the study of Tai et al., a penetratin-modified PAMAM G5.0/HA complex was established to deliver ASOs to the retina via topical instillation. Compared with other nano-based drug delivery systems, this complex exhibited much more distribution in the retina and prolonged the retention time of ASOs in the retina for more than 8 h.62

The amino groups in PAMAM have mucoadhesive properties by interacting with the negatively charged cellular membrane, thus improving the therapeutic effects.78 Lai et al. developed a liposome system coated with PAMAM G3.0 that simultaneously entrapped berberine hydrochloride (BBH) and chrysophanol (CHR) for topical instillation drug delivery to the retina. The PAMAM G3.0 coating could not only enhance bio-adhesion on corneal epithelium but also allow greater permeability of negatively charged molecules than of positively charged molecules into the corneal layer due to diffusion through the sclera. After topical instillation, the accumulation of BBH and CHR was effectively promoted by the system, and a better therapeutic effect was achieved in reducing ROS levels and protecting against light-induced retinal damage.63

4. Future perspectives

Although topical drug delivery to the posterior segment of the eye is challenging, there remains an emerging number of nano-based drug delivery systems aiming at delivering drugs or nucleic acids to the retina via this non-invasive pathway. There is a leading trend in the design and development towards active targeting to increase drug concentration in specific sites, increased complexity in the synthesis with multiple functions that combine therapeutic and diagnostic capabilities and leverage the non-corneal pathway to facilitate drug delivery to the retina (Fig. 7).
image file: d3bm00505d-f7.tif
Fig. 7 Summary of future perspectives on topical drug delivery. Increasing attention has been paid to these three aspects aiming at delivering drugs or nucleic acids to the retina through this non-invasive pathway.

A lot of novel nanocarriers are introduced to improve ocular permeability and bioavailability, but their distribution in ocular tissues, especially in the retina, after topical instillation still needs to be tested. The actively targeted therapies will be able to increase drug concentration in specific sites, such as RPE, photoreceptors, retinal or choroidal neovascular, thus further enhancing the bioavailability and efficacy with reduced toxicity to normal ocular tissues.79 But the balance among rates of drug release, absorption, and selective target binding that encourage sufficient and uniform drug distribution throughout the retinal lesions remains a challenge in the design and development.

Simple functional nano-based delivery systems can no longer meet the current need for ocular drug delivery. The trend has moved toward complex and multi-component materials that blur the boundaries in traditional categories, even though the increased complexity may add to the difficulties and costs for the sake of large-scale production for clinical translation in the future. Not only is the design more complicated in the synthesis but delivery systems are also made to be multifunctional that combine diagnostic and therapeutic functions, enabling visual tracking during the ocular disease treatment.80–82 These delivery systems are still in preclinical trials and not yet in clinical trials.

The conjunctival–scleral pathway, a type of noncorneal pathway, is in principle the one followed by topical instillation to reach the retina, though this pathway is still under investigation and debate regarding the potential for therapeutic concertation. The surface area of the conjunctiva is 17 times larger than that of the cornea. In addition, the conjunctiva showed superior permeability to drugs compared to the cornea.83 Hydrophilic and small-sized formulations or molecules can diffuse across the conjunctiva and sclera from the ocular surface into the eye without entering the aqueous humor. Given that the openings in the conjunctival epithelium are larger than those in the cornea, it is possible that large-molecule drugs can also penetrate via this route.84 Cell-penetrating peptides are common examples of modifications to nanocarriers to improve drug delivery to the retina from the ocular surface. These peptides often make in vivo uptake into the retina easier via the conjunctival–scleral pathway.85

5. Conclusions

The involvement of nanomedicine in ocular diseases has brought numerous advancements over conventional treatments. Topical instillation, as a noninvasive way, is an optimal option for many ocular diseases due to its potential for eliminating the risks of intravitreal injection and the toxicity of systemic drug delivery. Although the delivery of drugs safely and efficiently into lesions in the posterior segment of the eye topically is a challenging task, relentless efforts have been dedicated to the development of novel nano-based delivery systems in the hope of possible clinical translation from bench to bedside. With ongoing innovations in this field, safe and effective eye drops are expected to revolutionize the present invasive treatment in the foreseeable future.

Conflicts of interest

There are no conflicts to declare.

Acknowledgements

This study was funded by the National Natural Science Foundation of China (82000939), the Foundation of National Infrastructures for Translational Medicine (Shanghai) (TMSK-2021-123), the Innovative Research Team of High-level Local Universities in Shanghai (SHSMU-ZDCX20210700), the Shanghai Pujiang Program (22PJD036), the Science and Technology Commission of Shanghai Municipality (20DZ22780800), and the Shanghai Key Clinical Specialty, Shanghai Eye Disease Research Center (2022ZZ01003).

References

  1. B. Y. Kim, J. T. Rutka and W. C. Chan, Nanomedicine, N. Engl. J. Med., 2010, 363(25), 2434–2443 CrossRef CAS PubMed .
  2. W. Poon, B. R. Kingston, B. Ouyang, W. Ngo and W. C. W. Chan, A framework for designing delivery systems, Nat. Nanotechnol., 2020, 15(10), 819–829 CrossRef CAS PubMed .
  3. L. Y. Chou, K. Ming and W. C. Chan, Strategies for the intracellular delivery of nanoparticles, Chem. Soc. Rev., 2011, 40(1), 233–245 RSC .
  4. Z. Tang, X. Fan, Y. Chen and P. Gu, Ocular nanomedicine, Adv. Sci., 2022, 9(15), e2003699 CrossRef PubMed .
  5. V. Weissig, T. K. Pettinger and N. Murdock, Nanopharmaceuticals (part 1): products on the market, Int. J. Nanomed., 2014, 9, 4357–4373 CrossRef CAS PubMed .
  6. A. Hogan, U. Behan and D. J. Kilmartin, Outcomes after combination photodynamic therapy and immunosuppression for inflammatory subfoveal choroidal neovascularisation, Br. J. Ophthalmol., 2005, 89(9), 1109–1111 CrossRef CAS PubMed .
  7. A. H. Rogers, J. S. Duker, N. Nichols and B. J. Baker, Photodynamic therapy of idiopathic and inflammatory choroidal neovascularization in young adults, Ophthalmology, 2003, 110(7), 1315–1320 CrossRef PubMed .
  8. N. M. Bressler and Treatment of Age-Related Macular Degeneration with Photodynamic Therapy (TAP) Study Group, Photodynamic therapy of subfoveal choroidal neovascularization in age-related macular degeneration with verteporfin: two-year results of 2 randomized clinical trials-tap report 2, Arch. Ophthalmol., 2001, 119(2), 198–207 CAS .
  9. M. J. Sirks, E. H. C. van Dijk, N. Rosenberg, C. E. M. Hollak, S. Aslanis, C. M. G. Cheung, I. Chowers, C. M. Eandi, K. B. Freund, F. G. Holz, P. K. Kaiser, A. J. Lotery, K. Ohno-Matsui, G. Querques, Y. Subhi, R. Tadayoni, C. C. Wykoff, D. Zur, R. M. H. Diederen, C. J. F. Boon and R. O. Schlingemann, Clinical impact of the worldwide shortage of verteporfin (Visudyne®) on ophthalmic care, Acta Ophthalmol., 2022, 100(7), e1522–e1532 Search PubMed .
  10. C. M. G. Cheung, C. S. Tan, R. Patalauskaite, P. Margaron and T. Y. Y. Lai, Ranibizumab with or without verteporfin photodynamic therapy for polypoidal choroidal vasculopathy: predictors of visual and anatomical response in the EVEREST II study, Retina, 2021, 41(2), 387–392 CrossRef CAS PubMed .
  11. VEGF Inhibition Study in Ocular Neovascularization (V.I.S.I.O.N.) Clinical Trial Group, et al., Pegaptanib sodium for neovascular age-related macular degeneration: two-year safety results of the two prospective, multicenter, controlled clinical trials, Ophthalmology, 2006, 113(6), 992–1001 CrossRef PubMed .
  12. E. S. Gragoudas, A. P. Adamis, E. T. Cunningham Jr., M. Feinsod, D. R. Guyer and VEGF Inhibition Study in Ocular Neovascularization Clinical Trial Group, Pegaptanib for neovascular age-related macular degeneration, N. Engl. J. Med., 2004, 351(27), 2805–2816 CrossRef CAS PubMed .
  13. A. Mandal, V. Gote, D. Pal, A. Ogundele and A. K. Mitra, Ocular Pharmacokinetics of a Topical Ophthalmic Nanomicellar Solution of Cyclosporine (Cequa®) for Dry Eye Disease, Pharm. Res., 2019, 36(2), 36 CrossRef PubMed .
  14. K. Cholkar, B. C. Gilger and A. K. Mitra, Topical, aqueous, clear cyclosporine formulation design for anterior and posterior ocular delivery, Transl. Vis. Sci. Technol., 2015, 4(3), 1 CrossRef PubMed .
  15. A. Dogra, K. Kaur, J. Ali, S. Baboota, R. S. Narang and J. K. Narang, Nanoformulations For Ocular Delivery of Drugs - A Patent Perspective, Recent Pat. Drug Delivery Formulation, 2019, 13(4), 255–272 CrossRef CAS PubMed .
  16. D. F. Goldberg, R. P. Malhotra, B. A. Schechter, A. Justice, S. L. Weiss and J. D. Sheppard, A Phase 3, Randomized, Double-Masked Study of OTX-101 Ophthalmic Solution 0.09% in the Treatment of Dry Eye Disease, Ophthalmology, 2019, 126(9), 1230–1237 CrossRef PubMed .
  17. M. Toyos, P. K. Gupta, B. Mitchell and P. Karpecki, The Effect of OTX-101 on Tear Production in Patients with Severe Tear-deficient Dry Eye Disease: A Pooled Analysis of Phase 2b/3 and Phase 3 Studies, Curr. Eye Res., 2022, 47(2), 220–224 CrossRef CAS PubMed .
  18. J. Sheppard, S. Kannarr, J. Luchs, R. Malhotra, A. Justice, A. Ogundele, C. Darby and J. Bacharach, Efficacy and Safety of OTX-101, a Novel Nanomicellar Formulation of Cyclosporine A, for the Treatment of Keratoconjunctivitis Sicca: Pooled Analysis of a Phase 2b/3 and Phase 3 Study, Eye & Contact Lens, 2020, 46(Suppl 1), S14–S19 CrossRef PubMed .
  19. R. Malhotra, D. K. Devries, J. Luchs, A. Kabat, B. A. Schechter, B. Shen Lee, L. Shettle, R. Smyth-Medina, A. Ogundele, C. Darby, J. Bacharach and P. Karpecki, Effect of OTX-101, a Novel Nanomicellar Formulation of Cyclosporine A, on Corneal Staining in Patients With Keratoconjunctivitis Sicca: A Pooled Analysis of Phase 2b/3 and Phase 3 Studies, Cornea, 2019, 38(10), 1259–1265 CrossRef PubMed .
  20. S. Moghassemi, A. Dadashzadeh, R. B. Azevedo and C. A. Amorim, Nanoemulsion applications in photodynamic therapy, J. Controlled Release, 2022, 351, 164–173 CrossRef CAS PubMed .
  21. E. Stefansson, T. Loftsson, M. Larsen, A. Papp, K. Kaarniranta, M. R. Munk, P. Dugel and R. Tadayoni, DX-211 study group, Topical treatment of diabetic macular edema using dexamethasone ophthalmic suspension: A randomized, double-masked, vehicle-controlled study, Acta Ophthalmol., 2023, 101(1), 22–33 CrossRef CAS PubMed .
  22. T. W. Prow, Toxicity of nanomaterials to the eye, Wiley Interdiscip. Rev.: Nanomed. Nanobiotechnol., 2010, 2(4), 317–333 CAS .
  23. M. Korenfeld, K. K. Nichols, D. Goldberg, D. Evans, K. Sall, G. Foulks, S. Coultas and K. Brazzell, Safety of KPI-121 Ophthalmic Suspension 0.25% in Patients With Dry Eye Disease: A Pooled Analysis of 4 Multicenter, Randomized, Vehicle-Controlled Studies, Cornea, 2021, 40(5), 564–570 CrossRef PubMed .
  24. M. Korenfeld, K. K. Nichols, D. Goldberg, D. Evans, K. Sall, G. Foulks, S. Coultas and K. Brazzell, Safety of KPI-121 Ophthalmic Suspension 0.25% in Patients With Dry Eye Disease: A Pooled Analysis of 4 Multicenter, Randomized, Vehicle-Controlled Studies, Cornea, 2021, 40(5), 564–570 CrossRef PubMed .
  25. T. Kim, K. Sall, E. J. Holland, R. K. Brazzell, S. Coultas and P. K. Gupta, Safety and efficacy of twice daily administration of KPI-121 1% for ocular inflammation and pain following cataract surgery, Clin. Ophthalmol., 2018, 13, 69–86 CrossRef PubMed .
  26. K. A. Beckman, J. A. Katz, P. A. Majmudar, A. G. Rips, N. S. Vaidya and A. T. Rostov, KPI-121 1% for pain and inflammation in ocular surgery, Pain Manage., 2022, 12(1), 17–23 CrossRef CAS PubMed .
  27. F. Lallemand, P. Daull, S. Benita, R. Buggage and J. S. Garrigue, Successfully improving ocular drug delivery using the cationic nanoemulsion, novasorb, J. Drug Delivery, 2012, 2012, 604204 Search PubMed .
  28. Clinical Study Report: NVG09B113, A multicenter, randomized, double-masked, 3 parallel arms, placebo controlled study to assess the efficacy and safety of NOVA22007 1 mg/ml (ciclosporin/cyclosporine) eye drops, emulsion administered in paediatric patients with active severe vernal keratoconjunctivitis with severe keratitis [CONFIDENTIAL internal manufacturer's report], Evry (FR), Santen SAS, 2016 Aug 22.
  29. A. Leonardi, G. Van Setten, M. Amrane, D. Ismail, J. S. Garrigue, F. C. Figueiredo and C. Baudouin, Efficacy and safety of 0.1% cyclosporine A cationic emulsion in the treatment of severe dry eye disease: a multicenter randomized trial, Eur. J. Ophthalmol., 2016, 26(4), 287–296 CrossRef PubMed .
  30. A. Mandal, D. Pal, V. Agrahari, H. M. Trinh, M. Joseph and A. K. Mitra, Ocular delivery of proteins and peptides, challenges and novel formulation approaches, Adv. Drug Delivery Rev., 2018, 126, 67–95 CrossRef CAS PubMed .
  31. F. G. Holz, R. Tadayoni, S. Beatty, A. Berger, M. G. Cereda, R. Cortez, C. B. Hoyng, P. Hykin, G. Staurenghi, S. Heldner, T. Bogumil, T. Heah and S. Sivaprasad, Multi-country real-life experience of anti-vascular endothelial growth factor therapy for wet age-related macular degeneration, Br. J. Ophthalmol., 2015, 99(2), 220–226 CrossRef PubMed .
  32. R. Suri, S. Beg and K. Kohli, Target strategies for drug delivery bypassing ocular barriers, J. Drug Delivery Sci. Technol., 2019, 55, 101389 CrossRef .
  33. Y. Wang and C. Wang, Novel eye drop delivery systems: advance on formulation design strategies targeting anterior and posterior segments of the eye, Pharmaceutics, 2022, 14(6), 1150 CrossRef CAS PubMed .
  34. R. Bisht, A. Mandal, J. K. Jaiswal and I. D. Rupenthal, Nanocarrier mediated retinal drug delivery: overcoming ocular barriers to treat posterior eye diseases, Wiley Interdiscip. Rev.: Nanomed. Nanobiotechnol., 2018, 10(2), e1473 Search PubMed .
  35. C. H. Tsai, P. Y. Wang, I. C. Lin, H. Huang, G. S. Liu and C. L. Tseng, Ocular drug delivery: role of degradable polymeric nanocarriers for ophthalmic application, Int. J. Mol. Sci., 2018, 19(9), 2830 CrossRef PubMed .
  36. R. Varela-Fernández, V. Díaz-Tomé, A. Luaces-Rodríguez, A. Conde-Penedo, X. García-Otero, A. Luzardo-Álvarez, A. Fernández-Ferreiro and F. J. Otero-Espinar, Drug Delivery to the Posterior Segment of the Eye: Biopharmaceutic and Pharmacokinetic Considerations, Pharmaceutics, 2020, 12(3), 269 CrossRef PubMed .
  37. M. Shimazawa, Y. Inoue, T. Masuda, R. Onodera, K. Tahara, Y. Shimizu, Y. Mibe, K. Tsuruma, H. Takeuchi and H. Hara, Topical Diclofenac-Loaded Liposomes Ameliorate Laser-Induced Choroidal Neovascularization in Mice and Non-Human Primates, Curr. Neurovasc. Res., 2017, 14(1), 46–52 CrossRef CAS PubMed .
  38. T. Cheng, J. Li, Y. Cheng, X. Zhang and Y. Qu, Triamcinolone acetonide-chitosan coated liposomes efficiently treated retinal edema as eye drops, Exp. Eye Res., 2019, 188, 107805 CrossRef CAS PubMed .
  39. J. Li, T. Cheng, Q. Tian, Y. Cheng, L. Zhao, X. Zhang and Y. Qu, A more efficient ocular delivery system of triamcinolone acetonide as eye drop to the posterior segment of the eye, Drug Delivery, 2019, 26(1), 188–198 CrossRef CAS PubMed .
  40. J. C. Altamirano-Vallejo, J. Navarro-Partida, A. Gonzalez-De la Rosa, J. H. Hsiao, J. S. Olguín-Gutierrez, A. C. Gonzalez-Villegas, B. C. Keller, L. Bouzo-Lopez and A. Santos, Characterization and Pharmacokinetics of Triamcinolone Acetonide-Loaded Liposomes Topical Formulations for Vitreoretinal Drug Delivery, J. Ocul. Pharmacol. Ther., 2018, 34(5), 416–425 CrossRef CAS PubMed .
  41. J. Navarro-Partida, J. C. Altamirano-Vallejo, A. Gonzalez-De la Rosa, J. Armendariz-Borunda, C. R. Castro-Castaneda and A. Santos, Safety and Tolerability of Topical Ophthalmic Triamcinolone Acetonide-Loaded Liposomes Formulation and Evaluation of Its Biologic Activity in Patients with Diabetic Macular Edema, Pharmaceutics, 2021, 13(3), 322 CrossRef CAS PubMed .
  42. M. Khalil, U. Hashmi, R. Riaz and S. Rukh Abbas, Chitosan coated liposomes (CCL) containing triamcinolone acetonide for sustained delivery: A potential topical treatment for posterior segment diseases, Int. J. Biol. Macromol., 2020, 143, 483–491 CrossRef CAS PubMed .
  43. Y. Gu, C. Xu, Y. Wang, X. Zhou, L. Fang and F. Cao, Multifunctional Nanocomposites Based on Liposomes and Layered Double Hydroxides Conjugated with Glycylsarcosine for Efficient Topical Drug Delivery to the Posterior Segment of the Eye, Mol. Pharm., 2019, 16(7), 2845–2857 CrossRef CAS PubMed .
  44. C. B. M. Platania, V. Fisichella, A. Fidilio, F. Geraci, F. Lazzara, G. M. Leggio, S. Salomone, F. Drago, R. Pignatello, F. Caraci and C. Bucolo, Topical Ocular Delivery of TGF-β1 to the Back of the Eye: Implications in Age-Related Neurodegenerative Diseases, Int. J. Mol. Sci., 2017, 18(10), 2076 CrossRef PubMed .
  45. C. B. M. Platania, M. Dei Cas, S. Cianciolo, A. Fidilio, F. Lazzara, R. Paroni, R. Pignatello, E. Strettoi, R. Ghidoni, F. Drago and C. Bucolo, Novel ophthalmic formulation of myriocin: implications in retinitis pigmentosa, Drug Delivery, 2019, 26(1), 237–243 CrossRef CAS PubMed .
  46. M. Yadav, N. Schiavone, A. I. G. Guzman-Aranguez, F. Giansanti, L. Papucci, M. J. PerezdeLara and I. P. Kaur, Atorvastatin-loaded solid lipid nanoparticles as eye drops: proposed treatment option for age-related macular degeneration (AMD), Drug Delivery Transl. Res., 2020, 10, 919–944 CrossRef CAS PubMed .
  47. A. Tatke, N. Dudhipala, K. Y. Janga, S. P. Balguri, B. Avula, M. M. Jablonski and S. Majumdar, In Situ Gel of Triamcinolone Acetonide-Loaded Solid Lipid Nanoparticles for Improved Topical Ocular Delivery: Tear Kinetics and Ocular Disposition Studies, Nanomaterials, 2018, 9(1), 33 CrossRef PubMed .
  48. A. Romeo, A. Bonaccorso, C. Carbone, G. Lupo, C. Daniela Anfuso, G. Giurdanella, C. Caggia, C. Randazzo, N. Russo, G. L. Romano, C. Bucolo, M. Rizzo, G. Tosi, J. Thomas Duskey, B. Ruozi, R. Pignatello and T. Musumeci, Melatonin loaded hybrid nanomedicine: DoE approach, optimization and in vitro study on diabetic retinopathy model, Int. J. Pharm., 2022, 627, 122195 CrossRef CAS PubMed .
  49. M. Ran, Y. Deng, J. Yan, A. Zhang, Y. Wei, X. Li, H. He, J. Gou, T. Yin, X. Tang, J. Kong, H. Zhang, H. Zhang and Y. Zhang, Neovascularization-directed bionic eye drops for noninvasive renovation of age-related macular degeneration, Chem. Eng. J., 2022, 450(3), 138291 CrossRef CAS .
  50. S. E. Radwan, A. El-Kamel, E. I. Zaki, S. Burgalassi, E. Zucchetti and R. M. El-Moslemany, Hyaluronic-Coated Albumin Nanoparticles for the Non-Invasive Delivery of Apatinib in Diabetic Retinopathy, Int. J. Nanomed., 2021, 16, 4481–4494 CrossRef PubMed .
  51. K. Jiang, Y. Hu, X. Gao, C. Zhan, Y. Zhang, S. Yao, C. Xie, G. Wei and W. Lu, Octopus-like Flexible Vector for Noninvasive Intraocular Delivery of Short Interfering Nucleic Acids, Nano Lett., 2019, 19(9), 6410–6417 CrossRef CAS PubMed .
  52. A. Badia, A. Duarri, A. Salas, J. Rosell, J. Ramis, M. F. Gusta, E. Casals, M. A. Zapata, V. Puntes and J. García-Arumí, Repeated Topical Administration of 3 nm Cerium Oxide Nanoparticles Reverts Disease Atrophic Phenotype and Arrests Neovascular Degeneration in AMD Mouse Models, ACS Nano, 2023, 17(2), 910–926 CrossRef CAS PubMed .
  53. P. S. Apaolaza, M. Busch, E. Asin-Prieto, K. Peynshaert, R. Rathod, K. Remaut, N. Dünker and A. Göpferich, Hyaluronic acid coating of gold nanoparticles for intraocular drug delivery: Evaluation of the surface properties and effect on their distribution, Exp. Eye Res., 2020, 198, 108151 CrossRef CAS PubMed .
  54. M. Bassetto, D. Ajoy, F. Poulhes, C. Obringer, A. Walter, N. Messadeq, A. Sadeghi, J. Puranen, M. Ruponen, M. Kettunen, E. Toropainen, A. Urtti, H. Dollfus, O. Zelphati and V. Marion, Magnetically Assisted Drug Delivery of Topical Eye Drops Maintains Retinal Function In Vivo in Mice, Pharmaceutics, 2021, 13(10), 1650 CrossRef CAS PubMed .
  55. X. Zhao, I. Seah, K. Xue, W. Wong, Q. S. W. Tan, X. Ma, Q. Lin, J. Y. C. Lim, Z. Liu, B. H. Parikh, K. N. Mehta, J. W. Lai, B. Yang, K. C. Tran, V. A. Barathi, K. H. Cheong, W. Hunziker, X. Su and X. J. Loh, Antiangiogenic Nanomicelles for the Topical Delivery of Aflibercept to Treat Retinal Neovascular Disease, Adv. Mater., 2022, 34(25), e2108360 CrossRef PubMed .
  56. K. Li, R. Li, P. Zou, L. Li, H. Wang, D. Kong, G. Zheng and L. L. Li, Glycopeptide-nanotransforrs eyedrops with enhanced permeability and retention for preventing fundus neovascularization, Biomaterials, 2022, 281, 121361 CrossRef CAS PubMed .
  57. J. Shen, H. Gao, L. Chen, Y. Jiang, S. Li, Y. Chao, N. Liu, Y. Wang, T. Wei, Y. Liu, J. Li, M. Chen, J. Zhu, J. Liang, X. Zhou, X. Zhang, P. Gu, Q. Chen and Z. Liu, Eyedrop-based macromolecular ophthalmic drug delivery for ocular fundus disease treatment, Sci. Adv., 2023, 9(4), eabq3104 CrossRef CAS PubMed .
  58. Y. Ge, A. Zhang, R. Sun, J. Xu, T. Yin, H. He, J. Gou, J. Kong, Y. Zhang and X. Tang, Penetratin-modified lutein nanoemulsion in situ gel for the treatment of age-related macular degeneration, Expert Opin. Drug Delivery, 2020, 17(4), 603–619 CrossRef CAS PubMed .
  59. S. Delgado-Tirado, D. Amarnani, G. Zhao, E. J. Rossin, D. Eliott, J. B. Miller, W. A. Greene, L. Ramos, S. Arevalo-Alquichire, D. Leyton-Cifuentes, L. Gonzalez-Buendia, D. Isaacs-Bernal, H. A. B. Whitmore, N. Chmielewska, B. V. Duffy, E. Kim, H. C. Wang, J. M. Ruiz-Moreno, L. A. Kim and J. F. Arboleda-Velasquez, Topical delivery of a small molecule RUNX1 transcription factor inhibitor for the treatment of proliferative vitreoretinopathy, Sci. Rep., 2020, 10(1), 20554 CrossRef CAS PubMed .
  60. S. Delgado-Tirado, L. Gonzalez-Buendia, M. An, D. Amarnani, D. Isaacs-Bernal, H. Whitmore, S. Arevalo-Alquichire, D. Leyton-Cifuentes, J. M. Ruiz-Moreno, J. F. Arboleda-Velasquez and L. A. Kim, Topical Nanoemulsion of a Runt-related Transcription Factor 1 Inhibitor for the Treatment of Pathologic Ocular Angiogenesis, Ophthalmol. Sci., 2022, 2(3), 100163 CrossRef PubMed .
  61. X. Yang, L. Wang, L. Li, M. Han, S. Tang, T. Wang, J. Han, X. He, X. He, A. Wang and K. Sun, A novel dendrimer-based complex co-modified with cyclic RGD hexapeptide and penetratin for noninvasive targeting and penetration of the ocular posterior segment, Drug Delivery, 2019, 26(1), 989–1001 CrossRef CAS PubMed .
  62. L. Tai, C. Liu, K. Jiang, X. Chen, L. Feng, W. Pan, G. Wei and W. Lu, A novel penetratin-modified complex for noninvasive intraocular delivery of antisense oligonucleotides, Int. J. Pharm., 2017, 529(1–2), 347–356 CrossRef CAS PubMed .
  63. S. Lai, Y. Wei, Q. Wu, K. Zhou, T. Liu, Y. Zhang, N. Jiang, W. Xiao, J. Chen, Q. Liu and Y. Yu, Liposomes for effective drug delivery to the ocular posterior chamber, J. Nanobiotechnol., 2019, 17(1), 64 CrossRef PubMed .
  64. M. Feghhi, B. Sharif Makhmalzadeh, F. Farrahi, M. Akmali and N. Hasanvand, Anti-microbial Effect and in Vivo Ocular Delivery of Ciprofloxacin-loaded Liposome through Rabbit's Eye, Curr. Eye Res., 2020, 45(10), 1245–1251 CrossRef CAS PubMed .
  65. A. Meza-Rios, J. Navarro-Partida, J. Armendariz-Borunda and A. Santos, Therapies Based on Nanoparticles for Eye Drug Delivery, Ophthalmol. Ther., 2020, 9(3), 1–14 CrossRef PubMed .
  66. E. Sánchez-López, M. Espina, S. Doktorovova, E. B. Souto and M. L. García, Lipid nanoparticles (SLN, NLC): Overcoming the anatomical and physiological barriers of the eye - Part II - Ocular drug-loaded lipid nanoparticles, Eur. J. Pharm. Biopharm., 2017, 110, 58–69 CrossRef PubMed .
  67. A. Mukherjee, A. K. Waters, P. Kalyan, A. S. Achrol, S. Kesari and V. M. Yenugonda, Lipid-polymer hybrid nanoparticles as a next-generation drug delivery platform: state of the art, emerging technologies, and perspectives, Int. J. Nanomed., 2019, 14, 1937–1952 CrossRef CAS PubMed .
  68. H. M. Kim and S. J. Woo, Ocular Drug Delivery to the Retina: Current Innovations and Future Perspectives, Pharmaceutics, 2021, 13(1), 108 CrossRef CAS PubMed .
  69. F. Masse, M. Ouellette, G. Lamoureux and E. Boisselier, Gold nanoparticles in ophthalmology, Med. Res. Rev., 2019, 39(1), 302–327 CrossRef PubMed .
  70. Y. Wang and C. Wang, Novel Eye Drop Delivery Systems: Advance on Formulation Design Strategies Targeting Anterior and Posterior Segments of the Eye, Pharmaceutics, 2022, 14(6), 1150 CrossRef CAS PubMed .
  71. G. Di Prima, S. Saladino, F. Bongiovì, G. Adamo, G. Ghersi, G. Pitarresi and G. Giammona, Novel inulin-based mucoadhesive micelles loaded with corticosteroids as potential transcorneal permeation enhancers, Eur. J. Pharm. Biopharm., 2017, 117, 385–399 CrossRef CAS PubMed .
  72. F. Zhang, H. Chen, J. Lan, K. Song and X. Wu, Preparation and in vitro/in vivo evaluations of novel ocular micelle formulations of hesperetin with glycyrrhizin as a nanocarrier, Exp. Eye Res., 2021, 202, 108313 CrossRef CAS PubMed .
  73. A. Mandal, R. Bisht, I. D. Rupenthal and A. K. Mitra, Polymeric micelles for ocular drug delivery: From structural frameworks to recent preclinical studies, J. Controlled Release, 2017, 248, 96–116 CrossRef CAS PubMed .
  74. A. R. Fernandes, E. Sanchez-Lopez, T. D. Santos, M. L. Garcia, A. M. Silva and E. B. Souto, Development and Characterization of Nanoemulsions for Ophthalmic Applications: Role of Cationic Surfactants, Materials, 2021, 14(24), 7541 CrossRef CAS PubMed .
  75. J. Rodríguez Villanueva, M. G. Navarro and L. Rodríguez Villanueva, Dendrimers as a promising tool in ocular therapeutics: Latest advances and perspectives, Int. J. Pharm., 2016, 511(1), 359–366 CrossRef PubMed .
  76. K. Nagpal, P. Kumar, A. Mohan and S. Thakur, Dendrimers for Therapeutic Delivery: Compositions, Characterizations, and Current Status, Crit. Rev. Ther. Drug Carrier Syst., 2019, 36(4), 277–304 CrossRef PubMed .
  77. A. Thareja, H. Hughes, C. Alvarez-Lorenzo, J. J. Hakkarainen and Z. Ahmed, Penetration Enhancers for Topical Drug Delivery to the Ocular Posterior Segment-A Systematic Review, Pharmaceutics, 2021, 13(2), 276 CrossRef CAS PubMed .
  78. J. G. Souza, K. Dias, S. A. Silva, L. C. de Rezende, E. M. Rocha, F. S. Emery and R. F. Lopez, Transcorneal iontophoresis of dendrimers: PAMAM corneal penetration and dexamethasone delivery, J. Controlled Release, 2015, 200, 115–124 CrossRef CAS PubMed .
  79. S. Kumar Dubey, R. Pradhan, S. Hejmady, G. Singhvi, H. Choudhury, B. Gorain and P. Kesharwani, Emerging innovations in nano-enabled therapy against age-related macular degeneration: A paradigm shift, Int. J. Pharm., 2021, 600, 120499 CrossRef CAS PubMed .
  80. M. Tang, X. Ji, H. Xu, L. Zhang, A. Jiang, B. Song, Y. Su and Y. He, Photostable and Biocompatible Fluorescent Silicon Nanoparticles-Based Theranostic Probes for Simultaneous Imaging and Treatment of Ocular Neovascularization, Anal. Chem., 2018, 90(13), 8188–8195 CrossRef CAS PubMed .
  81. L. Li, Z. Zeng, Z. Chen, R. Gao, L. Pan, J. Deng, X. Ye, J. Zhang, S. Zhang, C. Mei, J. Yu, Y. Feng, Q. Wang, A. Y. Yu, M. Yang and J. Huang, Microenvironment-Triggered Degradable Hydrogel for Imaging Diagnosis and Combined Treatment of Intraocular Choroidal Melanoma, ACS Nano, 2020, 14(11), 15403–15416 CrossRef CAS PubMed .
  82. G. Stati, F. Rossi, T. Trakoolwilaiwan, L. D. Tung, S. Mourdikoudis, N. T. K. Thanh and R. Di Pietro, Development and Characterization of Curcumin-Silver Nanoparticles as a Promising Formulation to Test on Human Pterygium-Derived Keratinocytes, Molecules, 2022, 27(1), 282 CrossRef CAS PubMed .
  83. A. J. Huang, S. C. Tseng and K. R. Kenyon, Paracellular permeability of corneal and conjunctival epithelia, Invest. Ophthalmol. Visual Sci., 1989, 30(4), 684–689 CAS .
  84. E. Dosmar, J. Walsh, M. Doyel, K. Bussett, A. Oladipupo, S. Amer and K. Goebel, Targeting Ocular Drug Delivery: An Examination of Local Anatomy and Current Approaches, Bioengineering, 2022, 9(1), 41 CrossRef CAS PubMed .
  85. S. Pescina, C. Ostacolo, I. M. Gomez-Monterrey, M. Sala, A. Bertamino, F. Sonvico, C. Padula, P. Santi, A. Bianchera and S. Nicoli, Cell penetrating peptides in ocular drug delivery: State of the art, J. Controlled Release, 2018, 284, 84–102 CrossRef CAS PubMed .

This journal is © The Royal Society of Chemistry 2023