Next Article in Journal
Nanotechnology-Enhanced Orthopaedic Surgery
Previous Article in Journal
How Reproducible Is Feraheme® (Ferumoxytol Injection)? Comparison of Size, Zeta Potential, and Complement Activation of Different Batches over 15 Years
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Delivery Systems for Infectious Eye Diseases: Advancements and Prospects

by
Binapani Mahaling
1,*,
Namrata Baruah
2 and
Aumreetam Dinabandhu
3
1
Medical College of Wisconsin, Froedtert Eye Institute, 925 N 87th St, Milwaukee, WI 53226, USA
2
Emory National Primate Research Center, Emory University, 954 Gatewood Rd NE, Atlanta, GA 30329, USA
3
Wilmer Eye Institute, The Johns Hopkins University School of Medicine, 1800 Orleans St, Baltimore, MD 21287, USA
*
Author to whom correspondence should be addressed.
J. Nanotheranostics 2024, 5(4), 133-166; https://doi.org/10.3390/jnt5040010
Submission received: 30 August 2024 / Revised: 27 September 2024 / Accepted: 2 October 2024 / Published: 6 October 2024

Abstract

:
Infectious ocular diseases like keratitis, conjunctivitis, and endophthalmitis pose significant clinical challenges due to the complexities of delivering drugs to the eye. Recent advancements in drug delivery systems offer promising improvements for treating these conditions. Key strategies include targeted delivery through physicochemical modifications, magnetic nanoparticles, and ligand-receptor interactions. This review explores the safety and biocompatibility of ocular drug delivery systems through in vivo ocular toxicity studies, in vitro cytotoxicity assays, hemocompatibility studies, ocular tolerance tests, and genotoxicity assays. It also examines combination therapies and stimuli-responsive delivery systems for their potential to enhance therapeutic efficacy. Furthermore, we discuss tailored and optimized drug delivery approaches for infectious ocular diseases, outlining current challenges and future directions for developing effective ocular drug delivery systems.

1. Introduction

Infectious eye diseases caused by microorganisms such as bacteria, viruses, fungi, and parasites, affect different structures of the eye. These infections, if left untreated, can result in severe inflammation, tissue damage, and vision impairment [1,2]. Common examples include conjunctivitis, keratitis, and endophthalmitis [1]. Nonetheless, they remain a substantial concern, particularly in regions with limited access to prompt and effective treatment options. Conventional treatments for infectious eye diseases, typically involving antimicrobial drugs administered topically or systemically, are often limited by low ocular bioavailability, rapid drug clearance, and the development of antimicrobial resistance [1,3]. Drug delivery systems (DDSs) have emerged as a promising solution to address these limitations by enhancing drug stability, bioavailability, and therapeutic efficacy [3,4,5,6,7]. Various DDS technologies, including nanoparticles, liposomes, hydrogels, and in-situ gelling systems, have shown potential in overcoming ocular barriers and optimizing drug delivery to infected tissues [8,9,10]. Targeted DDSs are designed to deliver therapeutic agents directly to specific sites within the eye, further enhancing treatment efficacy [11,12,13,14]. Combination therapies, such as antibiotics with anti-inflammatory or antifungal agents, offer synergistic effects against infections and resistance [8,15,16,17,18]. Moreover, stimuli-responsive DDSs, which react to physiological changes, allow for customizable, controlled drug release, representing a new frontier in ocular disease treatment [19,20,21,22,23]. Multiple parameters can be used to assess toxicity of these DDSs both in vivo and in vitro. Additionally, market products such as DuraSite® and Vigamox® exemplify the clinical translation of these advancements, providing more effective treatments for infectious ocular diseases while minimizing toxicity and enhancing patient compliance [24,25]. In this review, we provide a comprehensive overview of infectious eye diseases, advancements in DDSs including targeted and stimuli-responsive DDSs, combination therapies, and their safety and efficacy assessments.

2. Infectious Eye Diseases

Infectious eye diseases refer to a range of conditions that are caused by microorganisms such as bacteria, viruses, fungi, or parasites, and affect the various structures of the eye [1]. These diseases can lead to inflammation, tissue damage, and visual impairment if not promptly diagnosed and treated [2]. Common examples of infectious ocular diseases include conjunctivitis (pink eye), keratitis (corneal infection), endophthalmitis (infection inside the eye), and ocular manifestations of systemic infectious diseases like herpes simplex virus and toxoplasmosis [1]. From an epidemiological standpoint, infectious diseases rank significantly lower than the leading global causes of avoidable moderate-to-severe distance visual impairment (MSVI) and avoidable blindness. According to a comprehensive meta-analysis, in 2020, these conditions were estimated to affect 553 million and 43 million individuals, respectively [26].

2.1. Infectious Keratitis

Infectious keratitis is the inflammation of the cornea, which is the transparent, dome-shaped outer layer of the eye that covers the iris and the pupil, by the microorganisms. It is a medical emergency as it progresses very fast. Keratitis is the most common cause of corneal blindness and its prevalence in the USA is 30,000 cases per year. Infectious keratitis can be further classified into bacterial keratitis, viral keratitis, fungal keratitis, or parasitic keratitis. The symptoms include eye pain, redness, blurred vision, tearing, photophobia, foreign body sensations, discharge, and sometimes decreased visual acuity [1,27].
Bacterial keratitis is the most common cause of infectious keratitis. This can result from the invasion of the cornea by various bacterial species, with common culprits including Staphylococcus aureus, Streptococcus pneumoniae, Pseudomonas aeruginosa, and Haemophilus influenzae. Bacterial keratitis occurs when bacteria gain access to the cornea through an injury or a compromised ocular surface, such as in cases of contact lens wear, corneal trauma, or by a blocked nasolacrimal duct. The accumulation of bacterial toxins and cellular debris induces an inflammatory response by the host, involving the corneal epithelium, stromal keratocytes, resident immune cells, and infiltrating leukocytes, predominantly polymorphonuclear cells (PMNs). This local inflammasome activation is a double-edged sword: while it can effectively eliminate bacteria, the indiscriminate release of reactive oxygen species (ROS), lysosomal enzymes, and autolytic proteins can cause collateral damage to the surrounding stroma. This damage may result in permanent structural changes that impair vision (Figure 1A). The primary approach to address this is the administration of topical antibiotic eye drops or ointments [28,29]. In severe cases, oral antibiotics or even hospitalization may be required as treatment modalities [27].
Fungal keratitis occurs when fungi gain access to the cornea through an injury, contact lens use, or exposure to fungal-contaminated environments. It is most commonly caused by filamentous fungi, such as Fusarium, Aspergillus, and Candida species. The incidence of fungal keratitis is only 6–20% and is majorly prevalent in developing countries, tropical and sub-tropical regions [30,31]. During fungal infections, neutrophils make up 95% of the cellular infiltrate. Infected corneas exhibit progressive erosion and necrosis of corneal tissue, a thinning of the corneal epithelium, and a disordered corneal stroma. Corneal ulcers caused by filamentous fungi usually present with dry-looking infiltrates with feathery margins. These ulcers can also feature satellite lesions, a hypopyon, and endo-exudates (Figure 1B) [29,32]. Hypopyon is a condition where severe inflammation leads to pus accumulation in the anterior chamber of the eye due to infection. Endo-exudates are retinal exudates in the form of lipid- or protein-rich fluid deposition in the retina during infection. Antifungal medications in the form of topical eye drops or ointments are typically prescribed for relief [31].
Viral keratitis is a type of corneal infection caused by viruses, and it is characterized by inflammation of the cornea, which can lead to significant visual impairment if not promptly diagnosed and managed. The most common virus associated with viral keratitis is the herpes simplex virus (HSV), specifically HSV-1 [1,33]. The global incidence of herpes keratitis is about 1.5 million, including 40,000 new cases of severe monocular visual impairment or blindness each year [34]. Based on the part of cornea that is affected, herpes keratitis can be classified as epithelial, stromal, endothelial, or mixed, and based on occurrence, it can be primary or secondary. The primary clinical manifestations can include corneal opacity, edema, corneal scarring, and neovascularization, which can result in irreversible vision impairment and blindness. The infection by HSV-1 starts with the interaction of virus capsid glycoproteins such as gB, gC, gD, gH, and gL with the cell receptors such as nectin-1 and -2, herpes virus entry mediator (HVEM), and 3-O sulfated heparan sulfate (3-OS HS) on corneal epithelial cells [13]. Epithelial cell swelling due to HSV infection can cause the breakdown of the outer corneal layers and subsequent cell death [35]. In an infected cornea, HSV initiates viral replication in the corneal epithelial cells, and binds to Toll-like receptors on the cell surface, stimulating the influx of inflammatory cells and the release of cytokines and chemokines that gradually infiltrate the stroma. These inflammatory cells, including neutrophils, dendritic cells (DCs), natural killer cells, and macrophages, reportedly aid in clearing HSV from the cornea during the initial infection (Figure 1C) [36]. Stromal keratitis is likely driven by CD4+ T cells as part of the inflammatory response to stromal infection, alongside the direct effects of the virus [37]. The major treatment strategies involve oral or topical antiviral and topical steroids [1].
Parasitic keratitis is a rare but serious condition characterized by corneal infection caused by parasitic organisms such as Acanthamoeba species and microsporidia. Risk factors for parasitic keratitis include contact lens use, ocular trauma, exposure to contaminated environments, and a compromised immune system. Parasitic keratitis causes intense eye pain, inflammation, and damage to the epithelium and stroma, which can lead to vision loss if not diagnosed and treated promptly. It starts when the Acanthamoeba adheres to the corneal epithelium through a mannose-binding protein. Acanthamoeba trophozoites causes epithelial barrier breakdown through mechanisms including direct cytolysis, phagocytosis, and the induction of apoptosis. Following corneal epithelial cell barrier break down, it gradually invades the stroma. Once the infection reaches the stroma, it causes extensive damage to the collagen matrix, triggering intense inflammation [38]. The innate immune cells, specifically neutrophils and macrophages, play a crucial role in resolving parasitic keratitis. Notably, both neutrophils and macrophages are capable of killing trophozoites. Eosinophils are also present in the cornea of rabbits affected by Acanthamoeba, though the function is not fully known (Figure 1D) [39,40]. The treatment for parasitic keratitis is challenging and may involve multiple approaches. Antiparasitic medications in the form of eye drops, ointments, or oral medications are commonly used [41]. However, the complications associated with keratitis are corneal damage, vision loss, corneal scarring, and, in some cases, the need for corneal transplantation. Successful treatment for keratitis requires multiple challenges to be addressed to design an effective drug delivery system (DDS). A basic requirement is to concede the drug dosage along with maintaining therapeutic concentrations for prolonged periods.

2.2. Infectious Conjunctivitis

Conjunctivitis, commonly known as pink eye, is an inflammation associated with the dilation of blood vessels of the conjunctiva, the thin, transparent tissue that covers the white part of the eye and lines the inner surface of the eyelids. Infectious conjunctivitis is caused by bacteria and viruses [1]. It is estimated that acute conjunctivitis affects 6 million people annually in the United States alone. The cost of treating bacterial conjunctivitis is estimated to be $377 million to $857 million per year [20]. Both bacterial and viral conjunctivitis are highly contagious and can spread through direct contact with infected individuals [1].
Bacterial conjunctivitis is caused by the colonization of bacteria, most commonly by Staphylococcus aureus, Streptococcus pneumoniae, Moraxella catarrhalis, or Haemophilus influenzae, on the ocular surface, disrupting the epithelial barrier. It is a common form of conjunctivitis characterized by redness, discharge, and discomfort in the affected eye. Inflammatory cytokines (e.g., IL-1, IL-6, TNF-α) and chemokines (e.g., IL-8) secreted by infected epithelial cells play significant roles in recruiting and activating immune cells in the conjunctiva during bacterial infection. In bacterial conjunctivitis, the infiltration of neutrophils and macrophages into the conjunctiva is a key feature of the body’s response to infection (Figure 2A) [42,43]. Bacterial conjunctivitis is accountable for increased morbidity and provides a more challenging clinical scenario for physicians [44]. Children are most affected by bacterial conjunctivitis [20]. Bacterial conjunctivitis is typically treated with antibiotic eye drops or ointments. The specific antibiotic prescribed may depend on the suspected or identified bacteria. With appropriate treatment, bacterial conjunctivitis tends to resolve without complications. However, in rare cases, complications such as corneal involvement or the spread of infection to other parts of the eye may occur.
Viral conjunctivitis is caused by viruses, and it represents up to 80% of all cases of acute conjunctivitis. It affects both adults and children. A major cause of viral conjunctivitis (65 to 90%) is adenovirus [1,45]. The transmission of adenovirus-associated conjunctivitis occurs through direct contact with contaminated fomites, water, or fecal-oral contamination [4]. Typical symptoms of viral conjunctivitis include redness and swelling of the conjunctiva, watery discharge, hyperemia, chemosis, ipsilateral lymphadenopathy, discomfort, and a gritty sensation in the eye [45]. Adenoviral conjunctivitis begins when the adenovirus interacts with conjunctival epithelial cells. Primary cellular receptors, including CAR, CD46, and sialic acid, interact with the adenoviral fiber-knob protein to facilitate viral attachment to the host cell, while the interaction between the adenoviral penton base and integrins mediates viral internalization [4]. Both innate immune response viral conjunctivitis types are mediated by natural killer cells, neutrophiles, dendritic cells and macrophages and adoptive immune response by CD8 T cells, and T-helper 1cells (Figure 2B) [4,42,46]. The goblet cells of the conjunctiva are also involved in HSV-1 and adenovirus mediated conjunctivitis [47]. There is no effective treatment currently available for viral conjunctivitis.

2.3. Endophthalmitis

Endophthalmitis is a severe inflammation and infection of the internal structures of the eye, including the aqueous humor, vitreous humor, and surrounding tissues. Endophthalmitis can be classified as either exogenous or endogenous, depending on the source of the infection. Exogenous endophthalmitis arises when infecting organisms enter the eye through direct inoculation, such as during intraocular surgery, penetrating trauma, or contiguous spread from nearby tissues. On the other hand, endogenous endophthalmitis occurs when infectious agents are disseminated hematogenously into the eye from a remote focus of infection. Exogenous endophthalmitis is more common than the endogenous endophthalmitis. Endophthalmitis is mainly caused by gram positive bacteria like Staphylococcus aureus and Streptococcus spps. and gram-negative bacteria such as Klebsiella spp. and E. coli [8,48]. The symptoms of endophthalmitis, whether ocular or systemic, often lack specificity. Therefore, early diagnosis depends on the alertness of ophthalmologists and other physicians. It is considered a medical emergency that requires prompt diagnosis and immediate treatment to prevent vision loss and potential complications. The identification of the pathogen typically involves microbiological or molecular techniques, which entail staining and culturing specimens from the eye or bodily fluids such as blood, urine, or cerebrospinal fluid. Polymerase chain reaction (PCR) also serves as a valuable tool in confirming the diagnosis [49]. Both bacterial and fungal endophthalmitis are associated with infiltration of neutrophils and macrophages into the retina and vitreous humor [50]. The infiltration of immune cells and activated microglia are associated with the cytokine and chemokines secretion (Figure 2C,D) [51,52]. Intravitreal injections of antimicrobial agents are the principal method of treatment, along with systemic broad-spectrum antibiotics [48,49,53].

3. Optimized Drug Delivery Systems for Infectious Ocular Diseases

Infectious ocular diseases are emerging medical problems in recent years, with conjunctivitis, keratitis and endophthalmitis being the most important eye infections, which are generally treated with topical or systemic administration of conventional anti-microbial drugs [1]. However, conventional antimicrobial drugs have certain limitations such as poor ocular bioavailability, low ocular residence time, low ocular penetration, high rate of antibiotic resistance, and some allergic reactions [3]. The DDSs enable efficient delivery of antimicrobial agents to the ocular tissues affected by infectious agents by protecting drugs from degradation, increasing their solubility, and facilitating their sustained release, thereby improving drug bioavailability and therapeutic outcomes [3,5,6,8,54]. Different DDSs, such as nanoparticles, nanospheres, liposomes, micelles, niosomes, nanosponges, nanosuspensions, nanoemulsions, cubosomes, hydrogels, in-situ gelling systems, contact lenses, microneedles, and ocular implants, have been explored to optimize the delivery of antimicrobials/drugs for infectious ocular diseases [8,,9,10]. Hereby, we discuss the inherent characteristics of above DDSs in the context of infectious ocular diseases:
Nanoparticles and nanospheres are solid colloidal particles that serve as carriers for drugs. Their small size (typically 1–1000 nm) allows them to penetrate ocular barriers more effectively. Nanoparticles can be engineered to release drugs in a controlled manner, enhancing the drug’s retention on the ocular surface. This is particularly useful for treating ocular infections or chronic diseases like diabetic retinopathy [8,55,56]. Furthermore, they can be modified to be pH or enzyme-sensitive, enhancing targeted drug delivery.
Liposomes are vesicular structures composed of lipid bilayers that can encapsulate both hydrophilic and lipophilic drugs. Due to their biocompatibility and ability to fuse with cell membranes, liposomes are ideal for delivering drugs to ocular tissues. They can enhance the permeability of drugs across the cornea, making them suitable for treating anterior segment diseases like keratitis and conjunctivitis. Their versatility in surface modification allows for prolonged retention on the corneal surface [56,57,58].
Micelles are self-assembling colloidal particles formed by amphiphilic molecules. They offer enhanced solubility for hydrophobic drugs, making them useful in ocular formulations. Micelles are particularly effective in delivering drugs to the posterior segment of the eye due to their small size and ability to avoid clearance mechanisms. They can be used for delivering anti-inflammatory or anti-microbial agents with improved bioavailability [56,59].
Niosomes are non-ionic surfactant-based vesicles similar to liposomes but with enhanced stability. They are biocompatible and can encapsulate both hydrophilic and lipophilic drugs, making them versatile in ocular drug delivery. Their ability to prolong drug release and improve the retention of drugs in the eye makes them suitable for conditions requiring sustained drug action, and for diseases like keratitis, conjunctivitis and endophthalmitis [60,61,62].
Nanosponges are porous particles that can encapsulate drugs within their sponge-like structures, allowing for controlled and sustained drug release. They can deliver both hydrophilic and hydrophobic drugs efficiently. They can directly act against microorganisms. In ocular applications, nanosponges are useful in treating diseases where long-term drug exposure is required, as they reduce dosing frequency and improve patient compliance [56,63].
Nanosuspensions are colloidal dispersions of drug nanoparticles stabilized by surfactants. They improve the solubility and bioavailability of poorly water-soluble drugs and have the capability to overcome ocular barriers. In ocular applications, nanosuspensions can enhance drug penetration through the cornea and provide prolonged drug release. They are particularly useful for delivering drugs to the posterior segment of the eye, such as in the treatment of infectious retinal diseases [19,64,65].
Nanoemulsions are thermodynamically stable mixtures of oil, water, and surfactants, with droplet sizes in the nanometer range. These systems improve the solubility and bioavailability of hydrophobic drugs. In the eye, nanoemulsions enhance drug penetration and provide sustained release, making them ideal for delivering anti-inflammatory and anti-microbial drugs. Their small droplet size improves drug absorption across ocular barriers [20,56,66,67].
Cubosomes are nanostructured particles with a cubic crystalline phase formed from amphiphilic lipids. They are stable, biocompatible, and capable of carrying both hydrophilic and lipophilic drugs. Cubosomes can enhance drug delivery across ocular barriers and provide a controlled release, making them particularly useful for both anterior and posterior infectious eye diseases [68,69,70].
Hydrogels are three-dimensional networks of hydrophilic polymers that can absorb water and swell, allowing them to release drugs in a controlled manner. In situ gelling systems are liquid formulations that transition to a gel state upon contact with the eye. These systems enhance drug retention on the ocular surface and reduce the need for frequent dosing, making them ideal for treating conditions like keratitis and conjunctivitis [15,71,72,73].
Drug-eluting contact lenses offer a novel method for sustained drug delivery to the eye. By incorporating drug-loaded nanoparticles or hydrogels into the lens material, these lenses provide continuous drug release over extended periods. They improve drug bioavailability and patient compliance by eliminating the need for frequent eye drops. Contact lenses can be used to treat various anterior segment diseases, such as corneal and conjunctival infections [16,56,74].
Microneedles are tiny needles that can penetrate the ocular surface or sclera without causing significant pain or damage. These devices offer a minimally invasive method for delivering drugs directly to the anterior and posterior segment of the eye, bypassing ocular barriers [75,76,77].
Ocular implants are small, solid devices placed inside the eye to deliver drugs over an extended period. These implants provide continuous, controlled drug release, reducing the need for repeated injections or topical applications. Implants are particularly beneficial for posterior chronic and infectious diseases [78,79].
Each of these above DDSs offer distinct advantages for ocular applications, including enhanced drug stability and bioavailability, prolonged retention time, ocular penetration, and controlled release. Their selection depends on the specific disease being treated, the required duration of therapy, and the drug’s physicochemical properties as explained in Table 1, Table 2 and Table 3.

3.1. Drug Delivery Systems for Infectious Keratitis

Infectious keratitis is a condition characterized by corneal inflammation resulting from an infection, posing a potential threat to vision and constituting an urgent eye problem. Managing infectious keratitis effectively depends on identifying the specific microorganism responsible, and treatment often involves the frequent use of antibacterial, antifungal, or antiviral eye drops. However, these eye drops may face challenges such as limited bioavailability, unwanted side effects, and reduced patient compliance due to barriers presented by the cornea [80]. The corneal barriers include tight junctions formed by the corneal epithelium, reflex blinking, nasolacrimal drainage and drainage of drug to other ocular layers by aqueous barrier [11,81]. Advancements in DDSs have been put forward to overcome corneal barriers leading to enhanced drug residence time, increased permeability of the drug, enhanced drug solubility, and protection of the drug from acids and enzymatic effects, overcoming drug resistance and reductions in toxicity as described in Table 1.
Table 1. Drug delivery systems for infectious keratitis.
Table 1. Drug delivery systems for infectious keratitis.
Drug Delivery SystemDetails of Drug Delivery SystemDiseaseMechanism of ActionApplications
Nanoparticles Gelatin-capped silver nanoparticlesBacterial keratitisAntibacterial and anti-angiogenic properties of the silver nanoparticlesThe gelatin-capped silver nanoparticles alleviate S. aureus-induced bacterial keratitis in rabbit eyes and bacterial infection-induced corneal neovascularization [82].
Silver nanoparticles prepared from fungusViral keratitisShow antiviral activity by blocking interaction between cells and virus Silver nanoparticles reduce viral infectivity in a size-dependent manner against herpes simplex virus and human parainfluenza virus; smaller sizes resulting in higher antiviral activity [83].
Protease responsive TLR-4 conjugated gelatin nanoparticlesFungal keratitis Enhance drug residence time by anchoring to the corneaUsed for management of keratitis by sustained and stimuli responsive release of antifungal drug and inhibition of inflammation [9].
Nigella sativa aqueous extract and chitosan nanoparticlesParasitic keratitisOvercome drug resistanceThe combination of Nigella sativa aqueous extract and chitosan nanoparticles shows synergistic antiparasitic activity in an animal model of Acanthamoeba keratitis [84].
Nanospheres Silver nanospheresKeratitis Kill S. aureus by predominance of specific particle density and high-atom-densityShows higher biocompatibility, anti-bacterial activity, anti-angiogenic capability, and repair of infected corneal tissues as compared to rod and triangular silver nanoparticles [10].
Liposomes Liposome-dual TLR 3/9 agonist nanoparticlesHSP-1 induced conjunctivitis, keratitis, uveitisImmunomodulation in subconjunctival tissues and corneal epithelium and suppression of herpesvirus replication and signs of infectionSignificant lessening of ocular signs of infection and significantly fewer episodes of viral shedding compared to feline herpesvirus -1 infected cats. This is achieved by induction of both type I and II interferon responses along with suppression of feline herpesvirus -1 replication [57].
Tobramycin liposomesBacterial keratitis Single topical administration of tobramycin liposomes reduced frequent dosing by sustained drug releaseSingle topical administration of tobramycin liposomes provides bactericidal effects in Pseudomonas keratitis rabbit model and may provide better patient compliance [85].
Liposomal amphotericin B
(Fungisome)
Fungal keratitisLiposomal formulation slows down the ocular drug clearance and acts as a drug reservoirTopical application of Fungisome was more effective in reduction of fungal keratitis with lower toxicity than naked amphotericin B, though not statistically significant [58].
Combinatorial delivery of siRNA (serine phosphatase and glycogen phosphoryl)-loaded liposomes and chlorhexidineParasitic keratitisProvide enhanced corneal penetration and chlorhexidine- siRNA act against Acanthamoeba.After 15 days of eight daily administrations, the liposomal complex combined with chlorhexidine treatment is able to reverse the lesions associated with keratitis [86].
MicellesChtosan-poly(lactide-co-glycolide)/poloxamer mixed micellesBacterial keratitisShow enhanced antibacterial activity by high cellular uptake, corneal retention, muco-adhesiveness, and antibacterial effectExhibiting superior therapeutic effects in P. aeruginosa and S. aureus-infected BK mouse models by reducing the corneal bacterial load and preventing corneal damage [87].
Acyclovir-loaded Soluplus micelles Viral keratitis Increases its solubility, corneal permeability and sclera penetrationIn vivo efficacy in viral keratitis needed to be evaluated [88].
Itraconazole-loaded polymeric micellesFungal keratitisEnhance corneal permeability of hydrophobic drug and mucoadhesivityShows in vitro anti-fungal activity against Candida albicans through inhibition of chitin synthesis [89,90].
NiosomesCiprofloxacin-loaded Solulan C24- and Bile Salts-Modified NiosomeBacterial keratitisEnhance drug permeability and prolong ocular residence timeSignificantly improves the Pseudomonas aeruginosa induced corneal ulcer, enhanced healing rate, inhibition of inflammatory cytokines secretion in vivo in rabbit [60].
Natamycin-loaded nanoparticle noisomeFungal keratitisEnhance corneal penetration, protection against acidic and enzymatic effects, reduction in toxicityExhibits superior anti-inflammatory and antifungal activity as compared to free natamycin in rabbit in both Candida and Aspergillus keratitis [91,92].
Isoniazid-nanoparticles-amphotericin BParasitic keratitisYet to be tested for the eye and hence, can be exploredShows antiamoebic effects against Acanthamoeba castellanii [93].
Nanosuspensions Eudragit-
Ganciclovir Nanosuspension
Viral keratitis Enhance corneal penetration, provides sustained release at pH 7.4In vitro and in vivo efficacy study needed to be performed.
Ganciclovir’s main mode of action against CMV involves the inhibition of viral DNA replication through the activity of ganciclovir-5’-triphosphate (ganciclovir-TP) [94,95].
Moxifloxacin–pamoate nanosuspensionsBacterial keratitis Provide a significant increase in ocular drug absorptionImproved prevention and treatment of ocular keratitis in rat model of ocular Staphylococcus aureus infection [96].
Voriconazole-loaded Glycol chitosan nanosuspensions with 4-carboxyphenylboronic acid pinacol ester as a ROS-responsive group (GC-EV-VOR)Fungal keratitis Exhibits high penetration through corneal barriers, good retention in the cornea and controllable drug release under low concentrations of ROSImproved therapeutic effects with depletion of ROS, ocular inflammation and antifungal activity. Voriconazole exhibits antifungal activity by inhibition of cytochrome P450–dependent 14α-lanosterol demethylation, which is a vital step in cell membrane ergosterol synthesis by fungi [64,97].
Praziquantel nanosuspensionsParasitic keratitisThese nanosuspensions greatly modify the energetic metabolism of T. cysticerci in vivoTreating with nanosuspensions in T. crassiceps cysticerci-infected mice led to elevated levels of glycolysis related organic acids and promoted a partial reversal of the tricarboxylic acid cycle, urea cycle, and ketone body production within the parasites, in contrast to the groups treated with the Praziquantel [98].
Nanoemulsions Acyclovir-loaded termoresponsive in situ gel nanoemulsionViral keratitis Enhance ocular permeation and sustained drug releaseThe DDSs shows good ocular tolerance bioavailability; however, the in vitro and in vivo efficacy need to be evaluated [20].
Ciprofloxacin-loaded nanoemulsion fabricated from sing oleic acid and Labrafa Lipophile WL 1349, Tween 80 and Poloxamer 188 Bacterial keratitisEnhance corneal penetration and sustained drug releaseThe nanoemulsion system demonstrated prolonged release and a 2.1-fold increase in permeation compared to the commercial ciprofloxacin formulation. Additionally, it exhibits robust stability at room temperature and can be frozen for up to one month. These findings suggest promising potential for treating bacterial keratitis [99].
Luliconazole-loaded nanoemulsion composed of Capryol 90, ethoxylated hydrogenated castor oil, Transcutol® P and waterFungal keratitisEnhance drug release and antifungal activity and ocular bioavailability The nanoemulsion formulation displayed outstanding physicochemical properties, excellent tolerability, enhanced antifungal activity, and improved bioavailability in ocular tissues [100].
Amphotericin-loaded nanoemulsion formulated from cholesterol and stearyl amineParasitic keratitisImprove drug stability, bioactivity and sustained deliveryA single dose of nanoemulsion formulation could provide stability and sustained release of drug in vitro. However, in vivo evaluation needs to be performed [101].
CubosomesGatifloxacin
Loaded cubosomes
Bacterial keratitisEnhance corneal penetration, bioactivity of the drug, and overcome bacterial resistanceGatifloxacin-loaded cubosomal dispersions enhanced corneal permeation and a fourfold reduction in the minimum inhibitory concentration against a clinically isolated methicillin-resistant strain in a rat model of bacterial keratitis [102].
Fluconazole-loaded cubosomeFungal keratitisEnhance corneal penetration and improved efficacyFluconazole-loaded cubosomes shows two-fold corneal penetration, and higher efficacy and safety as compared to aqueous fluconazole solution in rat model [68].
HydrogelSupramolecular hydrogel generated by ganciclovir and 2′-deoxyguanosine in the presence of potassium ions.Herpes simplex keratitis Enhance corneal retention because of high viscoelasticity, excellent spreadability, and effective deformation recovery propertiesGanciclovir and 2′-deoxyguanosine-based supra molecular hydrogel exhibit longer ocular retention higher therapeutic efficacy compared to the clinical ganciclovir gel [72].
Norfloxacin-loaded PLGA-laden hydroxypropyl methylcellulose hydrogelBacterial keratitisEnhance corneal permeation, sustained drug release, and antibacterial propertiesThe DDS enhances corneal permeation, ensuring sustained delivery and effectiveness against Pseudomonas aeruginosa-induced infection and inflammation in a rabbit corneal ulcer model [73].
Econazole-cyclodextrin embedded in deacylated gellan gum and k-carrageenan hydrogel or hyaluronic acid hydrogelFungal keratitisEnhance drug solubility and ocular retention The econazole hydrogel shows superior ocular retention, sustained release, non-irritation, and a better safety profile compared to the control drug [94].
Poly-Epsilon-Lysine (PEK) Functionalized HydrogelsParasitic keratitisOffer amoebicidal activityThe PEK-hydrogels shows equivalent anti-microbial properties as compared to Chlorohexidine against A. castellanii in in vitro and ex vivo studies [103].
In-situ gelling systemAcyclovir-loaded sodium alginate based in situ gelling systemViral keratitisIncrease the half-life of the drug, prolong corneal contact time, eradicate drug elimination and increase the bioavailabilityOphthalmic in situ gels offer the potential for sustained delivery, leading to extended ocular residence time, enhanced shelf life, and precise dosing compared to conventional dosage forms [104].
Oxytetracycline-loaded gelatin-polyacrylic acid nanoparticles In Situ Poloxamer N-407 gelBacterial keratitisProvides sustained delivery of drugThe in situ gelling system provides sustained delivery of drug, minimizing irritation and exhibiting excellent antibacterial properties. It holds promise as a potential delivery system for treating bacterial keratitis [105].
Voriconazole-loaded poloxamer-188, poloxamer-407 and carboxymethyl cellulose based in situ gelFungal keratitisIncrease residence time, and the bioavailability of voriconazole in the ocular mucosa This in situ gelling system offers sustained delivery, potent antifungal effects, and causes no eye irritation. It shows promise for development as a potential delivery system for treating fungal keratitis [106].
Super aggregated amphotericin B with a thermoreversible in situ gelParasitic keratitisEnhance biocompatibility, low toxicity, and high residence time on the ocular surfaceThis in situ gelling system could enhance the residence time while reducing toxicity and being active against Acanthamoeba keratitis, thus, holds promise for treating parasitic keratitis [107].
Contact lensesMethacrylic acid-based imprinted valacyclovir-loaded contact lensesViral keratitisProvide sustained release and enhance ocular penetration of drugThese contact lenses improve drug solubility, boost drug bioavailability through enhanced ocular penetration, and demonstrate excellent ocular compatibility [74].
Melimine-coated contact lensBacterial keratitisProvide antibacterial properties against Pseudomonas aeruginosa by inhibiting adhesion of bacteria.Contact lenses coated with melimine decreased the occurrence of microbial keratitis linked to Pseudomonas aeruginosa in a rabbit model of keratitis [108].
Hybrid hydrogel-based contact lens which comprises quaternized chitosan, silver nanoparticles, and graphene oxide with voriconazoleFungal keratitisProvide sustained delivery of drug and silver nanoparticles and further enhances the antifungal properties of the developed contact lens.The hybrid hydrogel-based contact lens offers continuous dry release, exhibits excellent biocompatibility, and effectively treats fungal keratitis in a mouse model [109].
Miltefosine coated PLGA contact lensAcanthamoeba KeratitisProvide sustained and local delivery of drugThe drug-eluting contact lens not only shows similar physicochemical and biocompatible properties as compared to the commercial contact lens, but also provides sustained and local delivery of drug at a therapeutic concentration for treatment of parasitic keratitis [110].
Micro-needlesCore-shell microneedles containing Ag@ZIF8 nanoparticles in the core and Rapamycin in the hydrophilic shell Bacterial keratitisThe core provides antimicrobial properties, and shell provides anti-inflammatory and antiangiogenic properties. A single application of the core–shell microneedles patch in a rat model of bacterial keratitis demonstrates effective antimicrobial action, along with superior anti-angiogenic and anti-inflammatory effects compared to daily topical eyedrops [77].
Amphotericin B-loaded polyvinyl alcohol and polyvinyl pyrrolidone microneedle patchFungal keratitisEnhances drug solubility, corneal penetration, and provides sustained drug deliveryThe application of amphotericin B-loaded microneedles patch significantly decreased the Candida albicans presence within the cornea and enhanced the epithelial and stromal differentiation of the corneal membrane in rabbit model [75].
polyhexamethylene biguanide (PHMB)-loaded PLGA microneedle patch Acanthamoeba keratitisProvide sustained deliveryA solitary application of a biodegradable microneedle can replace the repetitive use of eyedrops in treating Aacanthamoeba keratitis [76].
Ocular implantsBesifloxacin HCl-loaded nanofibrous PCL/PEG mucoadhesive ocular insertsBacterial keratitisEnhance bio-adhesion and sustained deliveryThe ocular inserts demonstrated effective reduction of bacterial keratitis in rabbit eyes in a single dose, outperforming multiple doses of the commercial drug [78].

3.2. Drug Delivery Systems for Infectious Conjunctivitis

Infectious conjunctivitis is associated with inflammation due to infection by bacteria or virus [1]. These are generally treated with antimicrobial eyedrops or ointments [45]. However, the major shortcomings associated with the current treatment strategies are- poor solubility of drugs, low bioavailability of drugs due to conjunctival barrier properties, and development of resistance for current antimicrobial agents [111,112]. The conjunctiva’s protective functions, including guarding the ocular surface, producing the tear film, and aiding in drug clearance into systemic circulation or transportation to deeper eye tissues, represent significant hurdles in conjunctival drug delivery [113]. The advancements in DDSs have been put forward to overcome conjunctival barriers by enhancing drug residence time, drug solubility, drug stability; increasing permeability of the drug, providing sustained drug delivery, and overcoming drug resistance and reductions in toxicity as explained in Table 2.
Table 2. Drug delivery systems for infectious conjunctivitis.
Table 2. Drug delivery systems for infectious conjunctivitis.
Drug Delivery SystemDetails of Drug Delivery SystemDiseaseMechanism of ActionApplications
Nanoparticles Biological adhesion reticulate structure (BNP/CA-PEG) ConjunctivitisEnhances ocular surface retention by mucoadhesive properties BNP/CA-PEG showed significantly higher adhesion properties, sustained drug release and better treatment efficacy in an ocular rat model of conjunctivitis [111].
NanospheresLevofloxacin-loaded chitosan-cyclodextrin nanospheresConjunctivitisMay increase ocular retention by promoting the interaction of positively charged nanospheres with the negatively charged ocular tissueIn vitro, antibacterial activity against Gram-positive and Gram-negative bacteria showed double the activity in levofloxacin-loaded chitosan-cyclodextrin nanospheres compared to the free drug [114].
LiposomesLiposome containing gaseous ozone or ozonated oilConjunctivitisOzone generates free oxygen radicals that promote the formation of hydrogen peroxide and lipo-peroxide, contributing to bacterial lysis and eventual deathShows antibacterial properties against broad spectrum bacteria like Pseudomonas aeruginosa, and is more evident against Staphylococcus and Streptococcus spp. [115,116].
MicellesErythromycin-loaded polymeric micellesBacterial conjunctivitis Prolongs the drug release and permeation profile. Even after ocular permeation, the formulation retains antibacterial properties, which is a promising strategy against ocular infection [112].
DendrimersAminoterminated-poly(amidoamine) (PAMAM) dendrimerBacterial conjunctivitisEffective against gram negative bacteria by disrupting bacterial cell membraneEffective against gram-negative bacteria but does not cause cell death to corneal cells, thus expanding its application towards conjunctivitis [117].
NiosomesLomefloxacin Hcl (LXN)-loaded niosomesKetaritis and bacterial conjunctivitisEnhances corneal penetration of LXNEffective against Staphylococcus aureus induced conjunctivitis in vivo without having any ocular toxicity in rabbit model [61].
NanosuspensionIon-paired moxifloxacin–pamoate nanosuspensionKeratitis and conjuctivitisIncreases intraocular antibiotic absorptionSingle dosing per day of moxifloxacin–pamoate nanosuspension is as effective as three doses per day of bare moxifloxacin [19].
NanoemulsionMoxifloxacin hydrochloride-loaded nanoemulsion-based in situ gelBacterial conjunctivitis Provides sustained delivery and reduced toxicityThe nanosuspension improves ocular bioavailability of drugs through enhanced penetration into the eye and sustained delivery [66].
CubosomesCubosomes formed by 60 mg of ciprofloxacin, 100 mg phytantriol, 25 mg Lutrol, and hydration media pH equal 5.8Conjunctivitis and corneal ulcer Improves eye permeation, prolonged the ocular retention time, and enhanced the antimicrobial activity Single administration of cubosomal DDSs can maintain drug levels above the minimum inhibitory concentration (MIC), contrasting with commercial drops that necessitate administration four times daily [69].
Hydrogel Poloxamer 407 and chitosan hydrogel containing combination of neomycin sulphate and betamethasone sodium phosphateConjunctivitis Enhances permeation and without any irritationIt can inhibit infection and inflammation simultaneously while providing sustained release of drugs [15].
In situ gelling systemlevofloxacin hemihydrate containing ion-sensitive gellan gum based in situ gelling systemBacterial conjunctivitisProvides prolonged drug release (24 h), enhanced stability and shows good ocular tolerabilityThe gellan gum-based in situ gelling system provides higher bioavailability of drug in conjunctiva as compared to the commercial product Levotop PF® with a good ocular tolerability, indicating potential use for treatment of bacterial conjunctivitis [118].
Contact lensContact lenses loaded with moxifloxacin HCl and hyaluronic acid Bacterial conjunctivitisProvides sustained drug release for 96 h, enhanced retention in eyeIt shows better efficacy against Staphylococcus aureus induced conjunctivitis as compared to eye drops [16].

3.3. Drug Delivery Systems for Endophthalmitis

Endophthalmitis is a devastating disorder characterized by ocular inflammation due to infection of the intraocular cavity that can lead to irreversible visual loss. Current treatment strategies are majorly based on systemic or intravitreal antimicrobial agents. Intravitreal injection of antimicrobial agents along with anti-inflammatory drugs is sometimes utilized in treatment protocols [49,53]. Repeated intravitreal injections of antimicrobial agents represent the prevailing treatment approach; however, they may be linked to discomfort, ocular pain, elevated intraocular pressure, intraocular bleeding, heightened risk of retinal detachment, and retinal toxicity, all of which can worsen complications and frequently result in blindness [8,56,119]. The advancements in DDSs have been put forward to overcome static and dynamic barriers of the eye such as corneal and conjunctival barriers, blood ocular barriers, blood retinal barriers, and barriers associated with conjunctival and choroidal blood flow, enhancing ocular drug adsorption, drug residence time, permeability of the drug, and drug solubility, while overcoming drug resistance and reducing toxicity as explained in the Table 3.
Table 3. Drug delivery systems for endophthalmitis.
Table 3. Drug delivery systems for endophthalmitis.
Drug Delivery SystemsDetails of Drug Delivery SystemsDiseaseMechanism of ActionApplications
Nanoparticles Chitosan coated poly-l-lactide nanoparticlesEndophthalmitis Overcome the blood ocular and retinal barrier by enhanced penetrationThe azithromycin- and triamcinolone acetonide-loaded dual drug DDSs showed antibacterial properties against both gram positive and gram negative bacteria [8].
Nanospheres Vancomycine-loaded lipid nano- capsules/spheresEndophthalmitis Show higher bioavailability of drug in the vitreous region by enhanced corneal penetrationAntimicrobial activity is comparable to intravitreal injection of vancomycin. Further, the optimized formula was found to be nonirritating and safe for ophthalmic administration [120].
LiposomesLiposomes-loaded Amphotericin-B or Fluconazole EndophthalmitisShow lower efficacy against Candida albicans induced fungal endophthalmitisThe liposomal antifungal drug shows lower toxicity and lower intravitreal clearance; however, it shows lower efficacy. Amphotericin B acts on disruption of fungal cell wall synthesis, and fluconazole inhibits the synthesis of ergosterol to increase cellular permeability [121,122,123,124].
MicellesMiconazole nitrate-loaded micelles of tri-block copolymers Pf 127 and Pf 68Fungal endophthalmitis Enhance corneal penetrationSustained drug delivery and achieved the therapeutic level of drug at 8 h of release [59].
DendrimersDendrimeric polyguanidilyated translocators-gatifloxacin complexEndophthalmitis Enhance gatifloxacin solubility, enhanced epithelial permeability and antimicrobial activity against Staphylococcus aureusShows promising effects against S. aureus induced endophthalmitis by enhancing drug solubility, permeability, antimicrobial activity, and in vivo delivery, potentially allowing a once-daily dose regimen [125].
NiosomesNiosome made up of Tween 60, cholesterol and dicetyl phosphateOcular infectionProvides sustained delivery of Gentamicin with no ocular irritationMay be useful for both gram positive and gram-negative bacteria and can be explored for ocular infections like endophthalmitis [62].
NanospongesRBC-PLGA nanospongesEndophthalmitis Effective against Enterococcus faecalisThe biomimetic nanosponges neutralize cytolysin, protect the retina, preserve vision, and may provide an adjunct detoxification therapy for bacterial infections [63].
nanosuspensionsAcyclovir containing polymeric nanosuspensionMay be useful for ocular infectionEnhance drug solubility, ocular bioavailability, and sustained drug releaseThe nanosuspensions exhibit promising potential due to their ability to enhance drug solubility, ocular bioavailability, sustained drug release, and antifungal properties. However, additional in vitro and in vivo assessments are required to evaluate their efficacy further [65].
NanoemulsionMoxifloxacin Mucoadhesive NanoemulsionEndophthalmitis Effective against both gram positive and negative bacteriaProvides sustained drug release for enhanced corneal penetration and equivalent antibacterial properties as compared to commercial Vigamox® eyedrops [67].
CubosomesIn situ gel containing Natamycin-loaded cubosomesEndophthalmitisEnhance drug solubility and ocular penetration with less ocular irritationMay be effective against fungal endopthalmitis, though in vivo evaluation in a disease model is needed [70].
In situ gelling systemCiprofloxacin containing micro emulsion-based in situ gelling systems EndophthalmitisEnhances the drug’s absorption, penetration, and retention, thereby boosting its bioavailability Given that the drug concentration in the vitreous is approximately 0.4 µg/mL, surpassing the therapeutic threshold required for gram-negative bacteria, it presents a potential avenue for investigation in the context of endophthalmitis [126].
Intraocular implantsMoxifloxacin releasing hyaluronic acidEndophthalmitisMaintain therapeutic concentration of drug for Pseudomonas aeruginosa and Staphylococcus aureus for more than 5 days after implantationCan be utilized as a potential drug delivery method for the prevention and treatment of bacterial infections like endophthalmitis after ophthalmic surgery [79].
Intra-ocular lensIntraocular lens containing antibiotic solutions of 0.3% and 0.5% gatifloxacin and 0.5% and 1.5% levofloxacin Prevention of post cataract endophthalmitisEffective against Enterococcus faecalisProvide sustained release of antibiotics and prevent bacterial proliferation [127].

4. Targeted Drug Delivery Systems for Infectious Ocular Diseases

Targeted drug delivery for infectious ocular diseases involves the development of advanced DDSs that can precisely deliver therapeutic agents to specific sites within the eye. This approach enhances the efficacy of treatments while minimizing systemic side effects. The various strategies of causing DDSs to target different specific ocular tissues include controlling the physicochemical properties of the nanoparticles, magnetic nanoparticles, and ligand receptor-based targeting [11,12,13,14].

4.1. Ocular Tissue Targeting by Varying the Physicochemical Properties of the Drug Delivery Systems

Due to the inherent barrier properties of eye tissues, a major challenge in treating eye diseases is delivering therapeutic agents to the desired tissue in effective quantities and durations. In our previous study, we aimed to understand how the physicochemical properties of nanoparticles influence their spatiotemporal biodistribution in the mouse eye. For this purpose, core-shell nanoparticles with varying properties such as hydrophilicity, surface charge, and mucoadhesivity were designed by altering either the core or the shell, and were administered as eye drops to mice. The results demonstrated that all nanoparticles, regardless of the core or shell type, followed the conjunctival-scleral pathway. However, their spatiotemporal distribution varied on the basis of their physicochemical properties. For instance: polycaprolactone (PCL)-Gelatin showed the highest biodistribution in the cornea, PCL-Pluronic F68 in the conjunctiva, choroid, and retina, PCL-Chitosan in the conjunctiva and iris, and PCL in the lens and sclera compared to other nanoparticles [11,12]. These nanoparticles can be explored to target infectious diseases in these specific tissues of the eye.

4.2. Ocular Tissue Targeting by Magnetic Nanoparticles (MNPs)

MNPs stand out from other nanocarriers due to their unique magnetic properties, which make them particularly suitable for targeted drug delivery. Drug molecules can be attached to the surface of MNPs and injected into the body. An external magnetic field can then be used to concentrate these drug-laden nanoparticles in a specific area, minimizing damage to surrounding tissue. MNPs specifically localize in the retinal pigment epithelium (RPE), where they remain for several days after being injected intravitreally or near the lens in the anterior eye. This localization specificity is independent of the MNPs’ particle size and surface properties [13]. However, functionalizing the MNP surface with vascular endothelial growth factor (VEGF), a bioactive molecule capable of transcytosis from the RPE to posterior layers, successfully targeted the MNPs to the choroid. In contrast, MNPs functionalized with a control polypeptide, poly-L-lysine, exhibited the same localization pattern as the unmodified MNP particles [14]. These magnetic nanoparticles can be a promising treatment modality for endophthalmitis.

4.3. Ocular Tissue Targeting by Ligand-Receptor Interactions

Ligand-receptor-based targeting is a promising strategy for the treatment of ocular infectious diseases, leveraging the specific interactions between ligands and receptors to enhance drug delivery efficacy. Various ligands, including antibodies, proteins, peptides, fatty acids, the carbohydrate mitis, and aptamers are used to target specific receptors expressed on diseased cells, or microorganisms improving drug delivery efficiency and therapeutic outcomes in infectious ocular diseases [128,129,130,131]. There are multiple ligands that are used to modify DDSs explored for corneal targeting. For example: (i) nanoemulsions have been modified with stearoyl L-carnitine to target carnitine transporter 2 and amino acid transporter B in human corneal epithelial cells, (ii) VEGF receptors, including VEGFR-1, VEGFR-2, and VEGFR-3, have been explored for targeted delivery in ocular therapies to manage conditions like ocular angiogenesis, (iii) LyP-1 peptide-modified alginate-based nanoparticles for drug delivery targeting the cornea, (iv) lectin-decorated nanoparticles to enhance binding to inflamed tissues specifically due to infection [128,129,130,132].
For development of receptor-targeted DDSs for conjunctivitis, certain receptors can be explored, for example- (i) hyaluronic acid is often used to target CD44 receptors, which are overexpressed in inflamed or diseased conjunctival tissues. HA-modified drugs can enhance drug delivery to the conjunctiva by binding specifically to CD44 receptors [133]. (ii) LHRH and transferrin receptor can be targeted by nanoparticles conjugated with deslorelin or transferrin significantly enhancing conjunctival uptake of nanoparticles and hence, can be explored for conjunctivitis for enhanced efficacy and lower toxicity [134]. (iii) Lectins can specifically bind to carbohydrate moieties on the surface of conjunctival cells. Lectin-functionalized nanoparticles can enhance drug delivery by targeting these glycan structures [135]. Receptor-targeted DDSs offer a promising approach to improve the efficacy of treatments by specifically targeting infected cells or tissues. An example of receptor-targeted drug DDSs that could be applied for the treatment of endophthalmitis is as follows: Nanoparticles conjugated with ligands for TLRs enhance the delivery of antimicrobial agents to the infected sites by targeting immune cells that express TLRs, thereby improving the immune response against pathogens causing endophthalmitis [136]. These receptor-targeted DDSs aim to improve the specificity and efficacy of treatments for ocular infection by leveraging the interactions between ligands and receptors expressed on immune cells and infected tissues.

5. Combination Therapies for Infectious Ocular Diseases

The efficacy of current drug therapies for treating infectious ocular diseases is at risk of becoming obsolete due to inadequate physical, chemical, biological, and pharmacokinetic properties of drugs, along with low bioavailability caused by ocular barriers, inflammation associated with infection, and the elevated risk of developing resistance [8,137]. To tackle this problem, researchers have investigated development of DDSs and combinatorial therapy approaches. In combinatorial therapies, the major approaches investigated are (i) utilizing combinations of two or more antibiotics, (ii) pairing antifungal agents with antibiotics, (iii) combinations of two or more antifungal agents, (iv) merging anti-infective agents with anti-inflammatory drugs, (v) combinatorial therapy, including antimicrobial agents and photodynamic therapy, and (vi) antibacterial and comfort agents for eye-related conditions [8,15,16,17,18].
In treating bacterial eye infections, the choice of drugs can vary. Typically, antibiotics target specific aspects of bacterial physiology: interfering with cell wall formation for gram-positive bacteria and disrupting ribosomal protein synthesis for gram-negative bacteria, thus enhancing drug specificity. For acute-onset bacterial endophthalmitis, current therapy involves a combination of vancomycin for gram-positive coverage, and amikacin and ceftazidime for gram-negative coverage. This combination therapy approach may yield improved outcomes, particularly in cases involving antibiotic-resistant bacteria [17]. Moreover, combining antifungal agents with antibiotics demonstrates an appealing potential to reduce the duration, stress, and fatigue linked with topical treatment regimens against B-hemolytic Streptococcus spp., Aspergillus spp., and Fusarium spp. [138].
Combination antifungal therapy has been used for fungal eye infections to increase efficacy, reduce toxicity and improve the prognosis of the patient in infections that are difficult to treat. The concurrent administration of natamycin and voriconazole for treating fungal keratitis patients effectively eliminates ocular fungi, enhances ocular symptoms and visual acuity improvement, and maintains good safety with no significant increase in adverse reactions as compared to natamycin alone [139]. In posterior infectious diseases such as endophthalmitis, inflammation linked to infection represents a significant challenge. To tackle this issue, researchers have investigated combination therapy for disorders affecting the posterior segment of the eye.
A combination of anti-inflammatory agents like triamcinolone acetonide, prednisolone acetate, dexamethasone, and ketorolac, has been employed alongside anti-infective agents like antibiotics or antifungal agents [8,53,140,141,142]. By incorporating anti-inflammatory and anti-microbial agents into a single delivery system, researchers could achieve synergistic effects on both anti-inflammatory and antimicrobial properties, leading to improved treatment outcomes [8,143]. The sequential release of antibacterial followed by anti-inflammatory drugs in infectious diseases like endophthalmitis is essential. To achieve this, Ailinmg Yu et al. developed a supramolecular hydrogel formed by co-assembly of a sparingly soluble antibiotic (levofloxacin, Lev) and dexamethasone-peptide amphiphile (Dex-SA-RGD). In the in vitro drug release study, it was observed that Lev was swiftly released from the supramolecular hydrogel within 6 hours. Conversely, Dex-SA-RGD escaped from the hydrogel due to matrix erosion, subsequently functioning as a prodrug to facilitate the sequential slow release of active dexamethasone via hydrolysis. The Lev/Dex-SA-RGD supramolecular hydrogel exhibited potent antibacterial activity against both gram-negative and gram-positive strains [143].
In efforts to combat bacterial and fungal resistance, researchers have explored combining antimicrobial agents with photodynamic therapy, yielding synergistic effects [18,144,145]. Furthermore, to make the drug formulation more patient compliant and offer relief for microbial infections, a contact lens is designed with moxifloxacin, an antibacterial agent, and hyaluronic acid for added comfort during conjunctivitis treatment. This specialized lens ensures sustained delivery of the medication for up to 96 h, providing both therapeutic effectiveness and comfort for the eye [16]. Despite the potential effectiveness of various combination of therapeutic agents, conventional dosage forms face challenges, including limited access to infected ocular tissues/cells, poor solubility, short half-life, and toxicity, highlighting the pressing need for innovative scientific research to address emerging rapid resistance to these drugs. DDSs can overcome ocular barriers and target specific ocular tissues by varying physicochemical properties, while increasing drug solubility and enhancing the half-life of the drug with reductions in ocular toxicity [8,11,12,56,88].

6. Stimuli Responsive Drug Delivery Systems for Infectious Ocular Diseases

In recent years, there has been a significant surge of interest in stimuli-responsive DDSs in infectious ocular diseases. This heightened attention stems from their ability to precisely regulate the release of drugs in both space and time, allowing for customizable drug release tailored to the patient’s physiological or pathological state [146]. These DDSs can react to various stimuli, such as changes in temperature, pH levels, presence of ions, enzymatic activity, changes in ROS, exposure to light, magnetic fields, and ultrasound (Figure 3) [20,21,22,23,64].
Temperature-sensitive ocular DDSs utilize changes in temperature, which can be higher in inflamed or infected areas, to trigger drug release. Thermoresponsive gels are materials that undergo a reversible sol-gel transition in response to changes in temperature (Figure 3A). Thermoresponsive gels or nanoparticle-loaded thermoresponsive gels have been explored for all the infectious ocular diseases like keratitis, conjunctivitis and endophthalmitis [20,105,106]. Acyclovir-loaded thermosensitive in situ gel nanoemulsions showed a 2.8 time higher corneal penetration than the control drug, and also provided sustained acyclovir release without having corneal irritation, which sounds to be a promising treatment strategy for viral keratitis [20]. In the oxytetracycline-loaded gelatin-polyacrylic acid nanoparticles-loaded in poloxamer-N407 thermo- responsive hydrogel for the treatment of bacterial keratitis, the residence time of the drug is enhanced, which shows antibacterial activity against Pseudomonas aeruginosa in vitro and in vivo, which is comparable with the commercial counterpart. A Voriconazole-loaded thermos-responsive in situ gelling system was prepared using poloxamer 188, poloxamer 407 and carboxymethylcellulose. This DDS increased residence time and the bioavailability of voriconazole in the ocular mucosa, provided sustained drug release, and protected the cornea and conjunctiva from damage due to fungal infection [98]. Vancomycin-loaded poly (ethylene glycol) diacrylate and poly(N-isopropylacrylamide)-based thermo-responsive hydrogel DDS showed a lower infection score than the control drug in Staphylococcus aureus induced rat endophthalmitis model [105].
Enzyme-responsive ocular DDSs react to specific enzymes that are overexpressed in healthy and infected tissues. In the eye, various enzymes are present under both physiological and pathological conditions, such as matrix metalloproteinases (MMP) in tissues, hyaluronidase in the vitreous, and lysozyme and esterase in tears (Figure 3B) [147]. These enzymes can be targeted for development of enzyme-responsive DDSs. MMP-sensitive supramolecular nanoparticles (MMP-S NPs) were developed to enhance the photodynamic antibacterial effect against biofilm-associated bacterial keratitis. These nanoparticles were prepared via host-guest self-assembly between hydrophobic Ce6-conjugated β-cyclodextrin (β-CD) and hydrophilic MMP-9-sensitive peptide-modified adamantane. The MMP-9-sensitive peptide included three functional moieties: a cationic AMP sequence (YGRKKKRRQRRR), an MMP-9-responsive sequence (GPLGVRG), and a negatively charged peptide sequence (EEEEEE). The negatively charged EEEEEE acted as an antifouling layer to prevent adhesion to normal ocular surfaces or healthy corneal cells, but was stripped off by overexpressed MMP-9 at the infection site. This exposure of the cationic AMPs allowed the nanoparticles to penetrate deeply into the biofilm and bind to bacterial surfaces through electrostatic interactions, thereby enhancing the antimicrobial efficacy of photodynamic therapy (PDT) against Pseudomonas aeruginosa [148]. Chitosan-coated liposomes containing levofloxacin were developed to use lysozyme in tears as an intelligent “switch” for precise antibiotic delivery through the catalytic degradation of chitosan. The release rate of levofloxacin depends on the lysozyme concentration and the degree of chitosan deacetylation. This drug carrier shows significant potential to enhance targeted antibiotic delivery, achieve on-demand controlled release, increase drug delivery efficiency, reduce side effects, and improve anti-infective therapy for bacterial keratitis [23].
pH-sensitive ocular DDSs exploit the differences in pH between healthy and infected tissues to release drugs precisely where needed. The change in pH value triggers different nanocarriers to release drugs at specific sites using two main strategies (Figure 3C). Polymers with many acidic groups experience increased electrostatic repulsion and swelling at high pH. Conversely, polymers with basic groups ionize and exhibit electrostatic repulsion at low pH [147]. However, the eye can tolerate a pH range of 4 to 10, with the pH of tears being approximately 7.4. Natural or synthetic pH-responsive polymers are designed to respond effectively within this environment and explored for keratitis, conjunctivitis, and endophthalmitis [21,22,147]. Ciprofloxacin-loaded Poly (N-isopropylacrylamide-methacrylic acid-vinylpyrrolidone) nanogels demonstrated enhanced antibacterial effects in vitro and showed reasonable efficacy in treating severe keratitis in vivo. This developed system has the potential for localized application in the treatment of keratitis [22]. A pH-sensitive metal oxide framework was developed for chemotherapy and photodynamic therapy against bacterial endophthalmitis. This hybrid DDS utilizes a pH-responsive zeolitic imidazolate framework-8-polyacrylic acid material, loaded with ammonium methylbenzene blue, and further modified with AgNO3/dopamine for in situ reduction to silver nanoparticles. The system undergoes a secondary modification with vancomycin/NH2-polyethylene glycol, forming a composite nanomaterial. This system demonstrates antibacterial activity against Escherichia coli, Staphylococcus aureus, and methicillin-resistant S. aureus, and exhibits cytocompatibility with both corneal and retinal pigmented epithelial cells, along with in vivo biocompatibility after ocular injection. The DDS shows synergistic and long-term antibacterial activity [21].
Ion-responsive ocular DDSs take advantage of the presence of Na+, K+, Mg2+ and Ca2+ in tears. These systems offer controlled and sustained drug release, which is particularly beneficial given the anatomical and physiological barriers of the eye [147]. The ion-activated in situ gels can respond to the ionic composition of tear fluid, allowing the gel to form on the ocular surface and release the drug over an extended period. This approach helps in maintaining effective drug concentrations at the site of infection, reducing the frequency of administration, and improving patient compliance. For example, Kesavan et al. developed ion-responsive hydrogels using gellan gum or sodium alginate combined with a sodium methylcellulose sulfate to treat experimental bacterial keratitis. The gatifloxacin-loaded hydrogel demonstrated significantly stronger adhesion compared to commercially available gatifloxacin formulations, thereby effectively prolonging its retention time on the ocular surface. On the 4th day of treatment, there were notable differences in keratitis scores between the hydrogel group and the control group. Specifically, the hydrogel group showed significant improvements in symptoms such as chemosis, eye irritation, tear secretion, mucus removal, and eyelid swelling [149]. Díaz-Tomé et al. developed ion-activated in situ gels using gellan gum, κ-carrageenan, or hyaluronic acid for treating fungal keratitis. Their study demonstrated that, compared to a free drug solution, the residual amount of econazole in two hydrogel formulations was approximately double that of the free drug solution two hours after administration, as evidenced by PET-CT imaging without having any ocular irritation [150].
ROS sensitive ocular systems have shown significant promise in the treatment of infectious diseases, particularly due to their ability to target and release drugs in response to the oxidative stress associated with infections (Figure 3D) [64,151]. A ROS-sensitive nanoparticle-based eye drop has been developed using glycol chitosan (GC) as the nanocarrier and 4-carboxyphenylboronic acid pinacol ester (EB) as the ROS-responsive component. This innovative DDS is loaded with voriconazole (VOR). The developed GC-EB-VOR nanoparticle-based eye drop demonstrates high corneal penetration, controlled drug release under low ROS concentrations, effective ROS depletion, and promising antifungal efficacy, making it a potential therapy for fungal keratitis [50]. Another innovative approach involves using nanoparticles modified with ROS-sensitive polymers. For instance, researchers developed a system where poly (ethylene glycol)-thioketal-coated nanoparticles encapsulated with moxifloxacin and functionalized with a bacterial-targeting peptide were used to treat bacterial biofilms and endophthalmitis. The ROS-responsive degradation of the polymer coating ensures that the drug is released precisely at the site of infection, where ROS levels are high, thereby enhancing the antibacterial effects and reducing systemic toxicity [151].
The other stimuli responses, such as light, magnetic fields, and ultrasound responsive DDSs are reported for other ocular diseases such as glaucoma, age related macular degeneration, diabetic retinopathy, and uveitis, and can be extrapolated for infectious ocular diseases such as keratitis, conjunctivitis and endophthalmitis [147].

7. Safety and Biocompatibility of Ocular Drug Delivery Systems

The eye is an essential sensory organ that plays an important role in visualization of surroundings. Ocular infections can stem from bacteria, fungi, parasites, or viruses, each capable of inducing a range of diseases. At present, a variety of ophthalmic DDSs are accessible in the market, with others progressing through preclinical and clinical phases. Nonetheless, there is a scarcity of data regarding the safety, efficacy, and toxicology of these advanced ocular DDSs. Multiple observable and quantifiable parameters exist for assessing the toxicity of ocular DDSs both in vivo and in vitro (Figure 4).

7.1. In Vivo Ocular Toxicity Study

The globally accepted standard assay for evaluating acute ocular toxicity is known as the eye irritation test, or in vivo Draize eye test, developed by the FDA [54,105,126,154]. This method serves as an endorsed means to assess the safety and toxicity of materials employed in ophthalmic formulations. The DDSs are administered into the conjunctival sac, and signs of irritation such as redness, swelling, cloudiness, edema, hemorrhage, discharge, and blindness are observed in this test (Figure 4A) [7,61,105,106,118,126]. An eye irritation study can also be conducted by applying the DDSs to one eye while using the other eye as a control. Histological analysis can then be performed to assess inflammation and cell death. DDSs for ophthalmic applications are quickly advancing, aiming to enhance the safety and efficacy of targeted therapeutics while improving patient compliance (Figure 4A) [7,88]. Ocular organotypic models, or the Enucleated Eye Test (EET), use isolated rabbit eyes from animals utilized for other research or commercial purposes. DDSs can be applied at relevant concentrations, and observations are made for corneal opacity and swelling. Corneal opacity indicates protein denaturation, swelling, vacuolation, and damage to the epithelium and stroma. Bovine and porcine corneas, as well as enucleated chicken eyes, are widely accepted as reliable and accurate tissues for assessing eye irritation. The EET is considered a scientifically sound method for evaluating and identifying substances that are either non-irritants or cause irreversible eye damage [155,156]. The in vivo toxicity of DDSs can be assessed by measuring changes in intraocular pressure and evaluating retinal function using an electroretinogram after intravitreal injection. This evaluation is crucial for DDSs developed for treating posterior infectious eye diseases, such as endophthalmitis [55,155,157]. DDSs that penetrate ocular tissues or are administered intraocularly may impact retinal morphology and circulation. These effects can be experimentally assessed using optical coherence tomography (OCT) for retinal morphology, ultrasound Doppler measurements for circulation, and retinal fluorescein angiography for vascular analysis (Figure 4B) [155].

7.2. In Vitro Cytotoxicity Assays

A cytotoxicity assay for ocular applications is designed to evaluate the potential toxic effects of DDSs on ocular cells. This is crucial for ensuring the safety and efficacy of ocular DDSs (Figure 4C) [11,12]. The different cell line or cells explored for cytotoxicity study for ocular applications are majorly based on the targeted tissues in the eye. Commonly used cell lines/cells include human corneal epithelial cells (HCE), rabbit corneal epithelial cell line (SIRC), rabbit corneal keratocytes, retinal pigment epithelial cells (ARPE-19), human conjunctival epithelial cells (WKD), Muller glia (MIO-M1), Microglia (BV-2), and macrophages (RAW-267) [8,64,82,86,117]. The cytotoxicity studies were performed by MTT assay, Cell Titer-Glo Luminescent Cell Viability Assay, Cell Counting Kit-8 assay, LDH release assay, LIVE/DEAD assay, and morphological analysis after treatment with ocular DDSs [8,9,64,82,158,159]. The cells under oxidative stress after exposure can be evaluated by DCF (2′,7′-dichlorofluorescein)/DHE (Dihydroethidium) staining, lipid peroxidation, and Nytro tyrosine evaluation [52,82,158]. Maintaining barrier function by epithelial and endothelial cells in the eye is crucial. Changes in barrier integrity and function in the presence of DDSs can be evaluated using electron microscopy, transepithelial electrical resistance (TEER) measurements, and permeability studies [159].

7.3. In Vitro Hemocompatibility Study

The hemocompatibility study of DDSs for ocular applications are essential to ensure the safety and efficacy of these delivery systems [8,55]. This study evaluates how DDSs interact with blood components, which is critical for preventing adverse reactions such as blood clotting, hemolysis, and immune responses when the DDSs come into contact with the bloodstream, either directly or indirectly [8,55]. Understanding the hemocompatibility of nanoparticles helps in optimizing their design for ocular drug delivery, ensuring they are safe for use in medical treatments involving the eye. A hemolysis assay is a laboratory test used to evaluate the compatibility of DDS with red blood cells (RBCs). This assay measures the extent to which nanoparticles cause the rupture or lysis of RBCs, leading to the release of hemoglobin into the surrounding plasma [8,55,93,160]. Furthermore, the plasma recalcification assay and blood-clotting assay measures the time it takes for plasma to clot after the addition of calcium chloride, which triggers the coagulation cascade. It is essential to ensure that ocular DDSs do not induce unwanted coagulation or thrombotic events when they come into contact with blood or ocular tissues. By incorporating the plasma recalcification assay into the development process, researchers can ensure that ocular DDSs are both effective and safe, minimizing the risk of coagulation-related complications [8,55,160] (Figure 4D).

7.4. Ocular Tolerance Test (HET-CAM)

The Hen’s Egg Test on the Chorioallantoic Membrane (HET-CAM) is an in vitro assay used to assess the irritation potential of substances, including those intended for ocular applications (Figure 4E). This test serves as an alternative to the Draize rabbit eye test, reducing the need for animal testing. The HET-CAM test evaluates the irritant effects of a test substance on the highly vascularized chorioallantoic membrane of a fertilized chicken egg. This is employed to assess the predictive model for in vitro irritation by observing vascular damage, hemorrhage, hyperemia, and coagulation, and by computing an irritation score. The CAM contains arteries, veins, and capillaries, which provide reactions similar to inflammation-induced responses in the corneal tissue of rabbits during the Draize eye irritancy test [20,88,96,153].

7.5. In Vitro Genotoxicity Assay

A genotoxicity study for an ocular DDS is essential to evaluate whether the system causes genetic damage that could lead to mutations or cell death due to DNA strand break. These studies typically involve in vitro assays designed to detect various types of genetic damage, such as gene mutations, chromosomal aberrations, and DNA strand breaks (Figure 4C) [82,158]. The DNA strand break can be studied by comet assay, DNA gel electrophoresis, and DAPI staining [82,158]. Gene mutation assays detect changes in the DNA base sequence that result from DNA misrepair or replication errors. Mutations in critical genes can increase the risk of tumor formation. One of the classical methods for detecting such mutations is the bacterial mutation assay, known as the Ames test. This test helps ensure that the materials and drugs designed for ocular use do not induce genetic mutations that could lead to adverse effects such as tumor formation [96]. Assays for chromosomal damage detect alterations in chromosome structure, resulting in chromatid-type and chromosome-type breaks and rearrangements, or in chromosome number, leading to the production of aneuploid or polyploid cells. By incorporating assays that detect chromosomal damage, researchers can ensure that ocular DDSs are not only effective in treating eye conditions, but are also safe for long-term use, minimizing the risk of genetic alterations and associated health issues [161].

8. Marketed Products for Infectious Eye Diseases Based on Drug Delivery Systems

The market offers a range of innovative DDSs specifically designed to treat infectious eye diseases. These formulations enhance drug retention, bioavailability, and therapeutic efficacy, overcoming the challenges posed by ocular barriers. For instance, Azithromycin in DuraSite® is a 1% azithromycin formulation that utilizes a low-viscosity, gel-forming delivery system. This system is soluble in tear fluid, allowing for increased drug retention time on the ocular surface, making it effective for the treatment of bacterial conjunctivitis [24]. Similarly, Moxifloxacin hydrochloride ophthalmic solution 0.5% (Vigamox®) is an ocular adaptation of moxifloxacin, designed to enhance penetration into ocular tissues and fluids. It is commonly used for the treatment of bacterial eye infections [25]. Natamycin ophthalmic suspension (Natacyn®) is the only commercially available antifungal agent FDA approved for the treatment of superficial fungal eye infections, including fungal keratitis, blepharitis, and conjunctivitis [162]. Zirgan®, a 0.15% aqueous gel formulation of ganciclovir, is approved as a topical antiviral agent for the treatment of acute herpes simplex epithelial dendritic ulcerative keratitis [163]. In addition to treating infections, various formulations such as AzaSite Plus™, DexaSiteTM, Lotemax®, and FML® have been developed to address ocular inflammation related to eye surgery, infections, and post-infectious inflammatory conditions [164,165,166]. These products leverage various DDS technologies to enhance drug efficacy, retention, and safety in treating infectious eye diseases.

9. Conclusions and Future Directions

The landscape of DDSs for infectious ocular diseases has significantly advanced, offering new avenues for effective treatment of conditions such as keratitis, conjunctivitis, and endophthalmitis. Enhanced drug delivery methods, including targeted approaches and stimuli-responsive systems, have demonstrated the potential to improve therapeutic outcomes by increasing drug bioavailability and precision targeting within ocular tissues. Safety and biocompatibility remain critical considerations in the development of these advanced systems. In vivo and in vitro studies, including cytotoxicity assays, hemocompatibility tests, and genotoxicity evaluations are essential to ensure that new drug delivery technologies are safe for clinical applications. The HET-CAM ocular tolerance test in particular provides valuable insights into the irritation potential of these systems.
While significant progress has been made in developing innovative DDSs for ocular infections, several major challenges persist, including antimicrobial resistance, systemic side effects, ocular toxicity, chronic and recurrent infections, and the need for invasive procedures. In addition, issues such as drug stability, delivery efficiency, and patient compliance require ongoing attention. Overcoming these obstacles will necessitate the development of personalized therapies, rapid diagnostic tools, and a deeper understanding of antimicrobial peptides and host defense mechanisms, alongside advancements in drug stability and delivery technologies.

Author Contributions

B.M. and A.D. originally conceptualized and wrote this article during the COVID-19 pandemic lockdown; B.M., N.B. and A.D. have rewritten, reviewed and edited it. All authors have read and agreed to the published version of the manuscript.

Funding

No funding is received for this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors acknowledge BioRender and MS PowerPoint that were partly used for preparation of the figures.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Watson, S.; Cabrera-Aguas, M.; Khoo, P. Common eye infections. Aust. Prescr. 2018, 41, 67–72. [Google Scholar] [CrossRef] [PubMed]
  2. Stiles, J. 128—Ocular Infections. In Greene’s Infectious Diseases of the Dog and Cat, 5th ed.; Sykes, J.E., Ed.; W.B. Saunders: Philadelphia, PA, USA, 2021; pp. 1688–1709. [Google Scholar]
  3. Ghafoorianfar, S.; Ghorani-Azam, A.; Mohajeri, S.A.; Farzin, D. Efficiency of nanoparticles for treatment of ocular infections: Systematic literature review. J. Drug Deliv. Sci. Technol. 2020, 57, 101765. [Google Scholar] [CrossRef]
  4. Chigbu, D.I.; Labib, B.A. Pathogenesis and management of adenoviral keratoconjunctivitis. Infect. Drug Resist. 2018, 11, 981–993. [Google Scholar] [CrossRef] [PubMed]
  5. Kakkar, S.; Karuppayil, S.M.; Raut, J.S.; Giansanti, F.; Papucci, L.; Schiavone, N.; Kaur, I.P. Lipid-polyethylene glycol based nano-ocular formulation of ketoconazole. Int. J. Pharm. 2015, 495, 276–289. [Google Scholar] [CrossRef] [PubMed]
  6. Khalil, I.A.; Ali, I.H.; El-Sherbiny, I.M. Noninvasive biodegradable nanoparticles-in-nanofibers single-dose ocular insert: In vitro, ex vivo and in vivo evaluation. Nanomedicine 2019, 14, 33–55. [Google Scholar] [CrossRef]
  7. Eid, H.M.; Elkomy, M.H.; El Menshawe, S.F.; Salem, H.F. Development, optimization, and in vitro/in vivo characterization of enhanced lipid nanoparticles for ocular delivery of ofloxacin: The influence of pegylation and chitosan coating. AAPS Pharmscitech 2019, 20, 1–14. [Google Scholar] [CrossRef]
  8. Mahaling, B.; Baruah, N.; Ahamad, N.; Maisha, N.; Lavik, E.; Katti, D.S. A non-invasive nanoparticle-based sustained dual-drug delivery system as an eyedrop for endophthalmitis. Int. J. Pharm. 2021, 606, 120900. [Google Scholar] [CrossRef]
  9. Ahsan, S.M.; Rao, C.M. Condition responsive nanoparticles for managing infection and inflammation in keratitis. Nanoscale 2017, 9, 9946–9959. [Google Scholar] [CrossRef] [PubMed]
  10. Nguyen, D.D.; Luo, L.-J.; Lai, J.-Y. Toward understanding the purely geometric effects of silver nanoparticles on potential application as ocular therapeutics via treatment of bacterial keratitis. Mater. Sci. Eng. C 2021, 119, 111497. [Google Scholar] [CrossRef]
  11. Mahaling, B.; Katti, D.S. Understanding the influence of surface properties of nanoparticles and penetration enhancers for improving bioavailability in eye tissues in vivo. Int. J. Pharm. 2016, 501, 1–9. [Google Scholar] [CrossRef]
  12. Mahaling, B.; Katti, D.S. Physicochemical properties of core–shell type nanoparticles govern their spatiotemporal biodistribution in the eye. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 2149–2160. [Google Scholar] [CrossRef]
  13. Giannaccini, M.; Giannini, M.; Calatayud, M.P.; Goya, G.F.; Cuschieri, A.; Dente, L.; Raffa, V. Magnetic nanoparticles as intraocular drug delivery system to target retinal pigmented epithelium (RPE). Int. J. Mol. Sci. 2014, 15, 1590–1605. [Google Scholar] [CrossRef]
  14. Giannaccini, M.; Pedicini, L.; De Matienzo, G.; Chiellini, F.; Dente, L.; Raffa, V. Magnetic nanoparticles: A strategy to target the choroidal layer in the posterior segment of the eye. Sci. Rep. 2017, 7, 43092. [Google Scholar] [CrossRef]
  15. Deepthi, S.; Jose, J. Novel hydrogel-based ocular drug delivery system for the treatment of conjunctivitis. Int. Ophthalmol. 2019, 39, 1355–1366. [Google Scholar] [CrossRef]
  16. Maulvi, F.A.; Singhania, S.S.; Desai, A.R.; Shukla, M.R.; Tannk, A.S.; Ranch, K.M.; Vyas, B.A.; Shah, D.O. Contact lenses with dual drug delivery for the treatment of bacterial conjunctivitis. Int. J. Pharm. 2018, 548, 139–150. [Google Scholar] [CrossRef] [PubMed]
  17. Kodati, S.; Eller, A.W.; Kowalski, R.P. The Susceptibility of Bacterial Endophthalmitis Isolates to Vancomycin, Ceftazidime, and Amikacin: A 23-Year Review. Ophthalmol. Retin. 2017, 1, 206–209. [Google Scholar] [CrossRef]
  18. Sushma, M.V.; Sankaranarayanan, S.A.; Bantal, V.; Pemmaraju, D.B.; Rengan, A.K. Ethosomal Nanoformulations for Combinational Photothermal Therapy of Fungal Keratitis. Adv. Ther. 2023, 6, 2200331. [Google Scholar] [CrossRef]
  19. Josyula, A.; Omiadze, R.; Parikh, K.; Kanvinde, P.; Appell, M.B.; Patel, P.; Saeed, H.; Sutar, Y.; Anders, N.; He, P.; et al. An ion-paired moxifloxacin nanosuspension eye drop provides improved prevention and treatment of ocular infection. Bioeng. Transl. Med. 2021, 6, e10238. [Google Scholar] [CrossRef]
  20. Mahboobian, M.M.; Mohammadi, M.; Mansouri, Z. Development of thermosensitive in situ gel nanoemulsions for ocular delivery of acyclovir. J. Drug Deliv. Sci. Technol. 2020, 55, 101400. [Google Scholar] [CrossRef]
  21. Chen, H.; Yang, J.; Sun, L.; Zhang, H.; Guo, Y.; Qu, J.; Jiang, W.; Chen, W.; Ji, J.; Yang, Y.-W.; et al. Synergistic Chemotherapy and Photodynamic Therapy of Endophthalmitis Mediated by Zeolitic Imidazolate Framework-Based Drug Delivery Systems. Small 2019, 15, 1903880. [Google Scholar] [CrossRef] [PubMed]
  22. Davaran, S.; Lotfipour, F.; Sedghipour, N.; Sedghipour, M.R.; Alimohammadi, S.; Salehi, R. Preparation and in vivo evaluation of in situ gel system as dual thermo-/pH-responsive nanocarriers for sustained ocular drug delivery. J. Microencapsul. 2015, 32, 511–519. [Google Scholar]
  23. Dong, Y.; Wu, T.; Jiang, T.; Zhu, W.; Chen, L.; Cao, Y.; Xiao, Y.; Peng, Y.; Wang, L.; Yu, X.; et al. Chitosan-coated liposome with lysozyme-responsive properties for on-demand release of levofloxacin. Int. J. Biol. Macromol. 2024, 269, 132271. [Google Scholar] [CrossRef] [PubMed]
  24. Luchs, J. Azithromycin in DuraSite for the treatment of blepharitis. Clin. Ophthalmol. 2010, 4, 681–688. [Google Scholar] [CrossRef]
  25. Miller, D. Review of moxifloxacin hydrochloride ophthalmic solution in the treatment of bacterial eye infections. Clin. Ophthalmol. 2008, 2, 77–91. [Google Scholar] [CrossRef]
  26. Clare, G.; Kempen, J.H.; Pavésio, C. Infectious eye disease in the 21st century—An overview. Eye 2024, 38, 2014–2027. [Google Scholar] [CrossRef]
  27. Cabrera-Aguas, M.; Khoo, P.; Watson, S.L. Infectious keratitis: A review. Clin. Exp. Ophthalmol. 2022, 50, 543–562. [Google Scholar] [CrossRef]
  28. Ung, L.; Chodosh, J. Foundational concepts in the biology of bacterial keratitis. Exp. Eye Res. 2021, 209, 108647. [Google Scholar] [CrossRef] [PubMed]
  29. Das, S.; D’Souza, S.; Gorimanipalli, B.; Shetty, R.; Ghosh, A.; Deshpande, V. Ocular Surface Infection Mediated Molecular Stress Responses: A Review. Int. J. Mol. Sci. 2022, 23, 3111. [Google Scholar] [CrossRef] [PubMed]
  30. Ahmadikia, K.; Aghaei Gharehbolagh, S.; Fallah, B.; Naeimi Eshkaleti, M.; Malekifar, P.; Rahsepar, S.; Getso, M.I.; Sharma, S.; Mahmoudi, S. Distribution, Prevalence, and Causative Agents of Fungal Keratitis: A Systematic Review and Meta-Analysis (1990 to 2020). Front. Cell. Infect. Microbiol. 2021, 11, 698780. [Google Scholar] [CrossRef] [PubMed]
  31. Tuli, S.S. Fungal keratitis. Clin. Ophthalmol. 2011, 5, 275–279. [Google Scholar] [CrossRef] [PubMed]
  32. Mills, B.; Radhakrishnan, N.; Karthikeyan Rajapandian, S.G.; Rameshkumar, G.; Lalitha, P.; Prajna, N.V. The role of fungi in fungal keratitis. Exp. Eye Res. 2021, 202, 108372. [Google Scholar] [CrossRef]
  33. Kaye, S.; Choudhary, A. Herpes simplex keratitis. Prog. Retin. Eye Res. 2006, 25, 355–380. [Google Scholar] [CrossRef]
  34. Farooq, A.V.; Shukla, D. Herpes simplex epithelial and stromal keratitis: An epidemiologic update. Surv. Ophthalmol. 2012, 57, 448–462. [Google Scholar] [CrossRef]
  35. Missotten, L. Immunology and herpetic keratitis. Eye 1994, 8, 12–21. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, L.; Wang, R.; Xu, C.; Zhou, H. Pathogenesis of herpes stromal keratitis: Immune inflammatory response mediated by inflammatory regulators. Front. Immunol. 2020, 11, 766. [Google Scholar] [CrossRef]
  37. Rajasagi, N.K.; Rouse, B.T. The Role of T Cells in Herpes Stromal Keratitis. Front. Immunol. 2019, 10, 512. [Google Scholar] [CrossRef] [PubMed]
  38. Fanselow, N.; Sirajuddin, N.; Yin, X.T.; Huang, A.J.W.; Stuart, P.M. Acanthamoeba Keratitis, Pathology, Diagnosis and Treatment. Pathogens 2021, 10, 323. [Google Scholar] [CrossRef] [PubMed]
  39. Neelam, S.; Niederkorn, J.Y. Pathobiology and Immunobiology of Acanthamoeba Keratitis: Insights from Animal Models. Yale J. Biol. Med. 2017, 90, 261–268. [Google Scholar]
  40. Ortillés, Á.; Goñi, P.; Rubio, E.; Sierra, M.; Gámez, E.; Fernández, M.T.; Benito, M.; Cristóbal, J.Á.; Calvo, B. A Rabbit Model of Acanthamoeba Keratitis: Use of Infected Soft Contact Lenses After Corneal Epithelium Debridement With a Diamond Burr. Investig. Ophthalmol. Vis. Sci. 2017, 58, 1218–1227. [Google Scholar] [CrossRef]
  41. de Lacerda, A.G.; Lira, M. Acanthamoeba keratitis: A review of biology, pathophysiology and epidemiology. Ophthalmic Physiol. Opt. 2021, 41, 116–135. [Google Scholar] [CrossRef]
  42. Cvenkel, B.; Globočnik, M. Conjunctival scrapings and impression cytology in chronic conjunctivitis. Correlation with microbiology. Eur. J. Ophthalmol. 1997, 7, 19–23. [Google Scholar] [CrossRef]
  43. Livingston, E.T.; Mursalin, M.H.; Callegan, M.C. A Pyrrhic Victory: The PMN Response to Ocular Bacterial Infections. Microorganisms 2019, 7, 537. [Google Scholar] [CrossRef] [PubMed]
  44. Pippin, M.M.; Le, J.K. Bacterial Conjunctivitis. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  45. Azari, A.A.; Barney, N.P. Conjunctivitis: A systematic review of diagnosis and treatment. JAMA 2013, 310, 1721–1730. [Google Scholar] [CrossRef] [PubMed]
  46. Gallenga, C.E.; Maritati, M.; Mura, M.; Di Virgilio, F.; Conti, P.; Contini, C. Macrophage Activation in Follicular Conjunctivitis during the COVID-19 Pandemic. Microorganisms 2023, 11, 2198. [Google Scholar] [CrossRef]
  47. Bannier-Hélaouët, M.; Korving, J.; Ma, Z.; Begthel, H.; Giladi, A.; Lamers, M.M.; van de Wetering, W.J.; Yawata, N.; Yawata, M.; LaPointe, V.L.S.; et al. Human conjunctiva organoids to study ocular surface homeostasis and disease. Cell Stem Cell 2024, 31, 227–243.e212. [Google Scholar] [CrossRef] [PubMed]
  48. Kernt, M.; Kampik, A. Endophthalmitis: Pathogenesis, clinical presentation, management, and perspectives. Clin. Ophthalmol. 2010, 4, 121–135. [Google Scholar] [CrossRef] [PubMed]
  49. Sheu, S.-J. Endophthalmitis. kjo 2017, 31, 283–289. [Google Scholar] [CrossRef]
  50. Rao, N.A.; Hidayat, A.A. Endogenous mycotic endophthalmitis: Variations in clinical and histopathologic changes in candidiasis compared with aspergillosis. Am. J. Ophthalmol. 2001, 132, 244–251. [Google Scholar] [CrossRef]
  51. Soon, M.Y.; Allen, P.J.; Dawkins, R.C.H. Cytokine Expression in Staphylococcal and Streptococcal Endophthalmitis. Biomed. Hub. 2022, 7, 88–98. [Google Scholar] [CrossRef]
  52. Mahaling, B.; Pandala, N.; Wang, H.-C.; Lavik, E.B. Azithromycin protects retinal glia against oxidative stress-induced morphological changes, inflammation, and cell death. ACS Bio Med. Chem. Au 2022, 2, 499–508. [Google Scholar] [CrossRef]
  53. Peyman, G.A.; Herbst, R. Bacterial endophthalmitis: Treatment with intraocular injection of gentamicin and dexamethasone. Arch. Ophthalmol. 1974, 91, 416–418. [Google Scholar] [CrossRef] [PubMed]
  54. Zhou, W.; Wang, Y.; Jian, J.; Song, S. Self-aggregated nanoparticles based on amphiphilic poly (lactic acid)-grafted-chitosan copolymer for ocular delivery of amphotericin B. Int. J. Nanomed. 2013, 3715–3728. [Google Scholar]
  55. Mahaling, B.; Srinivasarao, D.A.; Raghu, G.; Kasam, R.K.; Reddy, G.B.; Katti, D.S. A non-invasive nanoparticle mediated delivery of triamcinolone acetonide ameliorates diabetic retinopathy in rats. Nanoscale 2018, 10, 16485–16498. [Google Scholar] [CrossRef] [PubMed]
  56. Mahaling, B.; Low, S.W.; Ch, S.; Addi, U.R.; Ahmad, B.; Connor, T.B.; Mohan, R.R.; Biswas, S.; Chaurasia, S.S. Next-Generation Nanomedicine Approaches for the Management of Retinal Diseases. Pharmaceutics 2023, 15, 2005. [Google Scholar] [CrossRef]
  57. Lappin, M.; Wotman, K.; Chow, L.; Williams, M.; Hawley, J.; Dow, S. Nanoparticle ocular immunotherapy for herpesvirus surface eye infections evaluated in cat infection model. PLoS ONE 2023, 18, e0279462. [Google Scholar] [CrossRef]
  58. Ghosh, A.K.; Rudramurthy, S.M.; Gupta, A.; Choudhary, H.; Singh, S.; Thakur, A.; Jatana, M. Evaluation of liposomal and conventional amphotericin B in experimental fungal keratitis rabbit model. Transl. Vis. Sci. Technol. 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed]
  59. Agrawal, V.; Kumar, M.; Pathak, K. Defining the properties of pH-sensitive polymeric micellar ocular delivery system of miconazole nitrate for the management of fungal endophthalmitis. Pharm. Nanotechnol. 2014, 2, 157–166. [Google Scholar] [CrossRef]
  60. Mohamed, S.A.; Abdelgawad, M.A.; Alaaeldin, R.; Fathalla, Z.; Moharram, H.; Abdallah, R.M.A.; Abdel-Rahman, I.M.; Abdel-Aziz, M.; Abuo-Rahma, G.E.A.; Ghoneim, M.M.; et al. Solulan C24- and Bile Salts-Modified Niosomes for New Ciprofloxacin Mannich Base for Combatting Pseudomonas-Infected Corneal Ulcer in Rabbits. Pharmaceuticals 2021, 15, 44. [Google Scholar] [CrossRef]
  61. Khalil, R.M.; Abdelbary, G.A.; Basha, M.; Awad, G.E.; El-Hashemy, H.A. Enhancement of lomefloxacin Hcl ocular efficacy via niosomal encapsulation: In vitro characterization and in vivo evaluation. J. Liposome Res. 2017, 27, 312–323. [Google Scholar] [CrossRef]
  62. Abdelbary, G.; El-Gendy, N. Niosome-encapsulated gentamicin for ophthalmic controlled delivery. AAPS Pharmscitech 2008, 9, 740–747. [Google Scholar] [CrossRef]
  63. LaGrow, A.L.; Coburn, P.S.; Miller, F.C.; Land, C.; Parkunan, S.M.; Luk, B.T.; Gao, W.; Zhang, L.; Callegan, M.C. A novel biomimetic nanosponge protects the retina from the Enterococcus faecalis cytolysin. Msphere 2017, 2, 10–1128. [Google Scholar] [CrossRef]
  64. Niu, P.; Wu, Y.; Zeng, F.; Zhang, S.; Liu, S.; Gao, H. Development of nanodrug-based eye drops with good penetration properties and ROS responsiveness for controllable release to treat fungal keratitis. NPG Asia Mater. 2023, 15, 31. [Google Scholar] [CrossRef]
  65. Dandagi, P.; Kerur, S.; Mastiholimath, V.; Gadad, A.; Kulkarni, A. Polymeric ocular nanosuspension for controlled release of acyclovir: In vitro release and ocular distribution. Iran. J. Pharm. Res. 2009, 8, 79–86. [Google Scholar]
  66. Prajapati, B.G.; Patel, A.G.; Paliwal, H. Fabrication of nanoemulsion-based in situ gel using moxifloxacin hydrochloride as model drug for the treatment of conjunctivitis. Food Hydrocoll. Health 2021, 1, 100045. [Google Scholar] [CrossRef]
  67. Youssef, A.A.A.; Thakkar, R.; Senapati, S.; Joshi, P.H.; Dudhipala, N.; Majumdar, S. Design of Topical Moxifloxacin Mucoadhesive Nanoemulsion for the Management of Ocular Bacterial Infections. Pharmaceutics 2022, 14, 1246. [Google Scholar] [CrossRef]
  68. Nasr, M.; Teiama, M.; Ismail, A.; Ebada, A.; Saber, S. In vitro and in vivo evaluation of cubosomal nanoparticles as an ocular delivery system for fluconazole in treatment of keratomycosis. Drug Deliv. Transl. Res. 2020, 10, 1841–1852. [Google Scholar] [CrossRef]
  69. Alharbi, W.S.; Hosny, K.M. Development and optimization of ocular in situ gels loaded with ciprofloxacin cubic liquid crystalline nanoparticles. J. Drug Deliv. Sci. Technol. 2020, 57, 101710. [Google Scholar] [CrossRef]
  70. Hosny, K.M.; Rizg, W.Y.; Alkhalidi, H.M.; Abualsunun, W.A.; Bakhaidar, R.B.; Almehmady, A.M.; Alghaith, A.F.; Alshehri, S.; El Sisi, A.M. Nanocubosomal based in situ gel loaded with natamycin for ocular fungal diseases: Development, optimization, in-vitro, and in-vivo assessment. Drug Deliv. 2021, 28, 1836–1848. [Google Scholar] [CrossRef]
  71. Mahaling, B.; Roy, C.; Ghosh, S. Silk–gelatin hybrid hydrogel: A potential carrier for RNA therapeutics. J. Mater. Chem. B 2024. [CrossRef]
  72. Hu, J.; Zhou, X.; Chen, S.; Yin, D.; Yang, Y.; Chen, M.; Gui, C.; Cai, Y.; Hong, J.; Cheng, Y. A supramolecular gel with unique rheological properties for treating corneal virus infection. Nano Today 2023, 50, 101841. [Google Scholar] [CrossRef]
  73. Gebreel, R.M.; Edris, N.A.; Elmofty, H.M.; Tadros, M.I.; El-Nabarawi, M.A.; Hassan, D.H. Development and Characterization of PLGA Nanoparticle-Laden Hydrogels for Sustained Ocular Delivery of Norfloxacin in the Treatment of Pseudomonas Keratitis: An Experimental Study. Drug Des. Dev. Ther. 2021, 15, 399–418. [Google Scholar] [CrossRef] [PubMed]
  74. Varela-Garcia, A.; Gomez-Amoza, J.L.; Concheiro, A.; Alvarez-Lorenzo, C. Imprinted Contact Lenses for Ocular Administration of Antiviral Drugs. Polymers 2020, 12, 2026. [Google Scholar] [CrossRef]
  75. Roy, G.; Galigama, R.D.; Thorat, V.S.; Mallela, L.S.; Roy, S.; Garg, P.; Venuganti, V.V.K. Amphotericin B containing microneedle ocular patch for effective treatment of fungal keratitis. Int. J. Pharm. 2019, 572, 118808. [Google Scholar] [CrossRef] [PubMed]
  76. Park, S.; Lee, K.; Kang, H.; Lee, Y.; Lee, J.; Kim, J.H.; Song, H.B.; Ryu, W. Single Administration of a Biodegradable, Separable Microneedle Can Substitute for Repeated Application of Eyedrops in the Treatment of Infectious Keratitis. Adv. Healthc. Mater. 2021, 10, 2002287. [Google Scholar] [CrossRef]
  77. Jiang, X.; Jin, Y.; Zeng, Y.; Shi, P.; Li, W. Self-Implantable Core–Shell Microneedle Patch for Long-Acting Treatment of Keratitis via Programmed Drug Release. Small 2024, 20, 2310461. [Google Scholar] [CrossRef]
  78. Polat, H.K.; Bozdağ Pehlivan, S.; Özkul, C.; Çalamak, S.; Öztürk, N.; Aytekin, E.; Fırat, A.; Ulubayram, K.; Kocabeyoğlu, S.; İrkeç, M.; et al. Development of besifloxacin HCl loaded nanofibrous ocular inserts for the treatment of bacterial keratitis: In vitro, ex vivo and in vivo evaluation. Int. J. Pharm. 2020, 585, 119552. [Google Scholar] [CrossRef]
  79. Kim, D.J.; Jung, M.-Y.; Park, J.-H.; Pak, H.-J.; Kim, M.; Chuck, R.S.; Park, C.Y. Moxifloxacin releasing intraocular implant based on a cross-linked hyaluronic acid membrane. Sci. Rep. 2021, 11, 24115. [Google Scholar] [CrossRef]
  80. Sharma, A.; Taniguchi, J. Review: Emerging strategies for antimicrobial drug delivery to the ocular surface: Implications for infectious keratitis. Ocul. Surf. 2017, 15, 670–679. [Google Scholar] [CrossRef]
  81. Bachu, R.D.; Chowdhury, P.; Al-Saedi, Z.H.; Karla, P.K.; Boddu, S.H. Ocular drug delivery barriers—Role of nanocarriers in the treatment of anterior segment ocular diseases. Pharmaceutics 2018, 10, 28. [Google Scholar] [CrossRef]
  82. Luo, L.-J.; Lin, T.-Y.; Yao, C.-H.; Kuo, P.-Y.; Matsusaki, M.; Harroun, S.G.; Huang, C.-C.; Lai, J.-Y. Dual-functional gelatin-capped silver nanoparticles for antibacterial and antiangiogenic treatment of bacterial keratitis. J. Colloid. Interface Sci. 2019, 536, 112–126. [Google Scholar] [CrossRef]
  83. Gaikwad, S.; Ingle, A.; Gade, A.; Rai, M.; Falanga, A.; Incoronato, N.; Russo, L.; Galdiero, S.; Galdiero, M. Antiviral activity of mycosynthesized silver nanoparticles against herpes simplex virus and human parainfluenza virus type 3. Int. J. Nanomed. 2013, 8, 4303–4314. [Google Scholar] [CrossRef]
  84. Elkadery, A.A.S.; Elsherif, E.A.; Ezz Eldin, H.M.; Fahmy, I.A.F.; Mohammad, O.S. Efficient therapeutic effect of Nigella sativa aqueous extract and chitosan nanoparticles against experimentally induced Acanthamoeba keratitis. Parasitol. Res. 2019, 118, 2443–2454. [Google Scholar] [CrossRef]
  85. Frucht-Perry, J.; Assil, K.K.; Ziegler, E.; Douglas, H.; Brown, S.I.; Schanzlin, D.J.; Weinreb, R.N. Fibrin-enmeshed tobramycin liposomes: Single application topical therapy of Pseudomonas keratitis. Cornea 1992, 11, 393–397. [Google Scholar] [CrossRef]
  86. Zorzi, G.K.; Schuh, R.S.; Maschio, V.J.; Brazil, N.T.; Rott, M.B.; Teixeira, H.F. Box Behnken design of siRNA-loaded liposomes for the treatment of a murine model of ocular keratitis caused by Acanthamoeba. Colloids Surf. B Biointerfaces 2019, 173, 725–732. [Google Scholar] [CrossRef]
  87. Ch, S.; Padaga, S.G.; Ghosh, B.; Roy, S.; Biswas, S. Chitosan-poly(lactide-co-glycolide)/poloxamer mixed micelles as a mucoadhesive thermo-responsive moxifloxacin eye drop to improve treatment efficacy in bacterial keratitis. Carbohydr. Polym. 2023, 312, 120822. [Google Scholar] [CrossRef]
  88. Varela-Garcia, A.; Concheiro, A.; Alvarez-Lorenzo, C. Soluplus micelles for acyclovir ocular delivery: Formulation and cornea and sclera permeability. Int. J. Pharm. 2018, 552, 39–47. [Google Scholar] [CrossRef]
  89. Jaiswal, M.; Kumar, M.; Pathak, K. Zero order delivery of itraconazole via polymeric micelles incorporated in situ ocular gel for the management of fungal keratitis. Colloids Surf. B: Biointerfaces 2015, 130, 23–30. [Google Scholar] [CrossRef] [PubMed]
  90. Li, R.; Rinaldi, M. In vitro antifungal activity of nikkomycin Z in combination with fluconazole or itraconazole. Antimicrob. Agents Chemother. 1999, 43, 1401–1405. [Google Scholar] [CrossRef] [PubMed]
  91. El-Mofty, H.M.; El-Nabarawi, M.A.; Abd El Rehem, R.T.; Teaima, M.H.; Abary, M.Y.S.; Salah, M.; Lotfy, N.M. Niosomes: Do They Increase the Potency of Topical Natamycin Ketorolac Formula in Treating Aspergillus Keratitis? An Experimental Study. J. Ocul. Pharmacol. Ther. 2020, 36, 545–554. [Google Scholar] [CrossRef]
  92. El-Nabarawi, M.A.; Abd El Rehem, R.T.; Teaima, M.; Abary, M.; El-Mofty, H.M.; Khafagy, M.M.; Lotfy, N.M.; Salah, M. Natamycin niosomes as a promising ocular nanosized delivery system with ketorolac tromethamine for dual effects for treatment of candida rabbit keratitis; in vitro/in vivo and histopathological studies. Drug Dev. Ind. Pharm. 2019, 45, 922–936. [Google Scholar] [CrossRef]
  93. Iqbal, K.; Abdalla, S.A.O.; Anwar, A.; Iqbal, K.M.; Shah, M.R.; Anwar, A.; Siddiqui, R.; Khan, N.A. Isoniazid conjugated magnetic nanoparticles loaded with Amphotericin B as a potent Antiamoebic agent against Acanthamoeba castellanii. Antibiotics 2020, 9, 276. [Google Scholar] [CrossRef] [PubMed]
  94. Peter, S.; Mathews, M.M.; Saju, F.; Paul, S. Development, Optimization and In Vitro Characterization of Eudragit-Ganciclovir Nanosuspension or Treating Herpes Simplex Keratitis. J. Pharm. Innov. 2023, 18, 1328–1337. [Google Scholar] [CrossRef]
  95. Matthews, T.; Boehme, R. Antiviral activity and mechanism of action of ganciclovir. Rev. Infect. Dis. 1988, 10 (Suppl. 3), S490–S494. [Google Scholar] [CrossRef] [PubMed]
  96. Kaul, S.; Nagaich, U.; Verma, N. Preclinical assessment of nanostructured liquid crystalline particles for the management of bacterial keratitis: In vivo and pharmacokinetics study. Drug Deliv. Transl. Res. 2022, 12, 1719–1737. [Google Scholar] [CrossRef] [PubMed]
  97. Saravolatz, L.D.; Johnson, L.B.; Kauffman, C.A. Voriconazole: A New Triazole Antifungal Agent. Clin. Infect. Dis. 2003, 36, 630–637. [Google Scholar] [CrossRef]
  98. Silva, L.D.; Arrúa, E.C.; Pereira, D.A.; Fraga, C.M.; Costa, T.L.d.; Hemphill, A.; Salomon, C.J.; Vinaud, M.C. Elucidating the influence of praziquantel nanosuspensions on the in vivo metabolism of Taenia crassiceps cysticerci. Acta Trop. 2016, 161, 100–105. [Google Scholar] [CrossRef]
  99. Youssef, A.A.A.; Cai, C.; Dudhipala, N.; Majumdar, S. Design of Topical Ocular Ciprofloxacin Nanoemulsion for the Management of Bacterial Keratitis. Pharmaceuticals 2021, 14, 210. [Google Scholar] [CrossRef] [PubMed]
  100. Yang, J.; Liang, Z.; Lu, P.; Song, F.; Zhang, Z.; Zhou, T.; Li, J.; Zhang, J. Development of a Luliconazole Nanoemulsion as a Prospective Ophthalmic Delivery System for the Treatment of Fungal Keratitis: In Vitro and In Vivo Evaluation. Pharmaceutics 2022, 14, 2052. [Google Scholar] [CrossRef]
  101. Prajapat, V.M.; Aalhate, M.; Sriram, A.; Mahajan, S.; Maji, I.; Gupta, U.; Kumari, D.; Singh, K.; Kalia, N.P.; Dua, K.; et al. Amphotericin B loaded nanoemulsion: Optimization, characterization and in-vitro activity against L. donovani promastigotes. Parasitol. Int. 2024, 100, 102848. [Google Scholar] [CrossRef]
  102. Nasr, M.; Saber, S.; Bazeed, A.Y.; Ramadan, H.A.; Ebada, A.; Ciorba, A.L.; Cavalu, S.; Elagamy, H.I. Advantages of Cubosomal Formulation for Gatifloxacin Delivery in the Treatment of Bacterial Keratitis: In Vitro and In Vivo Approach Using Clinical Isolate of Methicillin-Resistant Staphylococcus aureus. Materials 2022, 15, 3374. [Google Scholar] [CrossRef] [PubMed]
  103. Kennedy, S.M.; Deshpande, P.; Gallagher, A.G.; Horsburgh, M.J.; Allison, H.E.; Kaye, S.B.; Wellings, D.A.; Williams, R.L. Amoebicidal Activity of Poly-Epsilon-Lysine Functionalized Hydrogels. Investig. Ophthalmol. Vis. Sci. 2022, 63, 11. [Google Scholar] [CrossRef]
  104. Hussain, A.; Majumdar, A.; Malviya, N.; Dhere, M. Formulation and Evaluation of Ophthalmic Novel In-Situ Gel Containing Acyclovir for the Treatment of Herpes Simplex Keratitis. Res. J. Pharm. Technol. 2022, 15, 3747–3750. [Google Scholar] [CrossRef]
  105. Abbas, M.N.; Khan, S.A.; Sadozai, S.K.; Khalil, I.A.; Anter, A.; Fouly, M.E.; Osman, A.H.; Kazi, M. Nanoparticles loaded thermoresponsive in situ gel for ocular antibiotic delivery against bacterial keratitis. Polymers 2022, 14, 1135. [Google Scholar] [CrossRef] [PubMed]
  106. Okur, N.Ü.; Yozgatlı, V.; Okur, M.E.; Yoltaş, A.; Siafaka, P.I. Improving therapeutic efficacy of voriconazole against fungal keratitis: Thermo-sensitive in situ gels as ophthalmic drug carriers. J. Drug Deliv. Sci. Technol. 2019, 49, 323–333. [Google Scholar] [CrossRef]
  107. Büchele, M.L.C.; Filippin-Monteiro, F.B.; de Lima, B.; Camargo, C.d.J.; Restrepo, J.A.S.; Souza, L.C.; Creczynski-Pasa, T.B.; Caumo, K.S. Super aggregated amphotericin B with a thermoreversible in situ gelling ophthalmic system for amoebic keratitis treatment. Acta Trop. 2021, 224, 106144. [Google Scholar] [CrossRef]
  108. Dutta, D.; Vijay, A.K.; Kumar, N.; Willcox, M.D.P. Melimine-Coated Antimicrobial Contact Lenses Reduce Microbial Keratitis in an Animal Model. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5616–5624. [Google Scholar] [CrossRef]
  109. Huang, J.-F.; Zhong, J.; Chen, G.-P.; Lin, Z.-T.; Deng, Y.; Liu, Y.-L.; Cao, P.-Y.; Wang, B.; Wei, Y.; Wu, T.; et al. A Hydrogel-Based Hybrid Theranostic Contact Lens for Fungal Keratitis. ACS Nano 2016, 10, 6464–6473. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, L.; Kuang, L.; Ross, A.E.; Farhat, W.; Boychev, N.; Sharfi, S.; Kanu, L.N.; Liu, L.; Kohane, D.S.; Ciolino, J.B. Topical Sustained Delivery of Miltefosine Via Drug-Eluting Contact Lenses to Treat Acanthamoeba Keratitis. Pharmaceutics 2022, 14, 2750. [Google Scholar] [CrossRef]
  111. Kong, X.; Jia, Y.; Wang, H.; Li, R.; Li, C.; Cheng, S.; Chen, T.; Mai, Y.; Nie, Y.; Deng, Y. Effective Treatment of Haemophilus influenzae-Induced Bacterial Conjunctivitis by a Bioadhesive Nanoparticle Reticulate Structure. ACS Appl. Mater. Interfaces 2023, 15, 22892–22902. [Google Scholar] [CrossRef]
  112. Sipos, B.; Budai-Szűcs, M.; Kókai, D.; Orosz, L.; Burián, K.; Csorba, A.; Nagy, Z.Z.; Balogh, G.T.; Csóka, I.; Katona, G. Erythromycin-loaded polymeric micelles: In situ gel development, in vitro and ex vivo ocular investigations. Eur. J. Pharm. Biopharm. 2022, 180, 81–90. [Google Scholar] [CrossRef]
  113. Hosoya, K.; Lee, V.H.; Kim, K.J. Roles of the conjunctiva in ocular drug delivery: A review of conjunctival transport mechanisms and their regulation. Eur. J. Pharm. Biopharm. 2005, 60, 227–240. [Google Scholar] [CrossRef] [PubMed]
  114. De Gaetano, F.; Marino, A.; Marchetta, A.; Bongiorno, C.; Zagami, R.; Cristiano, M.C.; Paolino, D.; Pistarà, V.; Ventura, C.A. Development of Chitosan/Cyclodextrin Nanospheres for Levofloxacin Ocular Delivery. Pharmaceutics 2021, 13, 1293. [Google Scholar] [CrossRef] [PubMed]
  115. Marchegiani, A.; Magagnini, M.; Cerquetella, M.; Troiano, P.; Franchini, I.; Franchini, A.; Scapagnini, G.; Spaterna, A. Preoperative topical liposomal ozone dispersion to reduce bacterial colonization in conjunctival sac and periocular skin: Preliminary study in dogs. Exp. Eye Res. 2019, 189, 107848. [Google Scholar] [CrossRef] [PubMed]
  116. Grandi, G.; Cavallo, R.; Zanotto, E.; Cipriani, R.; Panico, C.; Protti, R.; Scapagnini, G.; Davinelli, S.; Costagliola, C. In vitro antimicrobial activity of ozonated oil in liposome eyedrop against multidrug-resistant bacteria. Open Med. 2022; 17, 1057–1063. [Google Scholar] [CrossRef]
  117. Calabretta, M.K.; Kumar, A.; McDermott, A.M.; Cai, C. Antibacterial activities of poly(amidoamine) dendrimers terminated with amino and poly(ethylene glycol) groups. Biomacromolecules 2007, 8, 1807–1811. [Google Scholar] [CrossRef] [PubMed]
  118. Bhalerao, H.; Koteshwara, K.; Chandran, S. Levofloxacin hemihydrate in situ gelling ophthalmic solution: Formulation optimization and in vitro and in vivo evaluation. AAPS PharmSciTech 2019, 20, 1–12. [Google Scholar] [CrossRef]
  119. Radhika, M.; Mithal, K.; Bawdekar, A.; Dave, V.; Jindal, A.; Relhan, N.; Albini, T.; Pathengay, A.; Flynn, H.W. Pharmacokinetics of intravitreal antibiotics in endophthalmitis. J. Ophthalmic Inflamm. Infect. 2014, 4, 22. [Google Scholar] [CrossRef]
  120. Jounaki, K.; Makhmalzadeh, B.S.; Feghhi, M.; Heidarian, A. Topical ocular delivery of vancomycin loaded cationic lipid nanocarriers as a promising and non-invasive alternative approach to intravitreal injection for enhanced bacterial endophthalmitis management. Eur. J. Pharm. Sci. 2021, 167, 105991. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, K.R.; Peyman, G.A.; Khoobehi, B. Efficacy of liposome-bound amphotericin B for the treatment of experimental fungal endophthalmitis in rabbits. Investig. Ophthalmol. Vis. Sci. 1989, 30, 1527–1534. [Google Scholar]
  122. Gupta, S.K.; Dhingra, N.; Velpandian, T.; Jaiswal, J. Efficacy of fluconazole and liposome entrapped fluconazole for C. albicans induced experimental mycotic endophthalmitis in rabbit eyes. Acta Ophthalmol. Scand. 2000, 78, 448–450. [Google Scholar] [CrossRef]
  123. Gupta, S.K.; Velpandian, T.; Dhingra, N.; Jaiswal, J. Intravitreal Pharmacokinetics of Plain and Liposome-Entrapped Fluconazole in Rabbit Eyes. J. Ocul. Pharmacol. Ther. 2000, 16, 511–518. [Google Scholar] [CrossRef]
  124. Velpandian, T.; Narayanan, K.; Nag, T.C.; Ravi, A.K.; Gupta, S.K. Retinal toxicity of intravitreally injected plain and liposome formulation of fluconazole in rabbit eye. Indian J. Ophthalmol. 2006, 54, 237–240. [Google Scholar] [CrossRef]
  125. Durairaj, C.; Kadam, R.S.; Chandler, J.W.; Hutcherson, S.L.; Kompella, U.B. Nanosized dendritic polyguanidilyated translocators for enhanced solubility, permeability, and delivery of gatifloxacin. Investig. Ophthalmol. Vis. Sci. 2010, 51, 5804–5816. [Google Scholar] [CrossRef] [PubMed]
  126. Eram, F. In-vivo Evaluation and Characterization of Novel In-Situ Gelling System as Controlled Delivery System Containing Ciprofloxacin for Ocular Drug Delivery. J. Drug Deliv. Ther. 2020, 10, 32–39. [Google Scholar] [CrossRef]
  127. Tsuchiya, Y.; Kobayakawa, S.; Tsuji, A.; Tochikubo, T. Preventive effect against post-cataract endophthalmitis: Drug delivery intraocular lens versus intracameral antibiotics. Curr. Eye Res. 2008, 33, 868–875. [Google Scholar] [CrossRef] [PubMed]
  128. Narayana, S.; Ahmed, M.G.; Gowda, B.H.J.; Shetty, P.K.; Nasrine, A.; Thriveni, M.; Noushida, N.; Sanjana, A. Recent advances in ocular drug delivery systems and targeting VEGF receptors for management of ocular angiogenesis: A comprehensive review. Future J. Pharm. Sci. 2021, 7, 186. [Google Scholar] [CrossRef]
  129. Tang, B.; Wang, Q.; Zhang, G.; Zhang, A.; Zhu, L.; Zhao, R.; Gu, H.; Meng, J.; Zhang, J.; Fang, G. OCTN2- and ATB0,+-targeted nanoemulsions for improving ocular drug delivery. J. Nanobiotechnol. 2024, 22, 130. [Google Scholar] [CrossRef] [PubMed]
  130. Zhong, Z.; Cai, L.; Li, C. Characterization and targeting ability evaluation of cell-penetrating peptide LyP-1 modified alginate-based nanoparticles. RSC Adv. 2020, 10, 32443–32449. [Google Scholar] [CrossRef] [PubMed]
  131. Amit, C.; Muralikumar, S.; Janaki, S.; Lakshmipathy, M.; Therese, K.L.; Umashankar, V.; Padmanabhan, P.; Narayanan, J. Designing and enhancing the antifungal activity of corneal specific cell penetrating peptide using gelatin hydrogel delivery system. Int. J. Nanomed. 2019, 14, 605–622. [Google Scholar] [CrossRef]
  132. Chahud, F.; Ramalho, L.N.; Ramalho, F.S.; Haddad, A.; Roque-Barreira, M.C. The lectin KM+ induces corneal epithelial wound healing in rabbits. Int. J. Exp. Pathol. 2009, 90, 166–173. [Google Scholar] [CrossRef]
  133. García-Posadas, L.; Contreras-Ruiz, L.; López-García, A.; Villarón Álvarez, S.; Maldonado, M.J.; Diebold, Y. Hyaluronan receptors in the human ocular surface: A descriptive and comparative study of RHAMM and CD44 in tissues, cell lines and freshly collected samples. Histochem. Cell Biol. 2012, 137, 165–176. [Google Scholar] [CrossRef]
  134. Kompella, U.B.; Sundaram, S.; Raghava, S.; Escobar, E.R. Luteinizing hormone-releasing hormone agonist and transferrin functionalizations enhance nanoparticle delivery in a novel bovine ex vivo eye model. Mol. Vis. 2006, 12, 1185–1198. [Google Scholar] [PubMed]
  135. Qaddoumi, M.; Lee, V.H.L. Lectins as Endocytic Ligands: An Assessment of Lectin Binding and Uptake to Rabbit Conjunctival Epithelial Cells. Pharm. Res. 2004, 21, 1160–1166. [Google Scholar] [CrossRef] [PubMed]
  136. Miller, F.C.; Coburn, P.S.; Huzzatul, M.M.; LaGrow, A.L.; Livingston, E.; Callegan, M.C. Targets of immunomodulation in bacterial endophthalmitis. Prog. Retin. Eye Res. 2019, 73, 100763. [Google Scholar] [CrossRef]
  137. Bertino Jr, J.S. Impact of antibiotic resistance in the management of ocular infections: The role of current and future antibiotics. Clin. Ophthalmol. 2009, 3, 507–521. [Google Scholar] [CrossRef] [PubMed]
  138. Scotty, N.C.; Brooks, D.E.; Rose, C.D.S. In vitro efficacy of an ophthalmic drug combination against corneal pathogens of horses. Am. J. Vet. Res. 2008, 69, 101–107. [Google Scholar] [CrossRef]
  139. Cai, J.; Yang, C.; Wei, Q.; Lian, H.; An, L.; Zhang, R. Natamycin versus natamycin combined with voriconazole in the treatment of fungal keratitis. Pak. J. Med. Sci. 2023, 39, 775. [Google Scholar] [CrossRef]
  140. Cheng, Y.-H.; Chang, Y.-F.; Ko, Y.-C.; Liu, C.J.-l. Development of a dual delivery of levofloxacin and prednisolone acetate via PLGA nanoparticles/thermosensitive chitosan-based hydrogel for postoperative management: An in-vitro and ex-vivo study. Int. J. Biol. Macromol. 2021, 180, 365–374. [Google Scholar] [CrossRef] [PubMed]
  141. Topete, A.; Serro, A.P.; Saramago, B. Dual drug delivery from intraocular lens material for prophylaxis of endophthalmitis in cataract surgery. Int. J. Pharm. 2019, 558, 43–52. [Google Scholar] [CrossRef]
  142. Topete, A.; Tang, J.; Ding, X.; Filipe, H.P.; Saraiva, J.A.; Serro, A.P.; Lin, Q.; Saramago, B. Dual drug delivery from hydrophobic and hydrophilic intraocular lenses: In-vitro and in-vivo studies. J. Control. Release 2020, 326, 245–255. [Google Scholar] [CrossRef]
  143. Yu, A.; Hu, Y.; Ma, X.; Mo, L.; Pan, M.; Bi, X.; Wu, Y.; Wang, J.; Li, X. Sequential drug release of co-assembled supramolecular hydrogel as synergistic therapy against Staphylococcus aureus endophthalmitis. Chem. Eng. J. 2022, 427, 130979. [Google Scholar] [CrossRef]
  144. Shih, M.-H.; Huang, F.-C. Effects of Photodynamic Therapy on Rapidly Growing Nontuberculous Mycobacteria Keratitis. Investig. Ophthalmol. Vis. Sci. 2011, 52, 223–229. [Google Scholar] [CrossRef] [PubMed]
  145. Martinez, J.D.; Arboleda, A.; Naranjo, A.; Aguilar, M.C.; Durkee, H.; Monsalve, P.; Dubovy, S.R.; Donaldson, K.E.; Miller, D.; Amescua, G.; et al. Long-term outcomes of riboflavin photodynamic antimicrobial therapy as a treatment for infectious keratitis. Am. J. Ophthalmol. Case Rep. 2019, 15, 100481. [Google Scholar] [CrossRef] [PubMed]
  146. Nguyen, D.D.; Lai, J.-Y. Advancing the stimuli response of polymer-based drug delivery systems for ocular disease treatment. Polym. Chem. 2020, 11, 6988–7008. [Google Scholar] [CrossRef]
  147. Lin, X.; Wu, X.; Chen, X.; Wang, B.; Xu, W. Intellective and stimuli-responsive drug delivery systems in eyes. Int. J. Pharm. 2021, 602, 120591. [Google Scholar] [CrossRef]
  148. Han, H.; Gao, Y.; Chai, M.; Zhang, X.; Liu, S.; Huang, Y.; Jin, Q.; Grzybowski, A.; Ji, J.; Yao, K. Biofilm microenvironment activated supramolecular nanoparticles for enhanced photodynamic therapy of bacterial keratitis. J. Control. Release 2020, 327, 676–687. [Google Scholar] [CrossRef]
  149. Kesavan, K.; Kant, S.; Pandit, J.K. Therapeutic effectiveness in the treatment of experimental bacterial keratitis with ion-activated mucoadhesive hydrogel. Ocul. Immunol. Inflamm. 2016, 24, 489–492. [Google Scholar] [CrossRef] [PubMed]
  150. Díaz-Tomé, V.; Luaces-Rodríguez, A.; Silva-Rodríguez, J.; Blanco-Dorado, S.; García-Quintanilla, L.; Llovo-Taboada, J.; Blanco-Méndez, J.; García-Otero, X.; Varela-Fernández, R.; Herranz, M.; et al. Ophthalmic Econazole Hydrogels for the Treatment of Fungal Keratitis. J. Pharm. Sci. 2018, 107, 1342–1351. [Google Scholar] [CrossRef] [PubMed]
  151. Yu, J.; Xu, H.; Wei, J.; Niu, L.; Zhu, H.; Jiang, C. Bacteria-Targeting Nanoparticles with ROS-Responsive Antibiotic Release to Eradicate Biofilms and Drug-Resistant Bacteria in Endophthalmitis. Int. J. Nanomed. 2024, 19, 2939–2956. [Google Scholar] [CrossRef]
  152. Li, J.; Li, Z.; Liang, Z.; Han, L.; Feng, H.; He, S.; Zhang, J. Fabrication of a drug delivery system that enhances antifungal drug corneal penetration. Drug Deliv. 2018, 25, 938–949. [Google Scholar] [CrossRef]
  153. Alambiaga-Caravaca, A.M.; Calatayud-Pascual, M.A.; Rodilla, V.; Concheiro, A.; López-Castellano, A.; Alvarez-Lorenzo, C. Micelles of Progesterone for Topical Eye Administration: Interspecies and Intertissues Differences in Ex Vivo Ocular Permeability. Pharmaceutics 2020, 12, 702. [Google Scholar] [CrossRef]
  154. Mahaling, B.; Sinha, N.R.; Sokupa, S.; Addi, U.R.; Mohan, R.R.; Chaurasia, S.S. Mustard gas exposure instigates retinal Müller cell gliosis. Exp. Eye Res. 2023, 230, 109461. [Google Scholar] [CrossRef]
  155. Draize, J.H. Methods for the study of irritation and toxicity of substances applied topically to the skin and the mucous membranes. J. Pharmacol. Exp. Ther. 1944, 82, 377–390. [Google Scholar]
  156. Mehra, N.K.; Cai, D.; Kuo, L.; Hein, T.; Palakurthi, S. Safety and toxicity of nanomaterials for ocular drug delivery applications. Nanotoxicology 2016, 10, 836–860. [Google Scholar] [CrossRef]
  157. Barile, F.A. Validating and troubleshooting ocular in vitro toxicology tests. J. Pharmacol. Toxicol. Methods 2010, 61, 136–145. [Google Scholar] [CrossRef] [PubMed]
  158. Lin, M.; Zou, R.; Shi, H.; Yu, S.; Li, X.; Guo, R.; Yan, L.; Li, G.; Liu, Y.; Dai, L. Ocular biocompatibility evaluation of hydroxyl-functionalized graphene. Mater. Sci. Eng. C 2015, 50, 300–308. [Google Scholar] [CrossRef] [PubMed]
  159. Lim, R.R.; Mahaling, B.; Tan, A.; Mehta, M.; Kaur, C.; Hunziker, W.; Kim, J.E.; Barathi, V.A.; Ghosh, A.; Chaurasia, S.S. ITF2357 regulates NF-κB signaling pathway to protect barrier integrity in retinal pigment epithelial cells. FASEB J. 2024, 38, e23512. [Google Scholar] [CrossRef]
  160. Mahaling, B.; Verma, M.; Mishra, G.; Chaudhuri, S.; Dutta, D.; Sivakumar, S. Fate of GdF3 nanoparticles-loaded PEGylated carbon capsules inside mice model: A step toward clinical application. Nanotoxicology 2020, 14, 577–594. [Google Scholar] [CrossRef] [PubMed]
  161. Alenius, H.; Catalán, J.; Lindberg, H.; Norppa, H.; Palomäki, J.; Savolainen, K. Chapter 3—Nanomaterials and Human Health. In Handbook of Nanosafety; Vogel, U., Savolainen, K., Wu, Q., van Tongeren, M., Brouwer, D., Berges, M., Eds.; Academic Press: San Diego, CA, USA, 2014; pp. 59–133. [Google Scholar]
  162. Patil, A.; Lakhani, P.; Majumdar, S. Current perspectives on natamycin in ocular fungal infections. J. Drug Deliv. Sci. Technol. 2017, 41, 206–212. [Google Scholar] [CrossRef]
  163. Chou, T.Y.; Hong, B.Y. Ganciclovir ophthalmic gel 0.15% for the treatment of acute herpetic keratitis: Background, effectiveness, tolerability, safety, and future applications. Ther. Clin. Risk Manag. 2014, 10, 665–681. [Google Scholar] [CrossRef] [PubMed]
  164. Novack, G.D. Eyes on new product development. J. Ocul. Pharmacol. Ther. 2013, 29, 785. [Google Scholar] [CrossRef] [PubMed]
  165. Kang, C.; Keam, S.J.; Shirley, M.; Syed, Y.Y. Loteprednol Etabonate (Submicron) Ophthalmic Gel 0.38%: A Review in Post-Operative Inflammation and Pain Following Ocular Surgery. Clin. Drug Investig. 2020, 40, 387–394. [Google Scholar] [CrossRef]
  166. Minigh, J. Fluorometholone. In xPharm: The Comprehensive Pharmacology Reference; Enna, S.J., Bylund, D.B., Eds.; Elsevier: New York, NY, USA, 2007; pp. 1–4. [Google Scholar]
Figure 1. Schematic of pathophysiology of infectious keratitis. (A) bacterial keratitis, (B) fungal keratitis, (C) viral keratitis and (D) parasitic keratitis. In case of bacterial and fungal infection, microorganisms enter the corneal epithelial cell through injury or trauma. In response to infection and injury, the immune cells infiltrate, and cytokines and chemokines are subsequently secreted by the injured epithelial cells and immune cells. However, in case of viral infection, viruses bind the cell surface receptors of corneal epithelial cells and invade the stroma. Parasitic pathogens like Acanthamoeba damage the cornea by entering through various means, form cysts, and cause stromal dissolution leading to irreversible corneal damage; 1 corneal epithelial barrier breakdown, 2 cellular infiltration, 3 cytokines and chemokines secretion, 4 thinning and breakdown of Bowman’s layer, 5 corneal epithelial cell damage, 6 stromal dissolution.
Figure 1. Schematic of pathophysiology of infectious keratitis. (A) bacterial keratitis, (B) fungal keratitis, (C) viral keratitis and (D) parasitic keratitis. In case of bacterial and fungal infection, microorganisms enter the corneal epithelial cell through injury or trauma. In response to infection and injury, the immune cells infiltrate, and cytokines and chemokines are subsequently secreted by the injured epithelial cells and immune cells. However, in case of viral infection, viruses bind the cell surface receptors of corneal epithelial cells and invade the stroma. Parasitic pathogens like Acanthamoeba damage the cornea by entering through various means, form cysts, and cause stromal dissolution leading to irreversible corneal damage; 1 corneal epithelial barrier breakdown, 2 cellular infiltration, 3 cytokines and chemokines secretion, 4 thinning and breakdown of Bowman’s layer, 5 corneal epithelial cell damage, 6 stromal dissolution.
Jnt 05 00010 g001
Figure 2. Schematic of pathophysiology of infectious conjunctivitis and endophthalmitis. (A) Bacterial conjunctivitis: It starts with the colonization of bacteria on the of disrupted epithelial cells by injury or trauma. In response to infection and injury, the immune cells infiltrate and cytokines and chemokines are subsequently secreted by the injured epithelial cells and immune cells. (B) Viral conjunctivitis: Virus binds the cell surface receptors and internalizes conjunctival epithelial cells and invades stroma. Goblet cells are also involved in the internalization in viral conjunctivitis. Both the innate and adaptive immune responses get activated against viral infection and secret cytokines and chemokines. (C) Bacterial endophthalmitis, and (D) Fungal endophthalmitis: Both bacterial and fungal endophthalmitis can originate from exogenous or endogenous sources of microorganisms. Exogenous endophthalmitis occurs when organisms enter the eye through intraocular surgery, penetrating trauma, or by spreading from adjacent tissues. In contrast, endogenous endophthalmitis occurs when infectious agents are carried to the eye through the bloodstream from a distant site of infection. This leads to blood retinal barrier breakdown, immune cells infiltration and microglia activation. The infiltered immune cells and activated microglia secret cytokines and chemokines and cause inflammation and neurodegeneration; 1 represents conjunctival epithelial barrier breakdown, 2 represents cellular infiltration, 3 represents cytokines and chemokines secretion, 4 represents activation of lymphocyte, 5 represents blood-retinal barrier breakdown, 6 represents activation of microglia.
Figure 2. Schematic of pathophysiology of infectious conjunctivitis and endophthalmitis. (A) Bacterial conjunctivitis: It starts with the colonization of bacteria on the of disrupted epithelial cells by injury or trauma. In response to infection and injury, the immune cells infiltrate and cytokines and chemokines are subsequently secreted by the injured epithelial cells and immune cells. (B) Viral conjunctivitis: Virus binds the cell surface receptors and internalizes conjunctival epithelial cells and invades stroma. Goblet cells are also involved in the internalization in viral conjunctivitis. Both the innate and adaptive immune responses get activated against viral infection and secret cytokines and chemokines. (C) Bacterial endophthalmitis, and (D) Fungal endophthalmitis: Both bacterial and fungal endophthalmitis can originate from exogenous or endogenous sources of microorganisms. Exogenous endophthalmitis occurs when organisms enter the eye through intraocular surgery, penetrating trauma, or by spreading from adjacent tissues. In contrast, endogenous endophthalmitis occurs when infectious agents are carried to the eye through the bloodstream from a distant site of infection. This leads to blood retinal barrier breakdown, immune cells infiltration and microglia activation. The infiltered immune cells and activated microglia secret cytokines and chemokines and cause inflammation and neurodegeneration; 1 represents conjunctival epithelial barrier breakdown, 2 represents cellular infiltration, 3 represents cytokines and chemokines secretion, 4 represents activation of lymphocyte, 5 represents blood-retinal barrier breakdown, 6 represents activation of microglia.
Jnt 05 00010 g002
Figure 3. Schematic representation of stimuli-responsive drug delivery systems (DDSs) for infectious ocular diseases. (A) Thermo-responsive DDSs. It offers a promising approach for treating ocular infections by providing controlled drug release in response to temperature changes. When exposed to increased temperatures, these systems either swell or shrink, triggering the release of the drug. (B) Enzyme-responsive DDSs. This system can be developed by targeting the enzymes present in the eye such as matrix metalloproteinases (MMPs) in tissues, hyaluronidase in the vitreous, and lysozyme and esterase in tears. (C) pH-responsive DDSs. It offers a promising approach for treating ocular infections by reacting to pH changes in the eye tissues. The eye can tolerate a pH range of 4 to 10, with the normal pH of tears being around 7.4. Natural or synthetic pH-responsive polymers are engineered to respond effectively within this ocular environment. Nanoparticles can be designed for high encapsulation efficiency at pH levels below 6.9 or above 8, releasing the drug optimally at a pH of 7.4. (D) Reactive oxygen species (ROS)-responsive drug delivery system. ROS play a crucial role in infection and inflammation and can serve as a trigger for drug release in ocular DDSs. The nanocarrier can be engineered to penetrate ocular barriers and release drugs in response at high ROS levels.
Figure 3. Schematic representation of stimuli-responsive drug delivery systems (DDSs) for infectious ocular diseases. (A) Thermo-responsive DDSs. It offers a promising approach for treating ocular infections by providing controlled drug release in response to temperature changes. When exposed to increased temperatures, these systems either swell or shrink, triggering the release of the drug. (B) Enzyme-responsive DDSs. This system can be developed by targeting the enzymes present in the eye such as matrix metalloproteinases (MMPs) in tissues, hyaluronidase in the vitreous, and lysozyme and esterase in tears. (C) pH-responsive DDSs. It offers a promising approach for treating ocular infections by reacting to pH changes in the eye tissues. The eye can tolerate a pH range of 4 to 10, with the normal pH of tears being around 7.4. Natural or synthetic pH-responsive polymers are engineered to respond effectively within this ocular environment. Nanoparticles can be designed for high encapsulation efficiency at pH levels below 6.9 or above 8, releasing the drug optimally at a pH of 7.4. (D) Reactive oxygen species (ROS)-responsive drug delivery system. ROS play a crucial role in infection and inflammation and can serve as a trigger for drug release in ocular DDSs. The nanocarrier can be engineered to penetrate ocular barriers and release drugs in response at high ROS levels.
Jnt 05 00010 g003
Figure 4. Schematic representation of ocular biocompatibility study. (A) In vivo biocompatibility study in rabbit [152,153]. The Draize test is an animal-based (rabbit) method used to evaluate the potential of chemicals to cause eye irritation. Additionally, the histology of the ocular tissue provides changes in structural integrity of the ocular tissue after administration of ocular formulations. (B) In vivo biocompatibility study in rodents. It could be used to evaluate the biocompatibility of ocular formulation after administration to the eye. Different techniques can be used to evaluate the structural and functional integrity of the eye. (i) Electroretinogram (ERG) is used to evaluate retinal function. (ii) Optokinetic tracking (OKT) is used to study visual acuity. (iii) Fluorescein angiography (FA) is an eye examination that utilizes a fluorescent dye and a specialized camera to assess blood circulation in the retina and choroid. (iv) The change in intraocular pressure due to administration of ocular formulation can be measured by a tonometer. (v) Intra ocular ultrasound is a non-invasive test that uses high-frequency sound waves to create an image of the eye. (C) In vitro biocompatibility study in ocular cells. It could be used to evaluate the toxic effects of a substance on cultured cells. Various techniques are employed to assess cell viability, the production of reactive oxygen species (ROS), and any genetic material damage resulting from exposure to new formulations. (D) In vitro hemocompatibility study. It can be used to assess the safety of the formulation for contact with blood. In vitro hemocompatibility studies can include a variety of evaluation methods, such as hemolysis, blood clotting, hematology, and platelet and leukocyte activation [26,154]. (E) Ocular tolerance test (HET-CAM) [153]. HET-CAM test can be used to evaluate the irritation potential of ocular formulations by observing the damage it causes to the chorioallantoic membrane. The negative control is a 0.9% NaCl solution, Positive control is a 0.1N NaOH solution. Bleeding, clotting, and vascular melting are used as parameters for a vascular irritation study.
Figure 4. Schematic representation of ocular biocompatibility study. (A) In vivo biocompatibility study in rabbit [152,153]. The Draize test is an animal-based (rabbit) method used to evaluate the potential of chemicals to cause eye irritation. Additionally, the histology of the ocular tissue provides changes in structural integrity of the ocular tissue after administration of ocular formulations. (B) In vivo biocompatibility study in rodents. It could be used to evaluate the biocompatibility of ocular formulation after administration to the eye. Different techniques can be used to evaluate the structural and functional integrity of the eye. (i) Electroretinogram (ERG) is used to evaluate retinal function. (ii) Optokinetic tracking (OKT) is used to study visual acuity. (iii) Fluorescein angiography (FA) is an eye examination that utilizes a fluorescent dye and a specialized camera to assess blood circulation in the retina and choroid. (iv) The change in intraocular pressure due to administration of ocular formulation can be measured by a tonometer. (v) Intra ocular ultrasound is a non-invasive test that uses high-frequency sound waves to create an image of the eye. (C) In vitro biocompatibility study in ocular cells. It could be used to evaluate the toxic effects of a substance on cultured cells. Various techniques are employed to assess cell viability, the production of reactive oxygen species (ROS), and any genetic material damage resulting from exposure to new formulations. (D) In vitro hemocompatibility study. It can be used to assess the safety of the formulation for contact with blood. In vitro hemocompatibility studies can include a variety of evaluation methods, such as hemolysis, blood clotting, hematology, and platelet and leukocyte activation [26,154]. (E) Ocular tolerance test (HET-CAM) [153]. HET-CAM test can be used to evaluate the irritation potential of ocular formulations by observing the damage it causes to the chorioallantoic membrane. The negative control is a 0.9% NaCl solution, Positive control is a 0.1N NaOH solution. Bleeding, clotting, and vascular melting are used as parameters for a vascular irritation study.
Jnt 05 00010 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mahaling, B.; Baruah, N.; Dinabandhu, A. Drug Delivery Systems for Infectious Eye Diseases: Advancements and Prospects. J. Nanotheranostics 2024, 5, 133-166. https://doi.org/10.3390/jnt5040010

AMA Style

Mahaling B, Baruah N, Dinabandhu A. Drug Delivery Systems for Infectious Eye Diseases: Advancements and Prospects. Journal of Nanotheranostics. 2024; 5(4):133-166. https://doi.org/10.3390/jnt5040010

Chicago/Turabian Style

Mahaling, Binapani, Namrata Baruah, and Aumreetam Dinabandhu. 2024. "Drug Delivery Systems for Infectious Eye Diseases: Advancements and Prospects" Journal of Nanotheranostics 5, no. 4: 133-166. https://doi.org/10.3390/jnt5040010

APA Style

Mahaling, B., Baruah, N., & Dinabandhu, A. (2024). Drug Delivery Systems for Infectious Eye Diseases: Advancements and Prospects. Journal of Nanotheranostics, 5(4), 133-166. https://doi.org/10.3390/jnt5040010

Article Metrics

Back to TopTop