Next Article in Journal
Meroterpenoids from Gongolaria abies-marina against Kinetoplastids: In Vitro Activity and Programmed Cell Death Study
Next Article in Special Issue
Baricitinib Lipid-Based Nanosystems as a Topical Alternative for Atopic Dermatitis Treatment
Previous Article in Journal
FOXO3-Activated circFGFBP1 Inhibits Extracellular Matrix Degradation and Nucleus Pulposus Cell Death via miR-9-5p/BMP2 Axis in Intervertebral Disc Degeneration In Vivo and In Vitro
Previous Article in Special Issue
Endogenous Lipid Carriers—Bench-to-Bedside Roadblocks in Production and Drug Loading of Exosomes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progress of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Ocular Drug Delivery Platforms

Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 55 Marin Drinov Str., 9000 Varna, Bulgaria
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(3), 474; https://doi.org/10.3390/ph16030474
Submission received: 14 February 2023 / Revised: 12 March 2023 / Accepted: 20 March 2023 / Published: 22 March 2023
(This article belongs to the Special Issue Current Insights on Lipid-Based Nanosystems 2023)

Abstract

:
Sufficient ocular bioavailability is often considered a challenge by the researchers, due to the complex structure of the eye and its protective physiological mechanisms. In addition, the low viscosity of the eye drops and the resulting short ocular residence time further contribute to the observed low drug concentration at the target site. Therefore, various drug delivery platforms are being developed to enhance ocular bioavailability, provide controlled and sustained drug release, reduce the number of applications, and maximize therapy outcomes. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) exhibit all these benefits, in addition to being biocompatible, biodegradable, and susceptible to sterilization and scale-up. Furthermore, their successive surface modification contributes to prolonged ocular residence time (by adding cationic compounds), enhanced penetration, and improved performance. The review highlights the salient characteristics of SLNs and NLCs concerning ocular drug delivery, and updates the research progress in this area.

1. Introduction

According to World Health Organization, the prevalence of eye conditions is expected to increase in the following years as a result of population aging, the associated rise of non-communicable diseases (diabetes, cardiovascular diseases), along with various lifestyle factors, such as an unhealthy diet, smoking, extensive usage of digital devices, etc. [1,2,3,4]. Furthermore, a recent analysis for the Global Burden of Disease Study forecasts that by 2050, around 474 million people will suffer from moderate to severe visual impairments, among which 61 million will develop complete blindness [5]. Although the human eye is one of the most accessible organs in terms of drug application, efficient ocular delivery is still a goal to be achieved. Possible explanations lie in the anatomical and physiological characteristics of the eyeball and its protective mechanisms, as well as in the technological properties of the ocular formulations [6]. According to location, the human eye may be distinguished into two segments: anterior, presented by the cornea, conjunctiva, iris, ciliary body, lens, and aqueous humor, and posterior, consisting of the sclera, choroid, retina, vitreous humor, and optic nerve [7,8]. The preferred route of administration in ophthalmology—topical instillation—provides the possibility for treatment of anterior segment diseases such as blepharitis, dry eye disease, conjunctivitis, ocular infections or injuries [9], however, reaching the posterior part of the eye and ensuring sufficient therapeutic concentration thereby is still a challenge. Eye drops, representing the majority of ophthalmic formulations, are relatively easy for self-administration, characterized by high patient approval, cost-effectiveness, and well-established formulation and manufacturing processes [10]. Their main limitations include their intrinsic low viscosity, a short ocular contact time, and the relatively large volume of applied drops, often leading to drug loss via physiological pathways [11,12,13].
Additionally, ocular defense mechanisms such as reflex blinking, tear turnover, nasolacrimal drainage, and static and dynamic anatomical barriers further hinder drug absorption, resulting in less than 5% of the instilled dose attaining deeper ocular tissues [14,15]. In ocular surface diseases, drug bioavailability may be partially improved through modulating the formulations’ viscosity, by including viscosity enhancers or using in situ gel-forming systems/semisolid dosage forms [16]. However, this strategy does not apply to posterior segment diseases. Unfortunately, diseases affecting the back part of the eye, e.g., age-related macular degeneration, diabetic retinopathy, and glaucoma, may often cause visual impairment or blindness unless treated efficiently [17,18]. The therapy of posterior segment eye diseases usually includes intravitreal injections, which enable drug delivery to the vitreous cavity. However, the invasive nature of this approach and the potential associated complications (e.g., endophthalmitis, retinal detachment) determine the low patient compliance [19,20]. Reaching the posterior segment via the peroral or intravenous route has also been associated with limited therapeutic success, due to the presence of blood–ocular barriers (the blood–retinal barrier, in particular), in addition to the potential risk of occurrence of side effects [21]. Altogether, these factors determine the necessity of further progress in the field of ocular delivery by improving the technological characteristics of conventional ophthalmic formulations, exploring advanced drug delivery systems, or combining both strategies.
Various nanoscale drug delivery systems, such as liposomes [22,23], niosomes [24,25], solid lipid/polymeric nanoparticles [26,27,28,29], nanostructured lipid carriers [30,31], nanomicelles [32,33], microemulsions [34,35], and dendrimers [36], have been successfully developed for ocular delivery purposes, and have been reported to achieve enhanced bioavailability, sustained and controlled drug release, and a reduction in the number of applications, as well as side effects. SLNs and NLCs raise great interest due to their excellent biocompatibility and tolerability, tunable physiochemical characteristics, and scaling-up capabilities [37,38,39]. Developed for the first in the 1990s by Professor Müller and Professor Gasco, SLNs represent a mixture of solids at ambient temperature and and lipids at physiological temperatures, dispersed in an aqueous phase containing surfactants [40,41]. Approximately 10 years later, a second generation of lipid nanoparticles was proposed—NLCs,—which additionally include liquid lipid(s) in their structure [42,43]. Both drug delivery systems are feasible carriers for hydrophilic and hydrophobic drugs. They are characterized by their long-term stability and favored uptake through biological membranes, owing to their lipid nature and nano dimensions [44,45]. The possibilities to impart mucoadhesiveness by surface coating with various polymers, or by incorporating them into semisolid/in situ gelling/formulations, further promotes their beneficial effects in ocular therapeutics.
The current review aimed to summarize the recent research progress of solid lipid nanoparticles and nanostructured lipid carriers in ocular delivery. In the first part, the anatomical and physiological features of the human eye and potential delivery routes have been discussed. The second part provides an overview of the specific characteristics of SLNs and NLCs, with respect to their compositions, suitable physicochemical properties tailored for effective ocular delivery, surface modification strategies, and sterilization feasibility. Recent advances in this area have also been outlined.

2. Eye Anatomy, Barriers and Routes in Ocular Drug Delivery

Generally, human eye structures are distinguished according to their location in the eyeball, where the eye is divided into two segments (anterior and posterior) (Figure 1A), or according to their functionalities, where it is divided into three different layers—an outer (fibrous), middle (vascular) and inner (neuronal) coat [46]. The outer layer (fibrous tunic) consists of the cornea (at its front) and sclera, occupying five-sixths of the coat [47]. Its main functions are related to maintaining the shape of the eyeball, and providing protection to the inner ocular tissues [48]. The middle layer, also referred to as uvea, is composed of the iris and the ciliary body (in the anterior), and the choroid, forming the posterior uvea (Figure 1) [49]. The retina represents the innermost layer, which is involved in the visual perception process by converting light energy into neuronal signals, which are transmitted to the visual cortex of the brain by the optic nerve [50,51].
For better perception, the anatomical and physiological features of the human eye will be discussed from the anterior to posterior segment.

2.1. Anterior Segment of the Eye

2.1.1. Tear Film

The tear film is the first hindrance for topically applied drugs, often referred to as a dynamic (physiological) ocular barrier (Figure 1B) due to its high turnover rate, (0.5–2.2 µL/min), determining a short ocular residence time, and limited drug penetration ability [9,52,53]. Spread onto the corneal and conjunctival epithelium, it provides a smooth and lubricated optical surface, prevents the occurrence of infections due to its antimicrobial compounds (lysozyme, lactoferrin, lipocalin), or by washing out foreign substances, and supplies oxygen and nutrients to the cornea [54]. Traditionally, the tear film is described as a three-layered structure—an outer lipid layer produced by the Meibomian glands, a middle aqueous layer, and an inner mucous layer secreted predominantly by the conjunctival goblet cells [55]. However, a more recent theory considers that the tear film consists of two layers—an outer lipid layer and an inner muco-aqueous, gel-like layer [55,56,57]. Regarding ocular delivery, both layers exhibit barrier functions, the lipid one for hydrophilic drugs and the muco-aqueous layer for hydrophobic drugs [58]. Other precorneal factors negatively influencing ocular bioavailability include drug binding with proteins/mucin in the tear film, as well as drug loss via nasolacrimal drainage [53]. The latter is affected by the volume of applied drops (larger volumes correspond to more significant loss) and the blink reflex [9,12].

2.1.2. Cornea

The cornea is the main route for drug absorption after topical instillation, often referred to as a static (anatomical) barrier (Figure 1B). It is a transparent, highly specialized, avascular structure comprising six layers: the corneal epithelium, Bowman’s layer, stroma, Dua’s layer, Descemet’s membrane, and endothelium [59,60]. Among these, the epithelium, the stroma, and the endothelium have a primary role in the drug/nanocarrier transport. Corneal epithelium is a five to seven-layered structure, composed of squamous, wing and basal cells [61]. Its lipophilic nature, and the existing intercellular tight junctions (zonula occludens) hinder the entry of hydrophilic substances and macromolecules [14,62]. Additionally, the presence of efflux transporters, such as breast cancer resistance protein (BCRP), multidrug resistance-associated proteins (MRPs), P-glycoprotein (P-gp), and enzymes (e.g., cytochrome P450), acting as metabolic barriers, may further decrease ocular drug bioavailability [58,63,64]. Beneath the epithelium is the stroma, which occupies approximately 90% of the corneal thickness [65]. It is a hydrophilic, gel-like structure made of collagen fibrils and mucopolysaccharides, and represents the main obstacle for the permeation of lipophilic compounds [66]. The corneal endothelium is a single layer composed of hexagonal-shaped cells involved in water transport towards the anterior chamber, as well as the maintenance of corneal transparency [67]. Unlike the epithelial layer, the endothelial junctions are considered “leaky” and enable the transport of macromolecules [11]. In general, drugs are transported across the cornea via transcellular (for lipophilic compounds) and paracellular (for hydrophilic molecules) pathways [68]. Factors affecting corneal absorption include a drug’s molecular weight (compounds up to 500 Da are able to permeate across the epithelium), lipophilicity (facilitated for lipophilic compounds; preferably log D values of 2–3), degree of ionization (non-ionized forms penetrate more easily), and the charge of the ionized species (facilitated penetration of cationic molecules) [21,69,70,71].

2.1.3. Conjunctiva

The conjunctiva is a transparent mucous membrane, which overlays the anterior ocular surface and the interior of the eyelids. It is involved in the production of mucus and the maintenance of the tear film, ensuring the lubrication of the eye, and also preventing the entrance of exogenous substances or microorganisms [53,72]. The conjunctiva may be divided into three areas: the bulbar conjunctiva, covering the anterior part of the sclera; the conjunctival fornices, forming the cul-de-sac; the palpebral conjunctiva located on the posterior eyelid’s surface [50]. Generally, the cul-de-sac is estimated to retain a volume of up to 30 µL—a capacity insufficient to preserve the entire volume of an applied drop (most often in the range of 40–70 µL), which leads to partial drug loss immediately after instillation [67]. The conjunctiva is considered to be more permeable when compared to the cornea, especially in terms of hydrophilic compounds, due to the wider intercellular spaces between the junctions in its structure, allowing for the passage of larger compounds (5000–10,000 Da), as well as owing to its bigger surface area. Nevertheless, conjunctival drug absorption is considered ineffective, mainly due to its high vascularity [71,73,74]. Conjunctival blood and lymph circulation functions as a dynamic barrier, leading to drug clearance and systemic absorption, hence the observed low drug concentration in the anterior chamber. Additionally, the existing transporters (amino acids transporters, P-gp) acting as efflux pumps further contribute to this process [63,75].

2.1.4. Iris

The iris is a circular, colored, contractile structure, which surrounds an aperture in its center (the pupil) (Figure 1A). It regulates the constriction or dilation of the pupil according to the light intensity, via parasympathetic/sympathetic activation, respectively [76]. It contains pigmented epithelial cells in its structure, enabling drug accumulation and altering its pharmacokinetics [77]. The melanin-containing cells in the eye (localized to the iris/ciliary body at the front and in the choroid/retinal pigment epithelium in the posterior) can bind drug molecules via electrostatic and van der Waals forces, as well as by charge interactions. The formed complex may be considered a “reservoir”, releasing drugs at a slow rate, therefore, it can also be used in a drug-targeting approach to achieve prolonged action in the corresponding (pigmented) ocular areas [78,79,80].

2.1.5. Ciliary Body

The ciliary body is part of the middle (vascular) layer in the eye and is involved in the maintenance of the shape of the lens via the ciliary muscle, and in the production of aqueous humor [53,81]. Furthermore, the ciliary epithelium and the endothelial cells of the iris blood vessels form the blood–aqueous barrier (BAB), which prevents molecules’ entrance from systemic circulation to the aqueous humor [82]. The tight junctions in its structure limit the paracellular transport of large hydrophilic molecules, unlike small lipophilic compounds, which can penetrate via the transcellular pathway, and are subsequently eliminated by the uveal blood flow and aqueous humor turnover [49,78,83,84]. Alternatively, the elimination of hydrophilic compounds from the anterior chamber is carried out solely by the aqueous humor through Schlemm’s canal, which determines their slower clearance [67,78].

2.1.6. Lens

The lens is located behind the iris and the pupil (Figure 1A), and is characterized by its transparent appearance, biconvex shape, great index of refraction, and high concentration of proteins in its structure (i.e., crystallins). Its main functions include light transmission and focusing it onto the retina to obtain a distinct image [85,86].

2.2. Posterior Segment of the Eye

The sclera, the choroid, and the retinal pigment epithelium (RPE) represent the posterior static ocular barriers used for drug delivery [63].

2.2.1. Sclera

The sclera is a white, dense tissue, made of collagen fibers (predominantly type I, and <5% type III) and proteoglycans [87]. The porous areas within the collagenous, aqueous medium determine the relatively easy passage of hydrophilic molecules when compared to hydrophobic ones. In addition to drugs’ lipo/hydrophilicity, other physicochemical characteristics, such as their charge, molecular weight, and molecular radius, also influence scleral permeability [19]. The proteoglycan matrix, negatively charged at physiological pH, hinders the permeation of positively charged compounds as a result of the electrostatic interactions in between [88]. Regarding the impact of molecular weight/radius, studies showed that molecules up to 70 kDa are able to permeate across the sclera [89], and there is an inverse relationship between radius and drug permeability—smaller molecules penetrate more easily [88].

2.2.2. Choroid

The choroid is a thin, vascularized, pigmented tissue, involved in the transport of nutrients and oxygen to the retina [90,91]. Concerning drug delivery, it may be considered as both a static and dynamic barrier (Figure 1B), the latter owing to its high blood flow rate, determining rapid drug elimination [7,92]. Choroidal blood vessels are characterized by fenestrated walls, which enable drugs to reach the extravascular space of the choroid. Still, their further distribution towards the retina is limited by the presence of the blood–retinal barrier (BRB) [14,78].

2.2.3. Retina

The retina is a thin, transparent tissue lining the inner ocular surface [50]. It is characterized by a complex structure—histologically, it can be divided into ten layers. The outermost layer, the retinal pigment epithelium, represents a significant barrier to ocular drug delivery, due to the existing tight junctions between the epithelial cells, hindering paracellular drug transport [93,94]. The retinal pigment epithelium participates in the formation of the blood–retinal barrier (the outer BRB), whereas the retinal capillary endothelial cells constitute the inner BRB [95].

2.2.4. Vitreous Body

The vitreous body is a clear, avascular gel-like substance occupying the majority of the eyeball (Figure 1A) [96]. It performs several important functions, including maintaining the shape of the eyeball, acting as a shock absorber, protecting the retina from mechanical stress, and participating in light transmission towards the retina [97]. The vitreous body may be also considered as an area for drug delivery to the posterior eye segment. Intravitreal permeation depends on drugs’ physicochemical characteristics, such as their charge (facilitated for negatively charged molecules, which do not interact electrostatically with the negatively charged vitreous humor constituents), size (small molecules diffuse easily), and lipophilicity (easier when compared to hydrophilic drugs). The last two parameters also influence drug clearance—larger and hydrophilic molecules are characterized by a longer half-life, due to their elimination via the anterior route (through the aqueous humor), in contrast to small lipophilic compounds, which are cleared via the posterior route (crossing the BRB) [19,21,69].

2.3. Alternative Routes of Ocular Delivery

The complex anatomical and physiological features of the eye elucidate the challenges in ocular drug delivery from a physiological point of view. To achieve higher therapeutic concentrations in the posterior segment, alternative routes of administration have been exploited, the most common of which are presented in Table 1. However, most of them (excluding the oral route) are invasive, and are not applicable by the patients themselves, therefore, research efforts are focused on the elaboration of advanced drug delivery platforms, aiming to improve drug bioavailability and therapeutic outcomes for both anterior and posterior eye segment diseases.

3. Feasibility of Lipid Nanoparticles in Ophthalmology

Lipid-based drug delivery systems, such as nanoemulsions, liposomes, niosomes, cubosomes, and lipid nanoparticles, have attracted an enormous scientific interest, due to their biocompatibility, biodegradability, and tolerability [108]. An excellent review summarizing the feasibility of all the aforementioned lipid-based nanocarriers in ophthalmology is provided here [109]. Emerging initially as an alternative to liposomes in terms of their superior physical stability, cost-effective process and materials, as well as being alternatives to polymeric nanoparticles, due to the absence of toxic degradation products, [37] SLNs have been explored as drug delivery systems for various routes of application—dermal [110,111], ocular [112,113], pulmonary [114], parenteral [115], nasal [116], and oral [117]. Another advantageous characteristic of the lipid nanocarriers is the possibility of encapsulating more than one therapeutic agent, leading to the elaboration of dual or multidrug lipid nanoparticles, characterized by a synergetic effect and improved therapeutic performance [118]. In ophthalmology, in particular, SLNs and the second-generation lipid particles—NLCs—are considered especially beneficial due to their ability to provide sustained drug release by acting as drug depot formulations, and enhance corneal penetration due to the corresponding activity of non-ionic surfactants included in their structure [119,120]. The latter may further contribute towards an improved ocular bioavailability, by opening the tight junctions between corneal epithelial cells, facilitating paracellular drug transport, and by inhibiting P-glycoprotein activity, limiting drug efflux [121,122,123].
The lipid nanoparticles’ transcorneal penetration mechanism has been studied by Nagai et al., according to which the process is implemented via energy-dependent endocytosis. The authors proposed three endocytosis pathways (clathrin-dependent, caveolae-dependent and macropinocytosis) as possible mechanisms for penetration of indomethacin-loaded nanoparticles, with an emphasis on the caveolae-dependent endocytosis [124]. Undoubtedly, nanoparticles’ permeation and internalization are highly affected by their physicochemical characteristics, such as size, size distribution pattern, zeta potential, and subsequent surface modification. Generally, nanoparticles up to 200 nm are reported to penetrate across the cornea [125]. In the case of periocular application, the excessive downsizing of their dimensions (e.g., ≈20 nm) may lead to their rapid clearance, as reported by Amritte et al. [126]. In their study Niamprem et al. investigated the penetration of fluorescent dye (Nile red)-loaded NLCs across porcine cornea, as a function of their size and surface modifications. According to the authors, NLCs with a size of 40 nm exhibited enhanced penetration when compared to larger (150 nm) nanoparticles.
Regarding their internalization, non-modified NLCs had a higher uptake in porcine corneal epithelial cells than PEG- and stearylamine-modified nanocarriers. The latter may be attributed to their superior mucoadhesive properties, arising from hydrogen boding between PEG molecules and mucin glycoproteins, or from ionic interactions between cationic stearylamine and anionic groups present in mucin regions [127].
Ocular drug delivery is also affected by the zeta potential of the nanocarriers. Positive values contribute to an increased ocular contact time, as a result of the occurred electrostatic interactions with the negatively charged corneal epithelium [125]. Regarding zeta potential’s impact on the colloidal stability of the nanocarriers, generally, absolute values of 30 mV are considered to be sufficient to provide repulsion between the nanoparticles in the dispersion and prevent their aggregation [128].

3.1. Lipid Nanoparticles—Structural Features and Recent Progress in Ocular Therapeutics

According to their main structural components, lipid nanoparticles may be distinguished into solid lipid nanoparticles (composed of solid-state lipids under ambient and physiological conditions) and nanostructured lipid carriers (additionally containing liquid lipids in their composition). In both cases, the lipid constituents are dispersed in an aqueous medium stabilized by surfactants [108]. Their specific structures and types are illustrated in Figure 2.

3.1.1. Solid Lipid Nanoparticles

Solid lipid nanoparticles are generally sphere-shaped colloidal systems, ranging between 50 and 1000 nm, and have been successfully explored as carriers for both hydrophilic and hydrophobic drugs [129]. The most frequently used solid lipids for their preparation include triglycerides (tristearin (Dynasan 118), tripalmitin (Dynasan 116), trimyristin (Dynasan 114)), a mixture or mixtures of mono-, di- and triglycerides (glyceryl behenate (Compritol 888 ATO), glyceryl palmitostearate (Precirol ATO 5)), waxes (beeswax, carnauba wax), fatty acids (lauric/stearic/myristic acid), and the corresponding fatty alcohols [130,131].
The chemical structure of lipids has a major impact on their physicochemical properties and delivery process of the nanoparticles, as reported by several studies. Boonme et al. investigated the effect of different lipids (glyceryl trimyristate, glyceryl tripalmitate, glyceryl tristearate, stearic acid, glyceryl monostearate) on the characteristics of SLNs obtained by the microemulsion technique. The selected lipids differ in the number of C atoms of the fatty acids chains, as well as their polarity. According to the obtained results, lipid polarity influences the capability to obtain microemulsions—the formation of such was reported in three of the studied formulations (comprising glyceryl monostearate, stearic acid and glyceryl trimyristate). This may be related to the absence of polar functional groups in the structure of glyceryl tripalmitate/glyceryl tristearate, as well as to their long (C-16/C-18) chains, determining large molecular volumes unable to penetrate into the hydrophobic region of the surfactant interface. The number of carbon atoms of the fatty acid residue also affects nanoparticle size—the smallest diameter was observed in the glyceryl trimyristate-based formulation, as a result of the shorter carbon chain (14 C atoms vs. C18 atoms) facilitating its penetration into the surfactant’s interface [132]. Palival et al. investigated the influence of several solid lipids (stearic acid, glycerol monostearate, tristearin, and Compritol 888 ATO) on the properties of methotrexate-loaded SLNs intended for oral delivery. According to the obtained results, the highest entrapment efficacy was reported for the Compritol 888-based SLNs, which may be related to the drug interchain intercalation [133].
The appropriate selection of a solid lipid or lipid mixture is an important subject, as it impacts the physicochemical characteristics (size, drug loading capacity), as well as drug release and storage stability, of the nanocarriers. Important issues to be considered during (pre)formulation studies include the solubility of drug in the lipid matrix, drug/lipid compatibility, and the lipid(s) crystalline behavior [134,135]. Based on the structural organization and drug location within the nanoparticles, three types of SLNs can be distinguished, as illustrated in Figure 2.
The homogenous matrix model is characterized by a uniformly allocated drug within the lipid matrix (molecularly dissolved or in form of amorphous clusters), mainly produced via the high-pressure homogenization method. The homogenous matrix particles result from the agitation of the dispersed drug in bulk lipid (when the cold technique is applied) or from the crystallization of cooled liquid droplets, in the case of hot homogenization. The latter is suitable for highly lipophilic drugs, without the necessity of using solubilizing agents [136].
The drug-enriched shell model involves predominantly localizing the drug in the outer shell of the nanoparticles, arising from phase separation and drug migration during the cooling stage of the process. Fast cooling induces the lipid in the center to precipitate, whereas the drug concentration in the residual liquid lipid increases, forming the outer shell. This model is characterized by fast drug release [137].
The drug-enriched core model is characterized by a high drug concentration in the melted lipid, leading to supersaturation of the drug and its precipitation during the cooling phase before lipid recrystallization. Further cooling subsequently leads to lipid recrystallization, and to the formation of a membrane overlaying the drug-enriched core [138].
In addition to the lipid constituents, a SLN formulation also contains surfactants, which facilitate the dispersion of lipids within the aqueous medium and stabilize the system by reducing the interfacial tension between both immiscible phases [139]. Generally, surfactants are included in the composition up to 5%w/w, and their selection is based upon several considerations, such as hydrophilic–lipophilic balance (HLB value), the route of administration of SLNs, safety profile, and compatibility with the other excipients [135,140]. In SLNs, intended for ophthalmic applications, the most-often included surfactants are non-ionic, such as polyoxyethylene sorbitan fatty acid esters (Polysorbates/Tweens), polyoxyethylene/polyoxypropylene block copolymers (Poloxamers/Pluronic), and amphoteric molecules, e.g., soy lecithin, due to their superior safety profiles compared to their anionic or cationic counterparts [119,131].
In their study, Silva et al., 2019 investigated the cytotoxicity of SLNs, containing the cationic surfactants cetyltrimethylammonium bromide (CTAB) and dimethyldioctadecylammonium bromide (DDAB), against five human cell lines of different origin. According to the obtained results CTAB-containing SLNs exhibited superior cytotoxicity in comparison to DDAB-SLNs, as the experimental concentration is closer to the critical micellar concentration of CTAB (the latter is related to cell lysis) [141].
SLNs may also contain cryoprotectants (e.g., trehalose, sorbitol, mannitol), in case the nanoparticles are subjected to lyophilization [142], as well as surface-modifying additives, such as polyethylene glycol, to confer stealth properties of the nanocarriers [143], or selective ligands, antibodies, etc., to provide targeted delivery [144,145]. In ocular therapeutics, SLNs are often modified using polyethylene glycol to improve their pharmacokinetic profile, or are coated with mucoadhesive polymers (e.g., chitosan), aiming to prolong their precorneal residence time [146,147].
In their study, Eid et al. investigated the impact of PEGylation and chitosan coating on the ocular bioavailability of ofloxacin-loaded SLNs. The addition of PEG stearate to the compositions determined higher transcorneal permeability, with a moderate effect on the mucoadhesion, in contrast to chitosan, which exerted the opposite effects. Ultimately, the developed PEGylated chitosan-coated SLNs improved the ocular bioavailability of ofloxacin by increasing the drug concentration in rabbits’ eyes two- to three-fold when compared to the plain drug [148]. The PEGylation approach was also adopted by Dang et al., who developed a PEGylated SLNs-laden contact lens, characterized by an enhanced latanoprost-loading capacity, smaller sizes (compared to non-PEGylated SLNs), and sustained drug release up to 96 h [149].
The development of hybrid drug-delivery platforms based on nanocarriers and a vehicle (semisolid formulations, in situ gels, contact lens) is an advantageous strategy for ocular delivery purposes, as it exploits the beneficial effects of both systems. In their study, Sun and Hu developed tacrolimus-loaded SLNs that were thermosensitive in situ gel, which were characterized by suitable gelling and rheological characteristics (gelation temperature 32 °C, pseudoplastic behavior), sustained drug release and improved pharmacodynamic effects when compared to the free drug and tacrolimus-loaded SLNs [150]. Improved biopharmaceutical and therapeutic outcomes were reported also for mizolastine-loaded hydrogel SLNs, manifesting in sustained drug release (up to 30 h) and reduced symptoms of allergic conjunctivitis in rabbits’ eyes [151].
Another beneficial SLN-based delivery strategy implemented in ocular therapeutics is the elaboration of dual solid lipid nanoparticles, as reported by Carbone et al. [152]. The authors aimed to improve the effectiveness of Candida albicans mycosis treatment by combining the antimycotic effect of clotrimazole and the antioxidant activity of alpha-lipoic acid. SLN as a delivery platform enabled the simultaneous loading of both drugs, and determined slow and controlled drug release, without an initial burst effect. The latter was achieved due to the successful incorporation of both drugs within the inner lipid matrix, and not on the nanoparticles’ surface [152].
An overview of the developed SLNs for ocular delivery purposes is provided in Table 2.
As presented in Table 2, SLNs have been successfully exploited for both anterior and posterior eye segment diseases. The reported therapeutic results may be attributed to various factors, such as the ability of SLNs to form a depot for the prolonged release of the drug, the fluidizing effect of included surfactants on the lipid bilayers of ocular membranes, facilitating drug permeation, as well as the large surface area of nanocarriers, providing maximized contact with the ocular mucosa [163,164]. It is also worth noting the ability of SLNs to encapsulate high molecular weight compounds, such as atorvastatin [158] and natamycin [154], which are also characterized by poor solubility, therefore, their ocular delivery through conventional ophthalmic formulations would be a challenge. Encapsulation of atorvastatin in SLNs further contributed to improved drug photostability, as confirmed by the photostability studies conducted according to ICH guidelines [158]. Liang et al. also reported overcoming the unfavorable characteristics of the drug by developing econazole-loaded SLNs. The antimycotic is characterized by low aqueous solubility and strong irritation potential, which restrain its application in the therapy of ocular fungal infections. The conducted in vivo studies showed enhanced corneal permeation, and no ocular irritation with the econazole-loaded SLNs [153]. Solid lipid nanoparticles are also beneficial in the therapy of posterior segment diseases, e.g., glaucoma, as confirmed by the superior intraocular pressure reduction [162], and higher therapeutic concentration in the iris, ciliary body, and retina [163].
The pre-clinical safety of SLNs was evaluated in polymeric nanospheres and liposomes in a recent study conducted by Gomes Souza et al. The authors elaborated sunitinib-loaded nanocarriers as topical formulation strategies for corneal neovascularization treatment. The sunitinib-loaded SLNs were selected as the optimal formulation due to their excellent tolerability profile, controlled drug release, and highest corneal retention [165].

3.1.2. Nanostructured Lipid Carriers

Nanostructured lipid carriers were initially developed to surmount the limitations associated with SLNs, such as their poor drug-loading capacity, owing to their perfectly arranged crystalline structure, and their propensity towards drug expulsion during storage, resulting from lipid crystallization [37,166]. The addition of spatially incompatible liquid lipid(s) to the formulations is beneficial in two aspects, it leads to the formation of a less-ordered crystalline structure (Figure 2), ensuring extra area for drug loading, decreases the crystalline degree of the lipid matrix and averts drug expulsion [128,167]. Usually, the liquid lipid is included up to 30% of the total lipid amount in the NLCs formulations [168,169]. As such, researchers often use castor/olive/argan oil, oleic acid, Miglyol® 812 (medium-chain triglycerides), propylene glycol dicaprylocaprate—Labrafac™ PG (Gattefosse, Saint-Priest, France), or caprylocaproyl macrogol-8 glycerides—Labrasol® (Gattefosse, Saint-Priest, France) [170,171,172].
The selection of both solid and liquid lipids is reported to influence NLCs’ size. According to Apostolou et al., NLCs comprising solid lipids, such as Precirol ATO 5 (Gattefosse, Saint-Priest, France), Compritol 888 ATO (Gattefosse, Saint-Priest, France) or Dynasan 118 (IOI Oleo GmbH, Hamburg, Germany), exhibit larger particle sizes when compared to glyceryl monostearate- and stearic acid-based nanocarriers. A possible explanation may lie in the higher molecular weight of the lipids, leading to the formation of a more complex structure, with a tendency of aggregation between the molecules, which results in an increased nanoparticle diameter [173]. Concerning the selection of liquid lipids, NLCs containing Mygliol® 812 (IOI Oleo GmbH, Hamburg, Germany) are generally characterized by larger size when compared to oleic acid or Capryol 90-containing ones (Gattefosse, Saint-Priest, France) [174,175,176].
NLCs can be classified into three models depending on the preparation methods, lipid matrix structure, and drug location [177].
The imperfect type is obtained by blending structurally different lipids, resulting in the formation of disorganized lipid matrix. The selected lipids, usually a small fraction of liquid oil mixed with larger amount of solid lipid, may differ in terms of fatty acid origin, in their carbon chain length or degree of saturation. This type of NLC is characterized by its high drug-loading capacity, proportionally related to the imperfections within the lipid matrix [178].
The amorphous-type NLCs are formed owing to the addition of specific lipids to the formulation, such as hydroxyoctacosanyl hydroxystearate and isopropyl myristate. These lipids contribute to the formation of a non-crystalline (amorphous) matrix, limiting drug expulsion as a result of solid lipid crystallization [143].
The multiple-type NLCs are oil-in-solid, fat-in-water nanocarriers, composed of numerous liquid oil nanocompartments within a solid lipid matrix, usually obtained through the hot homogenization technique. The greater amount of liquid lipid in the formulation leads to phase separation and the formation of the nanosized droplets upon the cooling phase. The multiple-type NLCs are characterized by high drug-loading capacities, due to the superior solubility of lipophilic drugs in liquid lipids compared to those in solid ones. Furthermore, the solid matrix exhibits a barrier function, limiting drug leakage and controlling the release process [178,179].
Similar to SLNs, the surface of NLCs can be modified with cationic additives (e.g., chitosan) to impart muco-adhesiveness, sustained drug release, and increased penetration, as reported by Selvaraj et al. [180], Sharma et al. [181], and Fu et al. [182]. Derivatives of chitosan (trimethyl chitosan) and chitin (chitosan oligosaccharide) have also been investigated as nanoparticle surface-coating materials, as they exhibit improved aqueous solubility at a neutral pH (including in the lacrimal fluid) and superior safety profiles compared to native chitosan, while at the same time retaining all of its beneficial characteristics (biodegradability, muco-adhesion, penetration-enhancing properties, etc.) [183,184].
Mucoadhesive NLCs have also been developed by functionalization with (3-aminomethylphenyl) boronic acid attached to chondroitin sulfate, to increase corneal residence time by specifically targeting the sialic acid residues on the ocular surface, which ultimately improves drug performance regarding dry eye disease [185]. In vivo relief of dry eye disease symptoms, accompanied by enhanced corneal retention, was also reported by Zhu et al., developing chondroitin sulfate and L-cysteine conjugate-modified dexamethasone NLCs [186].
In another study Abdelhakeem et al. elaborated on surface-modified eplerenone-loaded NLCs for the treatment of central serous chorioretinopathy. The authors evaluated the effect of three different coating polymers (hyaluronic acid, chitosan oligosaccharide lactate, and hydrogenated collagen) on the properties of the nanocarriers. The largest particle size was reported for the hyaluronic acid-coated NLCs, corresponding to the formulation’s highest eplerenone entrapment efficiency and viscosity. The higher viscosity determined the superior sustained drug release from hyaluronic acid-modified NLCs compared to the other NLCs models. The selected optimal formulations (hyaluronic acid/chitosan oligosaccharide lactate-coated) were characterized by an excellent ocular tolerability, as confirmed by the Draize test [187].
Nanostructured lipid carriers have been also an integral component of hybrid drug-delivery platforms, recently included into thermosensitive in situ gel-forming systems [188,189]. An interesting approach is described by Yu et al. in two of their studies, elaborating on baicalin NLCs and quercetin NLCs that were subsequently incorporated into dual pH and thermosensitive in situ gels. The dual stimuli-responsive formulation was based on carboxymethyl chitosan and Poloxamer 407, cross-linked by the natural cross-linker genipin. Both hybrid, NLC-loaded, in situ gels were characterized by prolonged drug release and precorneal residence time, and improved transcorneal penetration compared to eye drops [190,191].
Dual nanostructured lipid carriers have also been developed for ocular delivery purposes. In their study Youseff et al. developed simultaneously loaded natamycin/ciprofloxacin NLCs as a drug delivery system for microbial keratitis treatment. The selection of model drugs (an antifungal agent and fluoroquinolone antibiotic) was based on the complex etiology of corneal infections (which may be caused by bacteria/fungi/protozoa, when a secondary or co-infection is present). The elaborated dual NLCs were subsequently incorporated into in situ ionic gel formulations, aiming to further enhance the therapeutic efficacy by providing prolonged ocular surface contact time [120]. Dual therapeutic synergy was exploited also by Chen and Wu when developing brinzolamide- and latanoprost-loaded NLCs for the therapy of glaucoma (details of the study are presented in Table 3) [192].
Further overview of the recent progress of NLCs in ocular therapeutics is shown in Table 3.
Nanostructured lipid carriers are feasible delivery systems for both drugs and biologically active compounds, as illustrated in Table 3. Polyphenolic compounds are well-known for their antioxidant effects, which would be highly beneficial in the therapy of ocular degenerative diseases. However, these phytochemicals are usually characterized by poor aqueous solubility and an unfavorable pharmacokinetic profile, as reported for diosmin [203] and mangiferin [204]. Their encapsulation in NLCs led to an improvement of their disadvantageous physicochemical properties (e.g., low aqueous solubility), and further contributed to superior antioxidant activity (in the case of the mangiferin-loaded NLCs) and cytoprotective effects (for diosmin-loaded NLCs). Other beneficial outcomes following drug loading into NLCs include superior chemical/photo stability, estimated by the rapamycin-loaded NLCs [195], as well as the pronounced enhancement of the solubility of dasatinib upon encapsulation. The latter further contributes to the observed higher anti-proliferation and anti-migration effects [198].
In addition to the conventional topical application, NLCs have been formulated for periocular administration (transscleral delivery), as reported by González-Fernández et al. The authors prepared dexamethasone acetate-loaded NLCs intended for the treatment of posterior eye segment diseases (e.g., macular edema, age-related macular degeneration). The encapsulated prodrug acetate ester provided sustained drug release as a result of the required enzymatic conversion step, and enhanced scleral/choroidal permeability [205].

3.2. Sterilization Feasibility of SLNs and NLCs

Owing to their compositional similarities, NLCs and SLNs can be prepared by identical methods, such as high-pressure homogenization (hot/cold option), high-speed homogenization and/or ultrasonication, solvent emulsification/evaporation, microemulsion, phase inversion techniques, and the solvent injection method [143]. A comprehensive description of the various preparation methods has been detailed by Gordillo-Galeano and Mora-Huertas [131], Khairnar et al. [206] and Duong et al. [207]. However, of great importance for ocular application is one of the post-production steps, namely, the sterilization feasibility.
Techniques such as heat sterilization (autoclaving), sterile filtration and gamma irradiation have been used as sterilization methods for SLNs and NLCs intended for ophthalmic application. The selection of the specific method is based on several considerations, such as drug heat stability, composition constituents (melting point of lipids, choice of surfactants), nanoparticle size, and the viscosity of the solution in case of sterile filtration [83,162,208]. Autoclaving is the most commonly exploited technique for the sterilization of lipid nanoparticles in ophthalmology, however, with controversial results regarding its impact on the physiochemical characteristics of the nanocarriers. According to some reports, there is no significant change in the particle size [158,172,209] or entrapment efficiency [158] of developed lipid nanocarriers before and after sterilization, in contrast to others, which established an increase in particle size in the micrometer range [210]. The latter may be ascribed to the compromised surfactant film properties, as well as to the melting of lipids at 121 °C, leading to the formation of an o/w emulsion. During the successive cooling and lipid recrystallization, no energy input (i.e., homogenization) was applied to the system, resulting in the increase of particle size [210]. In their study Youshia et al. investigated the influence of autoclaving and sterilization by gamma irradiation on the physicochemical parameters of methazolamide-loaded cationic NLCs. According to the results, NLCs subjected to heat sterilization were characterized by significantly lower entrapment efficiency and zeta potential values. At the same time an increase in the particle size and polydispersity index was observed. On the contrary, gamma radiation did not induce significant alterations in the particles size, size distribution pattern, or in the degree of methazolamide entrapment [211]. However, one of the main limitations of this method is the formation of free radicals, therefore, subsequent studies need to be performed, in order to evaluate the chemical stability of the components. Additionally, different strategies may be applied to mitigate the adverse effects of radiation, such as adjustment of the applied dose, lyophilization of the samples, and the use of suitable (endure to γ-radiation) excipients [208].
Sterile filtration has also been exploited as a sterilization approach for lipid nanoparticles used in ophthalmic application, as described by Bonaccorso et al. [157]. The authors investigated the influence of different types of membranes (polypropylene, polyethylene sulfone, polyvinylidene fluoride; pore size of 0.22 µm) on the filtration feasibility of sorafenib-loaded SLNs. The obtained results showed that polypropylene and polyethylene sulfone filters restrain the filtration process by retaining the nanoparticles within the membrane, unlike the polyvinylidene fluoride membrane, which enables SLNs’ passage. Furthermore, the obtained SLN suspension after filtration was characterized by unaltered physiochemical parameters [157].

3.3. Clinical Application of SLNs and NLCs in Ocular Therapeutics

Several lipid-based ophthalmic nanocarriers have been successfully implemented into clinical practice, such as Visudyne® (Novartis Pharma AG, Basel, Switzerland), a liposomal verteporfine nanoformulation intended for the therapy of age-related macular degeneration, Durezol® (Alcon, Geneva, Switzerland), a difluprednate nanoemulsion for ocular inflammation treatment, and Restasis® (AbbVie, North Chicago, IL, USA), a cyclosporine nanoemulsion intended for the therapy of dry eye disease [212,213]. However, regardless of the positive outcomes garnered from conducted studies, currently, there are no SLN- or NLC-based ophthalmic formulations that have been translated into clinical applications or marketed. A search through the website www.clinicaltrials.gov (accessed on 1 March 2023) using the keyword ”solid lipid nanoparticles” resulted in 10 studies, whereas the keyword “nanostructured lipid carriers” led to 2 results. Currently, none of these trials are related to ocular delivery purposes. Further details are provided in Table S1.

4. Conclusions and Prospects

Solid lipid nanoparticles and nanostructured lipid carriers have shown significant potential for effective ocular drug delivery, as confirmed by the findings summarized in this review. Their advantageous characteristics such as biodegradability, biocompatibility, owing to the generally recognized as safe (GRAS) lipid constituents, and their possibility to provide controlled and sustained drug release, to improve transcorneal penetration and enhance ocular bioavailability, determine their increasing progress in ocular therapeutics. Furthermore, the surface of both types of nanocarriers can be modified to improve their pharmacokinetic characteristics, impart mucoadhesive properties, prolong corneal residence time, and enhance their therapeutic efficacy. The latter can also be achieved by incorporating them into semisolid/in situ gelling formulations and contact lenses (i.e., hybrid delivery systems), which is another promising research direction and would be of great benefit, especially in case of ocular surface diseases. Drug delivery to the posterior segment of the eye can also be accomplished via SLNs and NLCs by proper adjustment of the formulation-related parameters (lipid constituents/surfactant(s) selection; tuning particles’ size into the desired nano range), which would be of great significance in the therapy of vision-threatening diseases. However, despite all the promising outcomes from conducted studies, the research progress has not been implemented into clinical application yet. Some of the challenges related to this matter include the possibility of developing reproducible batches of lipid nanoparticles, which exhibit sufficient colloidal stability during storage. In this regard, the implementation of quality-by-design (QbD) approach during the (pre)formulation stage is a feasible strategy, as it provides the possibility to obtain a final product with predictable quality attributes, which would benefit and facilitate nanocarriers’ subsequent commercialization [214]. Ocular toxicity is another critical issue to be considered during the development of ophthalmic formulations. According to the findings from the reviewed articles, SLNs and NLCs showed no level of toxicity (based on in vitro or in vivo studies), however, further studies are needed to evaluate their long-term toxicity, as well as their fate after application in vivo [215]. Regarding their clinical application approval, it is crucial to establish unified protocols evaluating their safety and effectiveness [107]. Based on the promising results from the conducted studies, it can be concluded that the potential of SLNs and NLCs should be fully deployed in the near future.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph16030474/s1, Table S1. Solid lipid nanoparticles and nanostructured lipid carriers in clinical trials (terminated studies and studies with unknown status are excluded).

Author Contributions

Conceptualization, V.G. and V.A.; methodology, V.G. and V.A.; investigation, V.G. and V.A.; writing—original draft preparation, V.G. and V.A.; writing—review and editing, V.A.; visualization, V.G.; supervision, V.A.; project administration, V.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. World Report on Vision; World Health Organization: Geneva, Switzerland, 2019; ISBN 9789241516570. [Google Scholar]
  2. Usgaonkar, U.; Shet Parkar, S.R.; Shetty, A. Impact of the use of digital devices on eyes during the lockdown period of COVID-19 pandemic. Ind. J. Ophthalmol. 2021, 69, 1901–1906. [Google Scholar] [CrossRef]
  3. Shalini, S.; Ipsita, P.; Abhay, P.; Pandey, A.K. Life style disorders in ophthalmology and their management. Environ. Conserv. J. 2019, 20, 67–72. [Google Scholar]
  4. García-Marqués, J.V.; Talens-Estarelles, C.; García-Lázaro, S.; Wolffsohn, J.S.; Cerviño, A. Systemic, environmental and lifestyle risk factors for dry eye disease in a mediterranean caucasian population. Cont. Lens Anterior Eye. 2022, 45, 101539. [Google Scholar] [CrossRef] [PubMed]
  5. Bourne, R.R.A.; Steinmetz, J.D.; Flaxman, S.; Briant, P.S.; Taylor, H.R.; Resnikoff, S.; Casson, R.J.; Abdoli, A.; Gharbieh, E.A.; Afshin, A.; et al. Trends in prevalence of blindness and distance and near vision impairment over 30 years: An analysis for the Global Burden of Disease Study. Lancet Glob. Health. 2021, 9, e130–e143. [Google Scholar] [CrossRef]
  6. Gote, V.; Sikder, S.; Sicotte, J.; Pal, D. Ocular Drug Delivery: Present Innovations and Future Challenges. J. Pharmacol. Exp. Ther. 2019, 370, 602–624. [Google Scholar] [CrossRef]
  7. Patel, A.; Cholkar, K.; Agrahari, V.; Mitra, A.K. Ocular drug delivery systems: An overview. World J. Pharmacol. 2013, 2, 47–64. [Google Scholar] [CrossRef] [PubMed]
  8. Nayak, K.; Misra, M. A review on recent drug delivery systems for posterior segment of eye. Biomed. Pharmacother. 2018, 107, 1564–1582. [Google Scholar] [CrossRef]
  9. Bachu, R.D.; Chowdhury, P.; Al-Saedi, Z.H.F.; Karla, P.K.; Boddu, S.H.S. Ocular Drug Delivery Barriers-Role of Nanocarriers in the Treatment of Anterior Segment Ocular Diseases. Pharmaceutics 2018, 10, 28. [Google Scholar] [CrossRef] [Green Version]
  10. Wilson, C.G. Ophthalmic Formulation. In Specialized Pharmaceutical Formulation: The Science and Technology of Dosage Forms, 1st ed.; Tovey, G.D., Ed.; Royal Society of Chemistry: London, UK, 2022; pp. 1–44. [Google Scholar]
  11. Kuno, N.; Fujii, S. Recent Advances in Ocular Drug Delivery Systems. Polymers 2011, 3, 193–221. [Google Scholar] [CrossRef]
  12. Jünemann, A.G.M.; Chorągiewicz, T.; Ozimek, M.; Grieb, P.; Rejdak, R. Drug bioavailability from topically applied ocular drops. Does drop size matter? Ophthalmol. J. 2016, 1, 29–35. [Google Scholar] [CrossRef] [Green Version]
  13. Taghe, S.; Mirzaeei, S. Preparation and characterization of novel, mucoadhesive ofloxacin nanoparticles for ocular drug delivery. Braz. J. Pharm. Sci. 2019, 55, e17105. [Google Scholar] [CrossRef] [Green Version]
  14. Gaudana, R.; Ananthula, H.K.; Parenky, A.; Mitra, A.K. Ocular drug delivery. AAPS J. 2010, 12, 348–360. [Google Scholar] [CrossRef]
  15. Baranowski, P.; Karolewicz, B.; Gajda, M.; Pluta, J. Ophthalmic drug dosage forms: Characterisation and research methods. Sci. World J. 2014, 2014, 861904. [Google Scholar] [CrossRef] [Green Version]
  16. Wu, Y.; Liu, Y.; Li, X.; Kebebe, D.; Zhang, B.; Ren, J.; Lu, J.; Li, J.; Du, S.; Liu, Z. Research progress of in-situ gelling ophthalmic drug delivery system. Asian J. Pharm. Sci. 2019, 14, 1–15. [Google Scholar] [CrossRef]
  17. Bastawrous, A.; Burgess, P.I.; Mahdi, A.M.; Kyari, F.; Burton, M.J.; Kuper, H. Posterior segment eye disease in sub-Saharan Africa: Review of recent population-based studies. Trop. Med. Int. Health. 2014, 19, 600–609. [Google Scholar] [CrossRef] [Green Version]
  18. Tóth, G.; Szabó, D.; Sándor, G.L.; Nagy, Z.Z.; Limburg, H.; Németh, J. Hátsószegmens-betegségek okozta látásromlás és vakság Magyarországon az 50 évnél idosebb korú lakosság körében [Visual impairment and blindness caused by posterior segment diseases in Hungary in people aged 50 years and older]. Orv. Hetil. 2022, 163, 624–630. [Google Scholar] [CrossRef]
  19. Varela-Fernández, R.; Díaz-Tomé, V.; Luaces-Rodríguez, A.; Conde-Penedo, A.; García-Otero, X.; Luzardo-Álvarez, A.; Fernández-Ferreiro, A.; Otero-Espinar, F.J. Drug Delivery to the Posterior Segment of the Eye: Biopharmaceutic and Pharmacokinetic Considerations. Pharmaceutics 2020, 12, 269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Otero-Espinar, F.J.; Fernández-Ferreiro, A.; González-Barcia, M.; Blanco-Méndez, J.; Luzardo, A. Stimuli sensitive ocular drug delivery systems. In Drug Targeting and Stimuli Sensitive Drug Delivery Systems, 1st ed.; Grumezescu, A., Ed.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 211–270. [Google Scholar]
  21. Sánchez-López, E.; Espina, M.; Doktorovova, S.; Souto, E.B.; García, M.L. Lipid nanoparticles (SLN, NLC): Overcoming the anatomical and physiological barriers of the eye—Part I—Barriers and determining factors in ocular delivery. Eur. J. Pharm. Biopharm. 2017, 110, 70–75. [Google Scholar] [CrossRef] [PubMed]
  22. Dos Santos, G.A.; Ferreira-Nunes, R.; Dalmolin, L.F.; Dos Santos Ré, A.C.; Anjos, J.L.V.; Mendanha, S.A.; Aires, C.P.; Lopez, R.F.V.; Cunha-Filho, M.; Gelfuso, G.M.; et al. Besifloxacin liposomes with positively charged additives for an improved topical ocular delivery. Sci. Rep. 2020, 10, 19285. [Google Scholar] [CrossRef]
  23. Chen, X.; Wu, J.; Lin, X.; Wu, X.; Yu, X.; Wang, B.; Xu, W. Tacrolimus Loaded Cationic Liposomes for Dry Eye Treatment. Front. Pharmacol. 2022, 13, 838168. [Google Scholar] [CrossRef] [PubMed]
  24. Fathalla, D.; Fouad, E.A.; Soliman, G.M. Latanoprost niosomes as a sustained release ocular delivery system for the management of glaucoma. Drug Dev. Ind. Pharm. 2020, 46, 806–813. [Google Scholar] [CrossRef] [PubMed]
  25. 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] [PubMed]
  26. Vicente-Pascual, M.; Gómez-Aguado, I.; Rodríguez-Castejón, J.; Rodríguez-Gascón, A.; Muntoni, E.; Battaglia, L.; del Pozo-Rodríguez, A.; Solinís Aspiazu, M.Á. Topical Administration of SLN-Based Gene Therapy for the Treatment of Corneal Inflammation by De Novo IL-10 Production. Pharmaceutics 2020, 12, 584. [Google Scholar] [CrossRef]
  27. Wang, L.; Liu, W.; Huang, X. An approach to revolutionize cataract treatment by enhancing drug probing through intraocular cell line. Libyan J. Med. 2018, 13, 1500347. [Google Scholar] [CrossRef] [PubMed]
  28. Lou, X.; Hu, Y.; Zhang, H.; Liu, J.; Zhao, Y. Polydopamine nanoparticles attenuate retina ganglion cell degeneration and restore visual function after optic nerve injury. J. Nanobiotechnology 2021, 19, 436. [Google Scholar] [CrossRef]
  29. Francia, S.; Shmal, D.; Di Marco, S.; Chiaravalli, G.; Maya-Vetencourt, J.F.; Mantero, G.; Michetti, C.; Cupini, S.; Manfredi, G.; DiFrancesco, M.L.; et al. Light-induced charge generation in polymeric nanoparticles restores vision in advanced-stage retinitis pigmentosa rats. Nat. Commun. 2022, 13, 3677. [Google Scholar] [CrossRef]
  30. Rincón, M.; Espinoza, L.C.; Silva-Abreu, M.; Sosa, L.; Pesantez-Narvaez, J.; Abrego, G.; Calpena, A.C.; Mallandrich, M. Quality by Design of Pranoprofen Loaded Nanostructured Lipid Carriers and Their Ex Vivo Evaluation in Different Mucosae and Ocular Tissues. Pharmaceuticals 2022, 15, 1185. [Google Scholar] [CrossRef] [PubMed]
  31. Polat, H.K.; Kurt, N.; Aytekin, E.; Akdağ Çaylı, Y.; Bozdağ Pehlivan, S.; Çalış, S. Design of Besifloxacin HCl-Loaded Nanostructured Lipid Carriers: In Vitro and Ex Vivo Evaluation. J. Ocul. Pharmacol. Ther. 2022, 38, 412–423. [Google Scholar] [CrossRef]
  32. Sun, L.; Zhang, M.; Shi, Y.; Fang, L.; Cao, F. Rational design of mixed nanomicelle eye drops with structural integrity investigation. Acta Biomater. 2022, 141, 164–177. [Google Scholar] [CrossRef]
  33. Xu, X.; Sun, L.; Zhou, L.; Cheng, Y.; Cao, F. Functional chitosan oligosaccharide nanomicelles for topical ocular drug delivery of dexamethasone. Carbohydr. Polym. 2020, 227, 115356. [Google Scholar] [CrossRef]
  34. Nayak, K.; Misra, M. PEGylated microemulsion for dexamethasone delivery to posterior segment of eye. J. Biomater. Sci. Polym. Ed. 2020, 31, 1071–1090. [Google Scholar] [CrossRef] [PubMed]
  35. Bachu, R.D.; Stepanski, M.; Alzhrani, R.M.; Jung, R.; Boddu, S.H.S. Development and Evaluation of a Novel Microemulsion of Dexamethasone and Tobramycin for Topical Ocular Administration. J. Ocul. Pharmacol. Ther. 2018, 34, 312–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Bravo-Osuna, I.; Vicario-de-la-Torre, M.; Andrés-Guerrero, V.; Sánchez-Nieves, J.; Guzmán-Navarro, M.; de la Mata, F.J.; Gómez, R.; de Las Heras, B.; Argüeso, P.; Ponchel, G.; et al. Novel Water-Soluble Mucoadhesive Carbosilane Dendrimers for Ocular Administration. Mol. Pharm. 2016, 13, 2966–2976. [Google Scholar] [CrossRef]
  37. Ghasemiyeh, P.; Mohammadi-Samani, S. Solid lipid nanoparticles and nanostructured lipid carriers as novel drug delivery systems: Applications, advantages and disadvantages. Res. Pharm. Sci. 2018, 13, 288–303. [Google Scholar]
  38. Costa, C.P.; Barreiro, S.; Moreira, J.N.; Silva, R.; Almeida, H.; Sousa Lobo, J.M.; Silva, A.C. In Vitro Studies on Nasal Formulations of Nanostructured Lipid Carriers (NLC) and Solid Lipid Nanoparticles (SLN). Pharmaceuticals 2021, 14, 711. [Google Scholar] [CrossRef]
  39. Nguyen, V.H.; Thuy, V.N.; Van, T.V.; Dao, A.H.; Lee, B.-J. Nanostructured lipid carriers and their potential applications for versatile drug delivery via oral administration. OpenNano 2022, 8, 100064. [Google Scholar] [CrossRef]
  40. Czajkowska-Kośnik, A.; Szekalska, M.; Winnicka, K. Nanostructured lipid carriers: A potential use for skin drug delivery systems. Pharmacol. Rep. 2019, 71, 156–166. [Google Scholar] [CrossRef]
  41. Musielak, E.; Feliczak-Guzik, A.; Nowak, I. Optimization of the Conditions of Solid Lipid Nanoparticles (SLN) Synthesis. Molecules 2022, 27, 2202. [Google Scholar] [CrossRef] [PubMed]
  42. Poonia, N.; Kharb, R.; Lather, V.; Pandita, D. Nanostructured lipid carriers: Versatile oral delivery vehicle. Future Sci. OA 2016, 2, FSO135. [Google Scholar] [CrossRef] [Green Version]
  43. Abo El-Enin, H.A.; Elkomy, M.H.; Naguib, I.A.; Ahmed, M.F.; Alsaidan, O.A.; Alsalahat, I.; Ghoneim, M.M.; Eid, H.M. Lipid Nanocarriers Overlaid with Chitosan for Brain Delivery of Berberine via the Nasal Route. Pharmaceuticals 2022, 15, 281. [Google Scholar] [CrossRef]
  44. Mirchandani, Y.; Patravale, V.B.; Brijesh, S. Solid lipid nanoparticles for hydrophilic drugs. J. Control Release. 2021, 335, 457–464. [Google Scholar] [CrossRef] [PubMed]
  45. Neves, A.R.; Queiroz, J.F.; Weksler, B.; Romero, I.A.; Couraud, P.O.; Reis, S. Solid lipid nanoparticles as a vehicle for brain-targeted drug delivery: Two new strategies of functionalization with apolipoprotein E. Nanotechnology 2015, 26, 495103. [Google Scholar] [CrossRef] [PubMed]
  46. Lee, W.J. Eye. In Vitamin C in Human Health and Disease, 1st ed.; Lee, W.J., Ed.; Springer: Dordrecht, The Netherlands, 2019; pp. 177–182. [Google Scholar]
  47. Watson, P.G.; Young, R.D. Scleral structure, organisation and disease. A review. Exp. Eye Res. 2004, 78, 609–623. [Google Scholar] [CrossRef] [PubMed]
  48. Sridhar, M.S. Anatomy of cornea and ocular surface. Indian J. Ophthalmol. 2018, 66, 190–194. [Google Scholar] [CrossRef]
  49. Labelle, P. The Eye. In Pathologic Basis of Veterinary Disease, 6th ed.; Zachary, J.F., Ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 1265–1318.e1. [Google Scholar]
  50. Moiseev, R.V.; Morrison, P.W.J.; Steele, F.; Khutoryanskiy, V.V. Penetration Enhancers in Ocular Drug Delivery. Pharmaceutics 2019, 11, 321. [Google Scholar] [CrossRef] [Green Version]
  51. Salazar, J.J.; Ramírez, A.I.; De Hoz, R.; Salobrar-Garcia, E.; Rojas, P.; Fernández-Albarral, J.A.; López-Cuenca, I.; Rojas, B.; Triviño, A.; Ramírez, J.M. Anatomy of the Human Optic Nerve: Structure and Function; Ferreri, F.M., Ed.; IntechOpen: London, UK, 2018; pp. 1–46. [Google Scholar]
  52. Lin, S.; Ge, C.; Wang, D.; Xie, Q.; Wu, B.; Wang, J.; Nan, K.; Zheng, Q.; Chen, W. Overcoming the Anatomical and Physiological Barriers in Topical Eye Surface Medication Using a Peptide-Decorated Polymeric Micelle. ACS Appl. Mater. Interfaces 2019, 11, 39603–39612. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, X.; Zuo, Y.Y. Nanomedicine for Ocular Drug Delivery. In Nanomedicine, 1st ed.; Gu, N., Ed.; Springer: Singapore, 2022; pp. 755–786. [Google Scholar]
  54. McDermott, A.M. Antimicrobial Compounds in Tears. Exp. Eye Res. 2013, 117, 53–61. [Google Scholar] [CrossRef] [Green Version]
  55. Kopacz, D.; Niezgoda, Ł.; Fudalej, E.; Nowak, A.; Maciejewicz, P. Tear film—Physiology and Disturbances in Various Diseases and Disorders. In Ocular Surface Diseases—Some Current Date on Tear Film Problem and Keratoconic Diagnosis, 1st ed.; Kopacz, D., Ed.; IntechOpen: London, UK, 2021; pp. 1–17. [Google Scholar]
  56. Willcox, M.D.P.; Argüeso, P.; Georgiev, G.A.; Holopainen, J.M.; Laurie, G.W.; Millar, T.J.; Papas, E.B.; Rolland, J.P.; Schmidt, T.A.; Stahl, U.; et al. TFOS DEWS II Tear Film Report. Ocul. Surf. 2017, 15, 366–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Yang, Y.; Lockwood, A. Topical ocular drug delivery systems: Innovations for an unmet need. Exp. Eye Res. 2022, 218, 109006. [Google Scholar] [CrossRef]
  58. Jumelle, C.; Gholizadeh, S.; Annabi, N.; Dana, R. Advances and limitations of drug delivery systems formulated as eye drops. J. Control Release. 2020, 321, 1–22. [Google Scholar] [CrossRef]
  59. Shafaie, S.; Hutter, V.; Cook, M.T.; Brown, M.B.; Chau, D.Y.S. In Vitro Cell Models for Ophthalmic Drug Development Applications. Biores. Open Access. 2016, 5, 94–108. [Google Scholar] [CrossRef] [Green Version]
  60. Peris-Martínez, C.; García-Domene, M.C.; Penadés, M.; Luque, M.J.; Fernández-López, E.; Artigas, J.M. Spectral Transmission of the Human Corneal Layers. J. Clin. Med. 2021, 10, 4490. [Google Scholar] [CrossRef] [PubMed]
  61. Ruan, Y.; Jiang, S.; Musayeva, A.; Pfeiffer, N.; Gericke, A. Corneal Epithelial Stem Cells-Physiology, Pathophysiology and Therapeutic Options. Cells 2021, 10, 2302. [Google Scholar] [CrossRef] [PubMed]
  62. Shastri, D.H.; Silva, A.C.; Almeida, H. Ocular Delivery of Therapeutic Proteins: A Review. Pharmaceutics 2023, 15, 205. [Google Scholar] [CrossRef]
  63. Wang, R.; Gao, Y.; Liu, A.; Zhai, G. A review of nanocarrier-mediated drug delivery systems for posterior segment eye disease: Challenges analysis and recent advances. J. Drug Target 2021, 29, 687–702. [Google Scholar] [CrossRef] [PubMed]
  64. Nakano, M.; Lockhart, C.M.; Kelly, E.J.; Rettie, A.E. Ocular cytochrome P450s and transporters: Roles in disease and endobiotic and xenobiotic disposition. Drug Metab. Rev. 2014, 46, 247–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Lagali, N. Corneal Stromal Regeneration: Current Status and Future Therapeutic Potential. Curr. Eye Res. 2020, 45, 278–290. [Google Scholar] [CrossRef] [Green Version]
  66. Morrison, P.W.; Khutoryanskiy, V.V. Advances in ophthalmic drug delivery. Ther. Deliv. 2014, 5, 1297–1315. [Google Scholar] [CrossRef] [Green Version]
  67. Agrahari, V.; Mandal, A.; Agrahari, V.; Trinh, H.M.; Joseph, M.; Ray, A.; Hadji, H.; Mitra, R.; Pal, D.; Mitra, A.K. A comprehensive insight on ocular pharmacokinetics. Drug Deliv. Transl. Res. 2016, 6, 735–754. [Google Scholar] [CrossRef] [Green Version]
  68. Pak, J.; Chen, Z.J.; Sun, K.; Przekwas, A.; Walenga, R.; Fan, J. Computational Modeling of Drug Transport Across the In Vitro Cornea. Comput. Biol. Med. 2018, 92, 139–146. [Google Scholar] [CrossRef]
  69. Löscher, M.; Seiz, C.; Hurst, J.; Schnichels, S. Topical Drug Delivery to the Posterior Segment of the Eye. Pharmaceutics 2022, 14, 134. [Google Scholar] [CrossRef]
  70. Harikumar, S.I.; Sonia, A. Nanotechnological approaches in Ophthalmic delivery systems. Int. J. Drug Dev. Res. 2011, 3, 9–19. [Google Scholar]
  71. Kwatra, D.; Mitra, A.K. Drug delivery in ocular diseases: Barriers and strategies. World J. Pharmacol. 2013, 2, 78–83. [Google Scholar] [CrossRef]
  72. Akhter, M.H.; Ahmad, I.; Alshahrani, M.Y.; Al-Harbi, A.I.; Khalilullah, H.; Afzal, O.; Altamimi, A.S.A.; Najib Ullah, S.N.M.; Ojha, A.; Karim, S. Drug Delivery Challenges and Current Progress in Nanocarrier-Based Ocular Therapeutic System. Gels 2022, 8, 82. [Google Scholar] [CrossRef] [PubMed]
  73. de Oliveira, I.F.; Barbosa, E.J.; Peters, M.C.C.; Henostroza, M.A.B.; Yukuyama, M.N.; Dos Santos Neto, E.; Löbenberg, R.; Bou-Chacra, N. Cutting-edge advances in therapy for the posterior segment of the eye: Solid lipid nanoparticles and nanostructured lipid carriers. Int. J. Pharm. 2020, 589, 119831. [Google Scholar] [CrossRef]
  74. Pescina, S.; Lucca, L.G.; Govoni, P.; Padula, C.; Favero, E.D.; Cantù, L.; Santi, P.; Nicoli, S. Ex Vivo Conjunctival Retention and Transconjunctival Transport of Poorly Soluble Drugs Using Polymeric Micelles. Pharmaceutics 2019, 11, 476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. 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]
  76. Szabadi, E. Functional Organization of the Sympathetic Pathways Controlling the Pupil: Light-Inhibited and Light-Stimulated Pathways. Front. Neurol. 2018, 9, 1069. [Google Scholar] [CrossRef] [Green Version]
  77. Jakubiak, P.; Cantrill, C.; Urtti, A.; Alvarez-Sánchez, R. Establishment of an In vitro-In vivo Correlation for Melanin Binding and the Extension of the Ocular Half-Life of Small-Molecule Drugs. Mol. Pharm. 2019, 16, 4890–4901. [Google Scholar] [CrossRef]
  78. Tangri, P.; Khurana, S. Basics of ocular drug delivery systems. Int. J. Res. Pharm. Biomed. Sci. 2011, 2, 1541–1552. [Google Scholar]
  79. Rimpelä, A.K.; Reinisalo, M.; Hellinen, L.; Grazhdankin, E.; Kidron, H.; Urtti, A.; Del Amo, E.M. Implications of melanin binding in ocular drug delivery. Adv. Drug Deliv. Rev. 2018, 126, 23–43. [Google Scholar] [CrossRef]
  80. Rimpelä, A.K.; Hagström, M.; Kidron, H.; Urtti, A. Melanin targeting for intracellular drug delivery: Quantification of bound and free drug in retinal pigment epithelial cells. J. Control Release 2018, 283, 261–268. [Google Scholar] [CrossRef] [PubMed]
  81. Achouri, D.; Alhanout, K.; Piccerelle, P.; Andrieu, V. Recent advances in ocular drug delivery. Drug Dev. Ind. Pharm. 2013, 39, 1599–1617. [Google Scholar] [CrossRef] [PubMed]
  82. Dubald, M.; Bourgeois, S.; Andrieu, V.; Fessi, H. Ophthalmic Drug Delivery Systems for Antibiotherapy—A Review. Pharmaceutics 2018, 10, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Seyfoddin, A.; Shaw, J.; Al-Kassas, R. Solid lipid nanoparticles for ocular drug delivery. Drug Deliv. 2010, 7, 467–489. [Google Scholar] [CrossRef]
  84. Ali, J.; Fazil, M.; Qumbar, M.; Khan, N.; Ali, A. Colloidal drug delivery system: Amplify the ocular delivery. Drug Deliv. 2014, 23, 700–716. [Google Scholar]
  85. Hejtmancik, J.F.; Shiels, A. Overview of the Lens. Prog. Mol. Biol. Transl. Sci. 2015, 134, 119–127. [Google Scholar]
  86. Ruan, X.; Liu, Z.; Luo, L.; Liu, Y. The structure of the lens and its associations with the visual quality. BMJ Open Ophthalmol. 2020, 5, e000459. [Google Scholar] [CrossRef]
  87. Coudrillier, B.; Pijanka, J.; Jefferys, J.; Sorensen, T.; Quigley, H.A.; Boote, C.; Nguyen, T.D. Collagen structure and mechanical properties of the human sclera: Analysis for the effects of age. J. Biomech. Eng. 2015, 137, 410061–4100614. [Google Scholar] [CrossRef] [Green Version]
  88. Alshaikh, R.A.; Waeber, C.; Ryan, K.B. Polymer based sustained drug delivery to the ocular posterior segment: Barriers and future opportunities for the treatment of neovascular pathologies. Adv. Drug Deliv. Rev. 2022, 187, 114342. [Google Scholar] [CrossRef]
  89. Yadav, D.; Varma, L.T.; Yadav, K. Drug Delivery to Posterior Segment of the Eye: Conventional Delivery Strategies, Their Barriers, and Restrictions. In Drug Delivery for the Retina and Posterior Segment Disease, 1st ed.; Patel, J.K., Sutariya, V., Kanwar, J.R., Pathak, Y.V., Eds.; Springer: Cham, Switzerland, 2018; pp. 51–67. [Google Scholar]
  90. Djigo, A.D.; Bérubé, J.; Landreville, S.; Proulx, S. Characterization of a tissue-engineered choroid. Acta Biomater. 2019, 84, 305–316. [Google Scholar] [CrossRef]
  91. Hurley, J.B. Retina Metabolism and Metabolism in the Pigmented Epithelium: A Busy Intersection. Annu. Rev. Vis. Sci. 2021, 7, 665–692. [Google Scholar] [CrossRef] [PubMed]
  92. Del Amo, E.M.; Rimpelä, A.K.; Heikkinen, E.; Kari, O.K.; Ramsay, E.; Lajunen, T.; Schmitt, M.; Pelkonen, L.; Bhattacharya, M.; Richardson, D.; et al. Pharmacokinetic aspects of retinal drug delivery. Prog. Retin. Eye Res. 2017, 57, 134–185. [Google Scholar] [CrossRef]
  93. Naylor, A.; Hopkins, A.; Hudson, N.; Campbell, M. Tight Junctions of the Outer Blood Retina Barrier. Int. J. Mol. Sci. 2019, 21, 211. [Google Scholar] [CrossRef] [Green Version]
  94. Willermain, F.; Libert, S.; Motulsky, E.; Salik, D.; Caspers, L.; Perret, J.; Delporte, C. Origins and consequences of hyperosmolar stress in retinal pigmented epithelial cells. Front. Physiol. 2014, 5, 199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Díaz-Coránguez, M.; Ramos, C.; Antonetti, D.A. The inner blood-retinal barrier: Cellular basis and development. Vision Res. 2017, 139, 123–137. [Google Scholar] [CrossRef]
  96. Asencio-Duran, M.; Dabad-Moreno, J.; Vicandi-Plaza, B.; Muñoz, M.; Capote-Díez, M.; Santisteban, G. The Vitreous Body and Its Role in the Diagnosis of Eye Pathologies. Med. Res. Arch. 2021, 9, 9. [Google Scholar] [CrossRef]
  97. Mishra, D.; Gade, S.; Glover, K.; Sheshala, R.; Singh, T.R.R. Vitreous Humor: Composition, Characteristics and Implication on Intravitreal Drug Delivery. Curr. Eye Res. 2022, 48, 208–218. [Google Scholar] [CrossRef]
  98. Stevens, S. Administering a subconjunctival injection. Community Eye Health J. 2009, 22, 15. [Google Scholar]
  99. Rafiei, F.; Tabesh, H.; Farzad, F. Sustained subconjunctival drug delivery systems: Current trends and future perspectives. Int. Ophthalmol. 2020, 40, 2385–2401. [Google Scholar] [CrossRef]
  100. Nebbioso, M.; Livani, M.L.; Santamaria, V.; Librando, A.; Sepe, M. Intracameral lidocaine as supplement to classic topical anesthesia for relieving ocular pain in cataract surgery. Int. J. Ophthalmol. 2018, 11, 1932–1935. [Google Scholar]
  101. Alghamdi, E.A.S.; Al Qahtani, A.Y.; Sinjab, M.M.; Alyahya, K.M. Intracameral Injections. In Extemporaneous Ophthalmic Preparations, 1st ed.; Alghamdi, E.A.S., Al Qahtani, A.Y., Sinjab, M.M., Alyahya, K.M., Eds.; Springer: Cham, Switzerland, 2020; pp. 67–68. [Google Scholar]
  102. Shah, T.J.; Conway, M.D.; Peyman, G.A. Intracameral dexamethasone injection in the treatment of cataract surgery induced inflammation: Design, development, and place in therapy. Clin. Ophthalmol. 2018, 12, 2223–2235. [Google Scholar] [CrossRef] [Green Version]
  103. Adrianto, M.F.; Annuryanti, F.; Wilson, C.G.; Sheshala, R.; Thakur, R.R.S. In vitro dissolution testing models of ocular implants for posterior segment drug delivery. Drug Deliv. Transl. Res. 2022, 12, 1355–1375. [Google Scholar] [CrossRef] [PubMed]
  104. Marashi, A.; Zazo, A. Suprachoroidal injection of triamcinolone acetonide using a custom-made needle to treat diabetic macular edema post pars plana vitrectomy: A case series. J. Int. Med. Res. 2022, 50, 1–10. [Google Scholar] [CrossRef]
  105. Chiang, B.; Jung, J.H.; Prausnitz, M.R. The suprachoroidal space as a route of administration to the posterior segment of the eye. Adv. Drug Deliv. Rev. 2018, 126, 58–66. [Google Scholar] [CrossRef]
  106. Martin, D.F. Evolution of Intravitreal Therapy for Retinal Diseases-From CMV to CNV: The LXXIV Edward Jackson Memorial Lecture. Am. J. Ophthalmol. 2018, 191, xli–lviii. [Google Scholar] [CrossRef] [PubMed]
  107. Gorantla, S.; Rapalli, V.K.; Waghule, T.; Singh, P.P.; Dubey, S.K.; Saha, R.N.; Singhvi, G. Nanocarriers for ocular drug delivery: Current status and translational opportunity. RSC Adv. 2020, 10, 27835–27855. [Google Scholar] [CrossRef]
  108. Peng, C.; Kuang, L.; Zhao, J.; Ross, A.E.; Wang, Z.; Ciolino, J.B. Bibliometric and visualized analysis of ocular drug delivery from 2001 to 2020. J. Control Release. 2022, 345, 625–645. [Google Scholar] [CrossRef]
  109. Das, B.; Nayak, A.K.; Mallick, S. Lipid-based nanocarriers for ocular drug delivery: An updated review. J. Drug Deliv. Sci. Technol. 2022, 76, 103780. [Google Scholar] [CrossRef]
  110. Shahraeini, S.S.; Akbari, J.; Saeedi, M.; Morteza-Semnani, K.; Abootorabi, S.; Dehghanpoor, M.; Rostamkalaei, S.S.; Nokhodchi, A. Atorvastatin Solid Lipid Nanoparticles as a Promising Approach for Dermal Delivery and an Anti-inflammatory Agent. AAPS PharmSciTech. 2020, 21, 263. [Google Scholar] [CrossRef] [PubMed]
  111. Essaghraoui, A.; Belfkira, A.; Hamdaoui, B.; Nunes, C.; Lima, S.A.C.; Reis, S. Improved Dermal Delivery of Cyclosporine A Loaded in Solid Lipid Nanoparticles. Nanomaterials 2019, 9, 1204. [Google Scholar] [CrossRef] [Green Version]
  112. Kakkar, S.; Singh, M.; Mohan Karuppayil, S.; Raut, J.S.; Giansanti, F.; Papucci, L.; Schiavone, N.; Nag, T.C.; Gao, N.; Yu, F.X.; et al. Lipo-PEG nano-ocular formulation successfully encapsulates hydrophilic fluconazole and traverses corneal and non-corneal path to reach posterior eye segment. J. Drug Target 2021, 29, 631–650. [Google Scholar] [CrossRef]
  113. Gómez-Aguado, I.; Rodríguez-Castejón, J.; Beraza-Millor, M.; Vicente-Pascual, M.; Rodríguez-Gascón, A.; Garelli, S.; Battaglia, L.; del Pozo-Rodríguez, A.; Solinís, M.Á. mRNA-Based Nanomedicinal Products to Address Corneal Inflammation by Interleukin-10 Supplementation. Pharmaceutics 2021, 13, 1472. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, J.L.; Hanafy, M.S.; Xu, H.; Leal, J.; Zhai, Y.; Ghosh, D.; Williams, R.O., III; Smyth, H.D.C.; Cui, Z. Aerosolizable siRNA-encapsulated solid lipid nanoparticles prepared by thin-film freeze-drying for potential pulmonary delivery. Int. J. Pharm. 2021, 596, 120215. [Google Scholar] [CrossRef] [PubMed]
  115. Elbrink, K.; Van Hees, S.; Chamanza, R.; Roelant, D.; Loomans, T.; Holm, R.; Kiekens, F. Application of solid lipid nanoparticles as a long-term drug delivery platform for intramuscular and subcutaneous administration: In vitro and in vivo evaluation. Eur. J. Pharm. Biopharm. 2021, 163, 158–170. [Google Scholar] [CrossRef]
  116. Khanna, K.; Sharma, N.; Rawat, S.; Khan, N.; Karwasra, R.; Hasan, N.; Kumar, A.; Jain, G.K.; Nishad, D.K.; Khanna, S.; et al. Intranasal solid lipid nanoparticles for management of pain: A full factorial design approach, characterization & Gamma Scintigraphy. Chem. Phys. Lipids 2021, 236, 105060. [Google Scholar] [CrossRef]
  117. Parvez, S.; Yadagiri, G.; Gedda, M.R.; Singh, A.; Singh, O.P.; Verma, A.; Sundar, S.; Mudavath, S.L. Modified solid lipid nanoparticles encapsulated with Amphotericin B and Paromomycin: An effective oral combination against experimental murine visceral leishmaniasis. Sci. Rep. 2020, 10, 12243. [Google Scholar] [CrossRef]
  118. Angelova, A.; Angelov, B. Dual and multi-drug delivery nanoparticles towards neuronal survival and synaptic repair. Neural Regen Res. 2017, 12, 886–889. [Google Scholar] [CrossRef]
  119. Sahoo, R.K.; Biswas, N.; Guha, A.; Sahoo, N.; Kuotsu, K. Nonionic surfactant vesicles in ocular delivery: Innovative approaches and perspectives. Biomed Res. Int. 2014, 2014, 263604. [Google Scholar] [CrossRef] [Green Version]
  120. Youssef, A.A.A.; Dudhipala, N.; Majumdar, S. Dual Drug Loaded Lipid Nanocarrier Formulations for Topical Ocular Applications. Int. J. Nanomed. 2022, 17, 2283–2299. [Google Scholar] [CrossRef] [PubMed]
  121. Alvi, M.M.; Chatterjee, P. A prospective analysis of co-processed non-ionic surfactants in enhancing permeability of a model hydrophilic drug. AAPS PharmSciTech. 2014, 15, 339–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Jacob, S.; Nair, A.B.; Shah, J.; Gupta, S.; Boddu, S.H.S.; Sreeharsha, N.; Joseph, A.; Shinu, P.; Morsy, M.A. Lipid Nanoparticles as a Promising Drug Delivery Carrier for Topical Ocular Therapy—An Overview on Recent Advances. Pharmaceutics 2022, 14, 533. [Google Scholar] [CrossRef] [PubMed]
  123. Malvajerd, S.S.; Azadi, A.; Izadi, Z.; Kurd, M.; Dara, T.; Dibaei, M.; Zadeh, M.S.; Javar, H.A.; Hamidi, M. Brain delivery of curcumin using solid lipid nanoparticles and nanostructured lipid carriers: Preparation, optimization, and pharmacokinetic evaluation. ACS Chem. Neurosci. 2019, 10, 728–739. [Google Scholar] [CrossRef]
  124. Nagai, N.; Ogata, F.; Otake, H.; Nakazawa, Y.; Kawasaki, N. Energy-dependent endocytosis is responsible for drug transcorneal penetration following the instillation of ophthalmic formulations containing indomethacin nanoparticles. Int. J. Nanomedicine 2019, 14, 1213–1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. González-Fernández, F.M.; Bianchera, A.; Gasco, P.; Nicoli, S.; Pescina, S. Lipid-Based Nanocarriers for Ophthalmic Administration: Towards Experimental Design Implementation. Pharmaceutics 2021, 13, 447. [Google Scholar] [CrossRef]
  126. Amrite, A.C.; Edelhauser, H.F.; Singh, S.R.; Kompella, U.B. Effect of circulation on the disposition and ocular tissue distribution of 20 nm nanoparticles after periocular administration. Mol. Vis. 2008, 14, 150–160. [Google Scholar]
  127. Niamprem, P.; Srinivas, S.P.; Tiyaboonchai, W. Penetration of Nile red-loaded nanostructured lipid carriers (NLCs) across the porcine cornea. Colloids Surf. B Biointerfaces. 2019, 176, 371–378. [Google Scholar] [CrossRef]
  128. Scioli Montoto, S.; Muraca, G.; Ruiz, M.E. Solid Lipid Nanoparticles for Drug Delivery: Pharmacological and Biopharmaceutical Aspects. Front. Mol. Biosci. 2020, 7, 587997. [Google Scholar] [CrossRef] [PubMed]
  129. Duan, Y.; Dhar, A.; Patel, C.; Khimani, M.; Neogi, S.; Sharma, P.; Kumar, N.S.; Vekariya, R.L. A brief review on solid lipid nanoparticles: Part and parcel of contemporary drug delivery systems. RSC Adv. 2020, 10, 26777–26791. [Google Scholar] [CrossRef] [PubMed]
  130. Attama, A.A.; Momoh, M.A.; Builders, P.F. Lipid Nanoparticulate Drug Delivery Systems: A Revolution in Dosage Form Design and Development. In Recent Advances in Novel Drug Carrier Systems; Sezer, A.D., Ed.; IntechOpen: London, UK, 2012; pp. 107–140. [Google Scholar]
  131. Gordillo-Galeano, A.; Mora-Huertas, C.E. Solid lipid nanoparticles and nanostructured lipid carriers: A review emphasizing on particle structure and drug release. Eur. J. Pharm. Biopharm. 2018, 133, 285–308. [Google Scholar] [CrossRef]
  132. Boonme, P.; Souto, E.B.; Wuttisantikul, N.; Jongjit, T.; Pichayakorn, W. Influence of lipids on the properties of solid lipid nanoparticles from microemulsion technique. Eur. J. Lipid Sci. Technol. 2013, 115, 820–824. [Google Scholar] [CrossRef]
  133. Paliwal, R.; Rai, S.; Vaidya, B.; Khatri, K.; Goyal, A.K.; Mishra, N.; Mehta, A.; Vyas, S.P. Effect of lipid core material on characteristics of solid lipid nanoparticles designed for oral lymphatic delivery. Nanomedicine 2009, 5, 184–191. [Google Scholar] [CrossRef] [PubMed]
  134. Cavendish, M.; Nalone, L.; Barbosa, T.; Barbosa, R.; Costa, S.; Nunes, R.; da Silva, C.F.; Chaud, M.V.; Souto, E.B.; Hollanda, L.; et al. Study of pre-formulation and development of solid lipid nanoparticles containing perillyl alcohol. J. Therm. Anal. Calorim. 2019, 141, 767–774. [Google Scholar] [CrossRef]
  135. Hernández-Esquivel, R.-A.; Navarro-Tovar, G.; Zárate-Hernández, E.; Aguirre-Bañuelos, P. Solid Lipid Nanoparticles (SLN). In Nanocomposite Materials for Biomedical and Energy Storage Applications, 1st ed.; Sharma, A., Ed.; IntechOpen: London, UK, 2022; pp. 1–27. [Google Scholar]
  136. Balamurugan, K.; Chintamani, P. Lipid nano particulate drug delivery: An overview of the emerging trend. Pharma Innov. J. 2018, 7, 779–789. [Google Scholar]
  137. Müller, R.H.; Radtke, M.; Wissing, S.A. Solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) in cosmetic and dermatological preparations. Adv. Drug Deliv. Rev. 2002, 54, S131–S155. [Google Scholar] [CrossRef]
  138. Sumera; Anwar, A.; Ovais, M.; Khan, A.; Raza, A. Docetaxel-loaded solid lipid nanoparticles: A novel drug delivery system. IET Nanobiotechnol. 2017, 11, 621–629. [Google Scholar] [CrossRef]
  139. Aguirre-Ramírez, M.; Silva-Jiménez, H.; Banat, I.M.; Díaz De Rienzo, M.A. Surfactants: Physicochemical interactions with biological macromolecules. Biotechnol. Lett. 2021, 43, 523–535. [Google Scholar] [CrossRef] [PubMed]
  140. Nguyen, T.-T.-L.; Duong, V.-A. Solid Lipid Nanoparticles. Encyclopedia 2022, 2, 952–973. [Google Scholar] [CrossRef]
  141. Silva, A.; Martins-Gomes, C.; Coutinho, T.; Fangueiro, J.; Sanchez-Lopez, E.; Pashirova, T.; Andreani, T.; Souto, E.B. Soft Cationic Nanoparticles for Drug Delivery: Production and Cytotoxicity of Solid Lipid Nanoparticles (SLNs). Appl. Sci. 2019, 9, 4438. [Google Scholar] [CrossRef] [Green Version]
  142. Amis, T.M.; Renukuntla, J.; Bolla, P.K.; Clark, B.A. Selection of Cryoprotectant in Lyophilization of Progesterone-Loaded Stearic Acid Solid Lipid Nanoparticles. Pharmaceutics 2020, 12, 892. [Google Scholar] [CrossRef]
  143. Dhiman, N.; Awasthi, R.; Sharma, B.; Kharkwal, H.; Kulkarni, G.T. Lipid Nanoparticles as Carriers for Bioactive Delivery. Front. Chem. 2021, 9, 580118. [Google Scholar] [CrossRef]
  144. Siram, K.; Karuppaiah, A.; Gautam, M.; Sankar, V. Fabrication of Hyaluronic Acid Surface Modified Solid Lipid Nanoparticles Loaded with Imatinib Mesylate for Targeting Human Breast Cancer MCF-7 Cells. J. Clust. Sci. 2022; in press. [Google Scholar]
  145. Kuo, Y.-C.; Chao, I.-W. Conjugation of melanotransferrin antibody on solid lipid nanoparticles for mediating brain cancer malignancy. Biotechnol. Prog. 2015, 32, 480–490. [Google Scholar] [CrossRef] [PubMed]
  146. Onugwu, A.L.; Attama, A.A.; Nnamani, P.O.; Onugwu, S.O.; Onuigbo, E.B.; Khutoryanskiy, V.V. Development and optimization of solid lipid nanoparticles coated with chitosan and poly(2-ethyl-2-oxazoline) for ocular drug delivery of ciprofloxacin. J. Drug Deliv. Sci.Technol. 2022, 74, 103527. [Google Scholar] [CrossRef]
  147. Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. 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, 183. [Google Scholar] [CrossRef] [PubMed]
  149. Dang, H.; Dong, C.; Zhang, L. Sustained latanoprost release from PEGylated solid lipid nanoparticle-laden soft contact lens to treat glaucoma. Pharm. Dev. Technol. 2022, 27, 127–133. [Google Scholar] [CrossRef] [PubMed]
  150. Sun, K.; Hu, K. Preparation and Characterization of Tacrolimus-Loaded SLNs in situ Gel for Ocular Drug Delivery for the Treatment of Immune Conjunctivitis. Drug Des. Devel. Ther. 2021, 15, 141–150. [Google Scholar] [CrossRef]
  151. El-Emam, G.A.; Girgis, G.N.; Hamed, M.F.; El-Azeem Soliman, O.A.; Abd El Gawad, A.E.G.H. Formulation and Pathohistological Study of Mizolastine–Solid Lipid Nanoparticles–Loaded Ocular Hydrogels. Int. J. Nanomed. 2021, 16, 7775–7799. [Google Scholar] [CrossRef]
  152. Carbone, C.; Fuochi, V.; Zielińska, A.; Musumeci, T.; Souto, E.B.; Bonaccorso, A.; Puglia, C.; Petronio, G.P.; Furneri, P.M. Dual-drugs delivery in solid lipid nanoparticles for the treatment of Candida albicans mycosis. Colloids Surf. B Biointerfaces 2020, 186, 110705. [Google Scholar] [CrossRef]
  153. Liang, Z.; Zhang, Z.; Yang, J.; Lu, P.; Zhou, T.; Li, J.; Zhang, J. Assessment to the Antifungal Effects in vitro and the Ocular Pharmacokinetics of Solid-Lipid Nanoparticle in Rabbits. Int. J. Nanomed. 2021, 16, 7847–7857. [Google Scholar] [CrossRef]
  154. Khames, A.; Khaleel, M.A.; El-Badawy, M.F.; El-Nezhawy, A.O.H. Natamycin solid lipid nanoparticles—Sustained ocular delivery system of higher corneal penetration against deep fungal keratitis: Preparation and optimization. Int. J. Nanomedicine 2019, 14, 2515–2531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Singh, M.; Guzman-Aranguez, A.; Hussain, A.; Srinivas, C.S.; Kaur, I.P. Solid lipid nanoparticles for ocular delivery of isoniazid: Evaluation, proof of concept and in vivo safety & kinetics. Nanomedicine 2019, 14, 465–491. [Google Scholar] [PubMed]
  156. Nair, A.B.; Shah, J.; Al-Dhubiab, B.E.; Jacob, S.; Patel, S.S.; Venugopala, K.N.; Morsy, M.A.; Gupta, S.; Attimarad, M.; Sreeharsha, N.; et al. Clarithromycin Solid Lipid Nanoparticles for Topical Ocular Therapy: Optimization, Evaluation and In Vivo Studies. Pharmaceutics 2021, 13, 523. [Google Scholar] [CrossRef] [PubMed]
  157. Bonaccorso, A.; Pepe, V.; Zappulla, C.; Cimino, C.; Pricoco, A.; Puglisi, G.; Giuliano, F.; Pignatello, R.; Carbone, C. Sorafenib Repurposing for Ophthalmic Delivery by Lipid Nanoparticles: A Preliminary Study. Pharmaceutics 2021, 13, 1956. [Google Scholar] [CrossRef]
  158. Yadav, M.; Schiavone, N.; Guzman-Aranguez, A.; Giansanti, F.; Papucci, L.; Perez de Lara, M.J.; Singh, M.; Kaur, I.P. Atorvastatin-loaded solid lipid nanoparticles as eye drops: Proposed treatment option for age-related macular degeneration (AMD). Drug Deliv. Transl. Res. 2020, 10, 919–944. [Google Scholar] [CrossRef] [PubMed]
  159. Cheng, Z.; Li, Y.; Wang, K.; Zhu, X.; Tharkar, P.; Shu, W.; Zhang, T.; Zeng, S.; Zhu, L.; Murray, M.; et al. Compritol solid lipid nanoparticle formulations enhance the protective effect of betulinic acid derivatives in human Müller cells against oxidative injury. Exp. Eye Res. 2022, 215, 108906. [Google Scholar] [CrossRef]
  160. Ahmad, I.; Pandit, J.; Sultana, Y.; Mishra, A.K.; Hazari, P.P.; Aqil, M. Optimization by design of etoposide loaded solid lipid nanoparticles for ocular delivery: Characterization, pharmacokinetic and deposition study. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 100, 959–970. [Google Scholar] [CrossRef]
  161. Freitas, L.G.A.; Isaac, D.L.C.; Lima, E.M.; Souza, L.G.; Abud, M.A.; Reis, R.G.D.; Tannure, W.T.; Ávila, M.P. Retinal changes in rabbit after intravitreal injection of sunitinib encapsulated into solid lipid nanoparticles and polymeric nanocapsules. Arq. Bras. Oftalmol. 2018, 81, 408–413. [Google Scholar] [CrossRef]
  162. Wang, F.Z.; Zhang, M.W.; Zhang, D.S.; Huang, Y.; Chen, L.; Jiang, S.M.; Shi, K.; Li, R. Preparation, optimization, and characterization of chitosan-coated solid lipid nanoparticles for ocular drug delivery. J. Biomed. Res. 2018, 32, 411–423. [Google Scholar]
  163. Taskar, P.S.; Patil, A.; Lakhani, P.; Ashour, E.; Gul, W.; ElSohly, M.A.; Murphy, B.; Majumdar, S. Δ9-Tetrahydrocannabinol Derivative-Loaded Nanoformulation Lowers Intraocular Pressure in Normotensive Rabbits. Transl. Vis. Sci. Technol. 2019, 8, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Wang, J.; Zhao, F.; Liu, R.; Chen, J.; Zhang, Q.; Lao, R.; Wang, Z.; Jin, X.; Liu, C. Novel cationic lipid nanoparticles as an ophthalmic delivery system for multicomponent drugs: Development, characterization, in vitro permeation, in vivo pharmacokinetic, and molecular dynamics studies. Int. J. Nanomed. 2017, 12, 8115–8127. [Google Scholar] [CrossRef] [Green Version]
  165. Gomes Souza, L.; Antonio Sousa-Junior, A.; Alves Santana Cintra, B.; Vieira Dos Anjos, J.L.; Leite Nascimento, T.; Palmerston Mendes, L.; de Souza Vieira, M.; do Nascimento Ducas, R.; Campos Valadares, M.; Antônio Mendanha, S.; et al. Pre-clinical safety of topically administered sunitinib-loaded lipid and polymeric nanocarriers targeting corneal neovascularization. Int. J. Pharm. 2023, 635, 122682. [Google Scholar] [CrossRef]
  166. Jaiswal, P.; Gidwani, B.; Vyas, A. Nanostructured lipid carriers and their current application in targeted drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 27–40. [Google Scholar] [CrossRef]
  167. Dhiman, S.; Mishra, N.; Sharma, S. Development of PEGylated solid lipid nanoparticles of pentoxifylline for their beneficial pharmacological potential in pathological cardiac hypertrophy. Artif. Cells Nanomed. Biotechnol. 2016, 44, 1901–1908. [Google Scholar] [CrossRef] [Green Version]
  168. Elmowafy, M.; Al-Sanea, M.M. Nanostructured lipid carriers (NLCs) as drug delivery platform: Advances in formulation and delivery strategies. Saudi Pharm. J. 2021, 29, 999–1012. [Google Scholar] [CrossRef] [PubMed]
  169. Shahzadi, I.; Fürst, A.; Knoll, P.; Bernkop-Schnürch, A. Nanostructured Lipid Carriers (NLCs) for Oral Peptide Drug Delivery: About the Impact of Surface Decoration. Pharmaceutics 2021, 13, 1312. [Google Scholar] [CrossRef] [PubMed]
  170. Chauhan, I.; Yasir, M.; Verma, M.; Singh, A.P. Nanostructured Lipid Carriers: A Groundbreaking Approach for Transdermal Drug Delivery. Adv. Pharm. Bull. 2020, 10, 150–165. [Google Scholar] [CrossRef] [PubMed]
  171. Kiss, E.L.; Berkó, S.; Gácsi, A.; Kovács, A.; Katona, G.; Soós, J.; Csányi, E.; Gróf, I.; Harazin, A.; Deli, M.A.; et al. Design and Optimization of Nanostructured Lipid Carrier Containing Dexamethasone for Ophthalmic Use. Pharmaceutics 2019, 11, 679. [Google Scholar] [CrossRef] [Green Version]
  172. El-Salamouni, N.S.; Farid, R.M.; El-Kamel, A.H.; El-Gamal, S.S. Effect of sterilization on the physical stability of brimonidine-loaded solid lipid nanoparticles and nanostructured lipid carriers. Int. J. Pharm. 2015, 496, 976–983. [Google Scholar] [CrossRef]
  173. Apostolou, M.; Assi, S.; Fatokun, A.A.; Khan, I. The Effects of Solid and Liquid Lipids on the Physicochemical Properties of Nanostructured Lipid Carriers. J. Pharm. Sci. 2021, 110, 2859–2872. [Google Scholar] [CrossRef]
  174. Malik, D.S.; Kaur, G. Nanostructured gel for topical delivery of azelaic acid: Designing, characterization, and in-vitro evaluation. J. Drug Deliv. Sci. Technol. 2018, 47, 123–136. [Google Scholar] [CrossRef]
  175. Bang, K.-H.; Na, Y.-G.; Huh, H.W.; Hwang, S.-J.; Kim, M.-S.; Kim, M.; Lee, H.-K.; Cho, C.-W. The Delivery Strategy of Paclitaxel Nanostructured Lipid Carrier Coated with Platelet Membrane. Cancers 2019, 11, 807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Cao, C.; Wang, Q.; Liu, Y. Lung cancer combination therapy: Doxorubicin and β-elemene co-loaded, pH-sensitive nanostructured lipid carriers. Drug Des. Devel. Ther. 2019, 13, 1087–1098. [Google Scholar] [CrossRef] [Green Version]
  177. Javed, S.; Mangla, B.; Almoshari, Y.; Sultan, M.; Ahsan, W. Nanostructured lipid carrier system: A compendium of their formulation development approaches, optimization strategies by quality by design, and recent applications in drug delivery. Nanotechnol. Rev. 2022, 11, 1744–1777. [Google Scholar] [CrossRef]
  178. Haider, M.; Abdin, S.M.; Kamal, L.; Orive, G. Nanostructured Lipid Carriers for Delivery of Chemotherapeutics: A Review. Pharmaceutics 2020, 12, 288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Khosa, A.; Reddi, S.; Saha, R.N. Nanostructured lipid carriers for site-specific drug delivery. Biomed. Pharmacother. 2018, 103, 598–613. [Google Scholar] [CrossRef]
  180. Selvaraj, K.; Kuppusamy, G.; Krishnamurthy, J.; Mahalingam, R.; Singh, S.K.; Gulati, M. Repositioning of Itraconazole for the Management of Ocular Neovascularization Through Surface-Modified Nanostructured Lipid Carriers. Assay Drug Dev. Technol. 2019, 17, 178–190. [Google Scholar] [CrossRef]
  181. Sharma, D.S.; Wadhwa, S.; Gulati, M.; Kumar, B.; Chitranshi, N.; Gupta, V.K.; Alrouji, M.; Alhajlah, S.; AlOmeir, O.; Vishwas, S.; et al. Chitosan modified 5-fluorouracil nanostructured lipid carriers for treatment of diabetic retinopathy in rats: A new dimension to an anticancer drug. Int. J. Biol. Macromol. 2023, 224, 810–830. [Google Scholar] [CrossRef] [PubMed]
  182. Fu, T.; Yi, J.; Lv, S.; Zhang, B. Ocular amphotericin B delivery by chitosan-modified nanostructured lipid carriers for fungal keratitis-targeted therapy. J. Liposome Res. 2017, 27, 228–233. [Google Scholar] [CrossRef]
  183. Pai, R.V.; Vavia, P.R. Chitosan oligosaccharide enhances binding of nanostructured lipid carriers to ocular mucins: Effect on ocular disposition. Int. J. Pharm. 2020, 577, 119095. [Google Scholar] [CrossRef] [PubMed]
  184. Li, J.; Jin, X.; Yang, Y.; Zhang, L.; Liu, R.; Li, Z. Trimethyl chitosan nanoparticles for ocular baicalein delivery: Preparation, optimization, in vitro evaluation, in vivo pharmacokinetic study and molecular dynamics simulation. Int. J. Biol. Macromol. 2020, 156, 749–761. [Google Scholar] [CrossRef]
  185. Tan, G.; Li, J.; Song, Y.; Yu, Y.; Liu, D.; Pan, W. Phenylboronic acid-tethered chondroitin sulfate-based mucoadhesive nanostructured lipid carriers for the treatment of dry eye syndrome. Acta Biomater. 2019, 99, 350–362. [Google Scholar] [CrossRef]
  186. Zhu, R.; Chen, W.; Gu, D.; Wang, T.; Li, J.; Pan, H. Chondroitin sulfate and L-Cysteine conjugate modified cationic nanostructured lipid carriers: Pre-corneal retention, permeability, and related studies for dry eye treatment. Int. J. Biol. Macromol. 2023, 228, 624–637. [Google Scholar] [CrossRef]
  187. Abdelhakeem, E.; El-Nabarawi, M.; Shamma, R. Effective Ocular Delivery of Eplerenone Using Nanoengineered Lipid Carriers in Rabbit Model. Int. J. Nanomed. 2021, 16, 4985–5002. [Google Scholar] [CrossRef]
  188. Yan, T.; Ma, Z.; Liu, J.; Yin, N.; Lei, S.; Zhang, X.; Li, X.; Zhang, Y.; Kong, J. Thermoresponsive Genistein NLC-dexamethasone-moxifloxacin multi drug delivery system in lens capsule bag to prevent complications after cataract surgery. Sci. Rep. 2021, 11, 181. [Google Scholar] [CrossRef] [PubMed]
  189. Tavakoli, N.; Taymouri, S.; Saeidi, A.; Akbari, V. Thermosensitive hydrogel containing sertaconazole loaded nanostructured lipid carriers for potential treatment of fungal keratitis. Pharm. Dev. Technol. 2019, 24, 891–901. [Google Scholar] [CrossRef] [PubMed]
  190. Yu, Y.; Feng, R.; Li, J.; Wang, Y.; Song, Y.; Tan, G.; Liu, D.; Liu, W.; Yang, X.; Pan, H.; et al. A hybrid genipin-crosslinked dual-sensitive hydrogel/nanostructured lipid carrier ocular drug delivery platform. Asian J. Pharm. Sci. 2019, 14, 423–434. [Google Scholar] [CrossRef] [PubMed]
  191. Yu, Y.; Xu, S.; Yu, S.; Li, J.; Tan, G.; Li, S.; Pan, W. A Hybrid Genipin-Cross-Linked Hydrogel/Nanostructured Lipid Carrier for Ocular Drug Delivery: Cellular, ex Vivo, and in Vivo Evaluation. ACS Biomater. Sci. Eng. 2020, 6, 1543–1552. [Google Scholar] [CrossRef]
  192. Chen, L.; Wu, R. Brinzolamide- and latanoprost-loaded nano lipid carrier prevents synergistic retinal damage in glaucoma. Acta Biochim. Pol. 2022, 69, 423–428. [Google Scholar] [CrossRef]
  193. Varela-Fernández, R.; García-Otero, X.; Díaz-Tomé, V.; Regueiro, U.; López-López, M.; González-Barcia, M.; Isabel Lema, M.; Javier Otero-Espinar, F. Lactoferrin-loaded nanostructured lipid carriers (NLCs) as a new formulation for optimized ocular drug delivery. Eur. J. Pharm. Biopharm. 2022, 172, 144–156. [Google Scholar] [CrossRef]
  194. Kumari, S.; Dandamudi, M.; Rani, S.; Behaeghel, E.; Behl, G.; Kent, D.; O’Reilly, N.J.; O’Donovan, O.; McLoughlin, P.; Fitzhenry, L. Dexamethasone-Loaded Nanostructured Lipid Carriers for the Treatment of Dry Eye Disease. Pharmaceutics 2021, 13, 905. [Google Scholar] [CrossRef] [PubMed]
  195. Zahir-Jouzdani, F.; Khonsari, F.; Soleimani, M.; Mahbod, M.; Arefian, E.; Heydari, M.; Shahhosseini, S.; Dinarvand, R.; Atyabi, F. Nanostructured lipid carriers containing rapamycin for prevention of corneal fibroblasts proliferation and haze propagation after burn injuries: In vitro and in vivo. J. Cell Physiol. 2019, 234, 4702–4712. [Google Scholar] [CrossRef] [PubMed]
  196. Kumar, M.; Tiwari, A.; Asdaq, S.M.B.; Nair, A.B.; Bhatt, S.; Shinu, P.; Al Mouslem, A.K.; Jacob, S.; Alamri, A.S.; Alsanie, W.F.; et al. Itraconazole loaded nano-structured lipid carrier for topical ocular delivery: Optimization and evaluation. Saudi J. Biol. Sci. 2022, 29, 1–10. [Google Scholar] [CrossRef] [PubMed]
  197. Patil, A.; Lakhani, P.; Taskar, P.; Wu, K.W.; Sweeney, C.; Avula, B.; Wang, Y.H.; Khan, I.A.; Majumdar, S. Formulation Development, Optimization, and In Vitro-In Vivo Characterization of Natamycin-Loaded PEGylated Nano-Lipid Carriers for Ocular Applications. J. Pharm. Sci. 2018, 107, 2160–2171. [Google Scholar] [CrossRef] [PubMed]
  198. Li, Q.; Yang, X.; Zhang, P.; Mo, F.; Si, P.; Kang, X.; Wang, M.; Zhang, J. Dasatinib loaded nanostructured lipid carriers for effective treatment of corneal neovascularization. Biomater. Sci. 2021, 9, 2571–2583. [Google Scholar] [CrossRef] [PubMed]
  199. Luo, Q.; Yang, J.; Xu, H.; Shi, J.; Liang, Z.; Zhang, R.; Lu, P.; Pu, G.; Zhao, N.; Zhang, J. Sorafenib-loaded nanostructured lipid carriers for topical ocular therapy of corneal neovascularization: Development, in-vitro and in vivo study. Drug Deliv. 2022, 29, 837–855. [Google Scholar] [CrossRef] [PubMed]
  200. Nirbhavane, P.; Sharma, G.; Singh, B.; Begum, G.; Jones, M.-C.; Rauz, S.; Vincent, R.; Denniston, A.K. Triamcinolone acetonide loaded-cationic nano-lipoidal formulation for uveitis: Evidences of improved biopharmaceutical performance and anti-inflammatory activity. Colloids Surf. B. Biointerfaces 2020, 190, 110902. [Google Scholar] [CrossRef]
  201. 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]
  202. Puglia, C.; Blasi, P.; Ostacolo, C.; Sommella, E.; Bucolo, C.; Platania, C.B.M.; Romano, G.L.; Geraci, F.; Drago, F.; Santonocito, D.; et al. Innovative Nanoparticles Enhance N-Palmitoylethanolamide Intraocular Delivery. Front. Pharmacol. 2018, 9, 285. [Google Scholar] [CrossRef]
  203. Zingale, E.; Rizzo, S.; Bonaccorso, A.; Consoli, V.; Vanella, L.; Musumeci, T.; Spadaro, A.; Pignatello, R. Optimization of Lipid Nanoparticles by Response Surface Methodology to Improve the Ocular Delivery of Diosmin: Characterization and In-Vitro Anti-Inflammatory Assessment. Pharmaceutics 2022, 14, 1961. [Google Scholar] [CrossRef]
  204. Santonocito, D.; Vivero-Lopez, M.; Lauro, M.R.; Torrisi, C.; Castelli, F.; Sarpietro, M.G.; Puglia, C. Design of Nanotechnological Carriers for Ocular Delivery of Mangiferin: Preformulation Study. Molecules 2022, 27, 1328. [Google Scholar] [CrossRef]
  205. González-Fernández, F.M.; Delledonne, A.; Nicoli, S.; Gasco, P.; Padula, C.; Santi, P.; Sissa, C.; Pescina, S. Nanostructured Lipid Carriers for Enhanced Transscleral Delivery of Dexamethasone Acetate: Development, Ex Vivo Characterization and Multiphoton Microscopy Studies. Pharmaceutics 2023, 15, 407. [Google Scholar] [CrossRef] [PubMed]
  206. Khairnar, S.V.; Pagare, P.; Thakre, A.; Nambiar, A.R.; Junnuthula, V.; Abraham, M.C.; Kolimi, P.; Nyavanandi, D.; Dyawanapelly, S. Review on the Scale-Up Methods for the Preparation of Solid Lipid Nanoparticles. Pharmaceutics 2022, 14, 1886. [Google Scholar] [CrossRef] [PubMed]
  207. Duong, V.-A.; Nguyen, T.-T.-L.; Maeng, H.-J. Preparation of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers for Drug Delivery and the Effects of Preparation Parameters of Solvent Injection Method. Molecules 2020, 25, 4781. [Google Scholar] [CrossRef]
  208. Zielińska, A.; Soles, B.B.; Lopes, A.R.; Vaz, B.F.; Rodrigues, C.M.; Alves, T.F.R.; Klensporf-Pawlik, D.; Durazzo, A.; Lucarini, M.; Severino, P.; et al. Nanopharmaceuticals for Eye Administration: Sterilization, Depyrogenation and Clinical Applications. Biology 2020, 9, 336. [Google Scholar] [CrossRef]
  209. Pardeike, J.; Weber, S.; Haber, T.; Wagner, J.; Zarfl, H.P.; Plank, H.; Zimmer, A. Development of an itraconazole-loaded nanostructured lipid carrier (NLC) formulation for pulmonary application. Int. J. Pharm. 2011, 419, 329–338. [Google Scholar] [CrossRef] [PubMed]
  210. Gokce, E.H.; Sandri, G.; Bonferoni, M.C.; Rossi, S.; Ferrari, F.; Güneri, T.; Caramella, C. Cyclosporine A loaded SLNs: Evaluation of cellular uptake and corneal cytotoxicity. Int. J. Pharm. 2008, 364, 76–86. [Google Scholar] [CrossRef]
  211. Youshia, J.; Kamel, A.O.; El Shamy, A.; Mansour, S. Gamma sterilization and in vivo evaluation of cationic nanostructured lipid carriers as potential ocular delivery systems for antiglaucoma drugs. Eur. J. Pharm. Sci. 2021, 163, 105887. [Google Scholar] [CrossRef]
  212. Thi, T.T.H.; Suys, E.J.A.; Lee, J.S.; Nguyen, D.H.; Park, K.D.; Truong, N.P. Lipid-Based Nanoparticles in the Clinic and Clinical Trials: From Cancer Nanomedicine to COVID-19 Vaccines. Vaccines 2021, 9, 359. [Google Scholar] [CrossRef]
  213. Khiev, D.; Mohamed, Z.A.; Vichare, R.; Paulson, R.; Bhatia, S.; Mohapatra, S.; Lobo, G.P.; Valapala, M.; Kerur, N.; Passaglia, C.L.; et al. Emerging Nano-Formulations and Nanomedicines Applications for Ocular Drug Delivery. Nanomaterials 2021, 11, 173. [Google Scholar] [CrossRef]
  214. Buttini, F.; Rozou, S.; Rossi, A.; Zoumpliou, V.; Rekkas, D.M. The application of Quality by Design framework in the pharmaceutical development of dry powder inhalers. Eur. J. Pharm. Sci. 2018, 113, 64–76. [Google Scholar] [CrossRef] [PubMed]
  215. Janagam, D.R.; Wu, L.; Lowe, T.L. Nanoparticles for drug delivery to the anterior segment of the eye. Adv. Drug Deliv. Rev. 2017, 122, 31–64. [Google Scholar] [CrossRef] [PubMed]
Figure 1. An overview of (A) ocular anatomy and routes for administration. (B) Ocular drug delivery barriers. * P-glycoprotein; ** Multidrug-resistant protein; *** Breast cancer resistance protein.
Figure 1. An overview of (A) ocular anatomy and routes for administration. (B) Ocular drug delivery barriers. * P-glycoprotein; ** Multidrug-resistant protein; *** Breast cancer resistance protein.
Pharmaceuticals 16 00474 g001
Figure 2. Different types of solid lipid nanoparticles and nanostructured lipid carriers.
Figure 2. Different types of solid lipid nanoparticles and nanostructured lipid carriers.
Pharmaceuticals 16 00474 g002
Table 1. Alternative routes of ocular drug delivery.
Table 1. Alternative routes of ocular drug delivery.
Alternative RouteSpecificsBenefitsLimitationsReferences
Sub-
conjunctival
(SC)
SC route includes SC injections,
administered in the lower or
upper fornix, as well as
instillation of SC implants;
Clinical indications include corneal/scleral lesions, glaucoma,
cytomegalovirus rhinitis.
Possibility to ensure high local drug concentration;
Improved penetration of water-soluble drugs due to the bypassing of the corneal epithelium.
Conjunctival and choroidal
blood/lymphatic flow;
Temporary pain at the
injection site;
Local irritations.
[98,99]
Intracameral
(IC)
Injections applied in the anterior chamber, often as a
prevention of postoperative
endophthalmitis after
cataract surgery;
Delivery of antibiotics,
steroids,
anesthetics.
Lower drug concentration needed;
Decreased side effects vs. topical steroid application;
Increased anesthesia during surgery when co-administered with topical anesthetics.
Potential complications,
such as toxic anterior segment
syndrome, corneal endothelial
toxicity.
[100,101,102]
TransscleralDrug delivery to the posterior
segment of the eye;
The sclera is thinnest
around the equator,
therefore, it is the preferred
area for injection.
Obviates the corneal
and conjunctival barrier;
Less-invasive procedure compared to intravitreal injections.
Static barriers (sclera, choroid,
retina) and dynamic barriers
(choroidal blood flow) reduce
drug bioavailability;
Necessity of high doses.
[84,99,103]
Supra-choroidal
(SC)
Drug injection under the choroid,
targeting the following areas: choroid and retina;
Microneedles have also been used for drug deposition into the SC space;
Clinical indications include:
posterior uveitis, macular edema.
Obviates the sclera and improves drug bioavailability within the choroid and retina;
Effective for the delivery of small molecules; Lower risk of intraocular pressure spikes.
Choroidal circulation;
Risk of occurrence of
choroidal hemorrhage
or detachment.
[99,104,105]
Intravitreal
(IV)
Direct injection to the vitreous body targeting posterior eye segment;
Drug delivery of vascular
endothelial growth factor
(VEGF) inhibitors, antibiotics,
corticosteroids;
IV injections are applied in the
therapy of age-related macular
degeneration, cytomegalovirus
retinitis,
diabetic macular edema,
retinal vein occlusions.
Bypasses the BRB;
Provides high local therapeutic concentration and prolonged drug levels;
Reduced systemic side effects.
Repetitive instillations lead to
serious ocular complications
and patient non-compliance.
Eye discomfort and
pain were reported following IV injections.
[53,106]
Systemic/OralDrugs are administered orally or
intravenously;
Therapeutic applications include:
scleritis,
cytomegalovirus retinitis.
Acceptance by the patients.Low bioavailability (<2%)—
barrier role of BAB, BRB;
Necessity of high doses,
corresponding to increased
risk of side effects.
[107]
Table 2. Recent progress of SLNs for ophthalmic application (5 years’ overview).
Table 2. Recent progress of SLNs for ophthalmic application (5 years’ overview).
CompositionDrug/DiseaseMethod of PreparationPhysicochemical
Characteristics
ResultsReferences
Tripalmitin
Tween 80
Glycerol
Econazole/
Fungal keratitis
Microemulsion methodSize 19.05 ± 0.28 nm
PDI 0.21 ± 0.01
ζ potential −2.20 ± 0.10 mV
EE = 94.18 ± 1.86%
Slow and controlled drug release (within 96 h);
Improved antifungal activity;
Enhanced bioavailability—drug concentration was above MIC within 3 h after application.
[153]
Precirol
ATO 5
Pluronic F68
Stearyl amine
Natamycin/
Fungal keratitis
Hot emulsification-ultrasonication techniqueSize 42 nm
PDI 0.224
ζ potential 26 mV
EE ≈ 85%
Prolonged drug release
(within 8 h);
Improved corneal penetration;
Superior antifungal activity vs. free drug;
Excellent ocular tolerability.
[154]
Compritol 888
ATO
Stearic acid
Tween 80
Soy lecithin
Isoniazid/
Ocular tuberculosis
Microemulsion methodSize 149.2 ± 4.9 nm
PDI 0.15 ± 0.02
ζ potential −0.35 ± 0.28 mV
EE = 65.2 ± 2.2%
Prolonged drug release (48 h);
Enhanced corneal permeability (1.6 fold);
Improved ocular bioavailability (4.2 fold) vs. drug solution.
[155]
Stearic acid
Tween 80
Transcutol P
Clarithromycin/
Bacterial endophthalmitis
High-speed mixing and the ultrasonication methodSize 157 ± 42.4 nm
PDI 0.13 ± 0.02
ζ potential −17.2 ± 3.1 mV
EE = 81.3 ± 4.6
Sustained drug release
(~80% in 8 h);
Improved transcorneal
permeation and bioavailability compared to drug solution.
[156]
Softisan 100
(Hydrogenated Coco-Glycerides)
Suppocire NB (C10–C18 Triglycerides)
Tween 80
Tegin O
DOTAP
DDAB
Sorafenib/
Uveal melanoma
Phase inversion temperature methodSize 127.85 ± 1.50 nm
PDI 0.215 ± 0.014
ζ potential 20 mV
EE= 75.0 ± 2.1%
Sustained drug release
(less than 25% of encapsulated drug released after 72 h);
Good physical stability, cytocompatibility and mucoadhesive properties of elaborated SLNs.
[157]
Compritol 888ATO
PEG 400
Poloxamer 188
Phospholipon 90H
Atorvastatin/
Age-related macular degeneration
Hot high-pressure homogenizationSize 256.3 ± 10.5 nm
PDI 0.26 ± 0.02
ζ potential −2.65 mV
EE= 73.1 ± 1.52%
Improved bioavailability
(8-fold in aqueous humor and 12-fold in vitreous humor) vs. free drug;
Proven safety in corneal/retinal cell lines;
Successful delivery to the retina, confirmed by intact fluorescein-labeled SLNs.
[158]
Com-
pritol 888 ATO/Compritol HD5 ATO
Pluronic F127
Betulinic acid (BA) derivatives H3, H5 and H7/
Retinal diseases (diabetic retinopathy, age-related macular degeneration, choroidal neovascularization)
Microemulsion methodSize 58.5± 9.8 nm
PDI 0.246
ζ potential 6.45 ± 5.58 mV
EE = 75.10%
Improved drug delivery and enhanced anti-oxidative efficacy of BA derivatives;
Suppressed glutamate-induced ROS production/necrosis in human Müller cells.
[159]
Gelucire 44/14 Compritol ATO 888
Tween 80
Etoposide/
Posterior segment-related diseases (e.g., age-related macular degeneration, diabetic retinopathy)
Melt-
emulsification and ultrasonication
technique
Size 239.43 ± 2.35 nm
PDI 0.261 ± 0.001
EE 80.96 ± 2.21%
Sustained etoposide concentration of etoposide in
vitreous body for
7 days after IV injection
Better toxicological profile vs. etoposide solution.
[160]
Stearic acid
Sodium taurodeoxycholate
Phosphati-
dylcholine
Sutinib
(Sb)/
Retinal diseases (age-related macular degeneration, diabetic retinopathy, retinal vein occlusions)
Microemulsion methodSize 140 nm
PDI 0.20
Excellent tolerability profile based on
in vivo study on 20 albino rabbits; After IV injections, Sb SLNs didn’t cause any abnormalities in ocular morphology in contrast to polymeric nanocapsules.
[161]
Chitosan
Phospholipids (Lipoid S100)
Glyceryl mono-
stearate
Tween 80
PEG 400
Methazolamide/
Glaucoma
Emulsion-solvent evaporation
method
Size 247.7 ± 17.3 nm
PDI
ζ potential 33.5 ± 3.9 mV
EE = 58.5 ± 4.5%
Prolonged drug release compared to drug solution;
Excellent tolerability
and marked reduction in
IOP vs. uncoated
methazolamide SLNs.
[162]
Compritol 888
ATO
Pluronic F68
Tween 80
Glycerol
Δ9
-Tetrahydrocannabinol-valine-hemisuccinate/
Glaucoma
UltrasonicationSize 287.80 ± 7.35 nm
PDI 0.29 ± 0.01
EE = 93.57 ± 4.68%
Greater reduction in the IOP with respect to intensity and duration compared to pilocarpine/timolol maleate eye drops;
High drug concentration in the iris/ciliary body and choroid/
retina.
[163]
Legend: DDAB—Didodecyldimethylammonium bromide; DOTAP—Dioleoyl-trimethylammonium–propane chloride; EE—Entrapment efficiency; IOP—Intraocular pressure; MIC—Minimum inhibitory concentration; PDI—Polydispersity index; ROS—Reactive oxygen species.
Table 3. Recent progress of NLCs for ophthalmic application (5 years’ overview).
Table 3. Recent progress of NLCs for ophthalmic application (5 years’ overview).
CompositionDrug/DiseaseMethod of
Preparation
Physicochemical
Characteristics
ResultsReferences
Glycerol
monostearate 40–55
Soy lecithin
Compritol 888 ATO
Cholesterol
Capryol 90
Miglyol 812 N
Kolliphor P 407
Kolliphor P 188
α-Tocopherol-PEG
Lactoferrin/
Keratoconus
Double
emulsion/
solvent evaporation method.
Size 119.45 ± 11.44 nm
PDI 0.151 ± 0.045
ζ potential 17.50 ± 2.53 mV
EE ≈ 75%
Controlled release profile;
Good physical stability
(up to 3 months);
Muco-adhesive
properties
(for at least 240 min);
Ocular tolerability.
[193]
Labrafac lipophile WL1349
Cholesterol
Tween 80
Dexamethasone
(DXM)/
Dry Eye Disease
Solvent diffusion
method
Size 19.51 ± 0.5 nm
PDI 0.08
ζ potential 9.8 mV
EE = 99.6 ± 0.5%
Cellular internalization
in HCECs and corneal
distribution in ex vivo
porcine cornea;
Significant reduction in inflammatory cytokines (MMP-9, IL-6 and TNF-α) related to DED pathogenesis vs. free DXM.
[194]
Precirol ATO5
Capryol PGMC
Stearylamine
Tween 80
Poloxamer 188
Rapamycin/
Corneal alkaline burn injury
Emulsification solvent diffusion and evaporation methodSize 216 ± 40 nm
ζ potential 14 ± 2.6 mV
EE = 97.66 ± 0.57%
Improved fibroblast uptake of encapsulated cargo via NLCs (1.5 times);
Superior in vivo corneal healing properties of NLCs vs. control groups.
[195]
Stearic acid, oleic acid
Poloxamer 407
Itraconazole/
Fungal keratitis
High-speed homogenization techniqueSize 150.67 nm
ζ potential −28 mV
EE = 94.65%
Ocular safe formulation according to HET−CAM test; Enhanced antifungal
activity of the NLCs
compared to commercial eye drops.
[196]
PrecirolATO 5,Castor oil, Span 80,
mPEG-2K-DSPE sodium salt
Poloxamer 188,
Tween 80, glycerin
Natamycin/
Fungal keratitis
High-pressure homogenizationSize 241.96 nm,
PDI 0.406
EE = 95.35%
Improved in vitro transcorneal permeation and flux of formulated
NT compared to drug
suspension.
[197]
Glycerin monostearate
Miglyol 812 N
Solutol HS 15
Gelucire 44/14
Soy lecithin
Dasatinib
(DAS)/
Corneal neovascularization
Melt-emulsification methodSize 78.53 ± 0.36 nm
PDI 0.21 ± 0.01
ζ potential −29.6 ± 1.0 mV
EE = 97.71% ± 0.89%
Enhanced solubility of
DAS (1200-fold) after
inclusion in NLCs;
Inhibition of the development of CNV and associated
corneal pathological
alterations in a mouse
model of CNV.
[198]
Monolaurin
Capryol-90
Cremophor RH40
Transcutol P
Glycerin
Sorafenib/
Corneal neovascularization
Microemulsion methodSize 111.87 ± 0.93 nm
PDI 0.15 ± 0.01
ζ potential−0.35 ± 0.08 mV
EE = 99.20 ± 0.86%
Excellent ocular tolerability (in vivo test on rabbits),
non-toxic in HCEC;
Approximately 6.7- and 1.3-fold higher drug concentrations in rabbit cornea and conjunctiva vs. free drug.
[199]
Compritol
888 ATO
Apifil (PEG-8 beeswax)
Miglyol 812N
Labrasol, Kolliphor
EL
Cremophor
RH60
Dexamethasone/
Ophthalmic inflammatory diseases, severe uveitis
Ultrasonication methodSize 92.18 ± 0.49 nm
PDI 0.12 ± 0.02
ζ potential −7.62 ± 0.26, EE = 88.31%
Good ocular tolerability;
Ability to penetrate across the cornea;
High concentration of NLCs in the stroma, according to porcine corneal penetration study.
[171]
Capmul MCM C10
Soya lecithin
Captex 200 P
Transcutol P
Polysorbate 80
Stearylamine
Triamcinolone acetonide/
Uveitis
Hot microemulsion methodSize 198.95 ± 12.82 nm
PDI 0.326 ± 0.04
ζ potential 35.8 ± 1.94 mV
EE = 88.14 ± 3.03 %
Sustained drug release
(84% within 24 h);
Ex vivo corneal
permeation of 51%;
Biocompatible and ocular tolerable formulation
(HET-CAM test).
[200]
Cholesterol
Stearic acid
Stearylamine
Oleic acid
Labrafil M 1944
Tween 80
Vancomycin
(VMC)/
Bacterial endophthalmitis
Cold homogenization techniqueSize 96.40 ± 0.71 nm
PDI 0.352 ± 0.011
ζ potential 29.7 ± 0.47 mV,
EE = 74.80 ± 4.30%
Improved transcorneal
penetration;
Biocompatible, non-irritant formulation (in vitro RBC hemolytic assay);
Enhanced (3-fold) intravitreal VMC concentration after topical application compared to drug solution.
[201]
Miglyol 812
Compritol 888 ATO
Lutrol F68
Palmitoylethanolamide
(PEA)/
Retinal diseases
(diabetic retinopathy, glaucoma)
High
shear homogenization
Size 208.6 ± 10.2 nm
PDI 0.18
ζ potential > 20 mV
Improved ocular
bioavailability: 40% and 100% higher PEA levels in vitreous body and retina compared to free drug.
[202]
Glyceryl monostearate
Labrafil M 2125 CS
Tween 80
Transcutol HP
Chitosan
5-Fluorouracil
(5-FU)/
Diabetic
retinopathy
Melt emulsification-ultrasonication methodSize 163.2 ± 2.3 nm
PDI 0.28 ± 1.52
ζ potential 21.4 ± 0.5 mV
EE = 85.0 ± 0.2 %
Higher and sustained
5-FU release vs. free drug;
Non-irritant formulations;
Antiangiogenic effect
confirmed by in vivo study in a diabetic
retinopathy rat model.
[181]
Capryol 90
Softisan 100
Tween 80
Diosmin/
Diabetic
retinopathy
Melt emulsification method and ultrasonicationSize 83.58 ± 0.77 nm
PDI 0.263 ± 0.067
ζ potential −18.5 ± 0.60 mV
EE = 99.53± 2.50
Very good physical stability of NLCs up to 60 days;
Cytocompatibility
assessed on ARPE-19 cells,
Cytoprotective effects.
[203]
Compritol 888 ATO
Miglyol 812
Lutrol F68
Mangiferin
(MNG)/
Oxidative stress related diseases, macular degeneration, diabetic retinopathy
High shear homogenization and ultrasoundSize 148.9 ± 0.1 nm
PDI 0.21 ± 0.02
ζ potential −23.5 ± 0.2 mV, EE ≈ 92%
Higher antioxidant activity of MNG NLCs vs.
free compound
according to ORAC assay;
Non-irritant formulations according to
HET−CAM Assay.
[204]
Glyceryl monostearate
Castor oil
Poloxamer 188
Brimonidine/
Glaucoma, ocular hypertension
High shear homogenizationSize 151.97 ±1.98 nm
PDI 0.230 ± 0.01
ζ potential −44.2 ± 7.81 mV
EE = 83.631 ± 0.495%
Improved permeability compared to analogous model SLNs;
Highest reduction in the IOP in rabbits (vs. SLNs and free drug).
[172]
Captex 200P (propylene glycol dicaprate)
Soya lecithin
Capmul®
MCM C10 (glyceryl monocaprate)
Tween 80
Transcutol P
Stearylamine
Captex 200P
Brinzolamide
(Brla)
Latanoprost
(Ltp)/
Glaucoma
Hot microemulsion methodSize165.28 ± 2.36 nm
PDI 0.31 ± 0.015
ζ potential 35.33 ± 0.37 mV
EE = 97.5 ± 2.16%
Adequate transcorneal
permeation (Brla and Ltp levels after 24 h were
≈82% and ≈84%, respectively);
Effective reduction
of IOP in rats’ eyes
with laser-induced
glaucoma.
[192]
Legend: ARPE—Human retinal pigment epithelial cell line, CNV—Corneal neovascularization, DED—Dry eye disease, HCEC—Human corneal epithelial cell lines, HET−CAM—Hen’s egg test on chorioallantoic membrane, IL-6—Interleukin-6, MMP—Matrix metalloproteinases, mPEG-2K-DSPE sodium salt—N-(Carbonyl-methoxypolyethylenglycol-2000)-1,2-distearoyl-sn-glycero-3-phosphoethanolamine sodium salt, ORAC—Oxygen radical absorbance capacity, TNF-α—Tumor necrosis factor α.
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

Gugleva, V.; Andonova, V. Recent Progress of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Ocular Drug Delivery Platforms. Pharmaceuticals 2023, 16, 474. https://doi.org/10.3390/ph16030474

AMA Style

Gugleva V, Andonova V. Recent Progress of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Ocular Drug Delivery Platforms. Pharmaceuticals. 2023; 16(3):474. https://doi.org/10.3390/ph16030474

Chicago/Turabian Style

Gugleva, Viliana, and Velichka Andonova. 2023. "Recent Progress of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Ocular Drug Delivery Platforms" Pharmaceuticals 16, no. 3: 474. https://doi.org/10.3390/ph16030474

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop