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

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.
Table 2. Recent progress of SLNs for ophthalmic application (5 years’ overview).
CompositionDrug/Disease Method 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]
PrecirolATO 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]
Compritol 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.

Download the full article as PDF here: Recent Progress of Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Ocular Drug Delivery Platforms

or read it here

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


Read more on Chitosan as a pharmaceutical excipient here:

Chitosan as a pharmaceutical excipient
Chitosan as a pharmaceutical excipient
You might also like