Nano-based DDS for Posterior Segment Diseases: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , ,

The complex anatomy of the eye presents a major challenge in the treatment of posterior segment eye diseases, hindering the effective delivery of medications. Conventional treatments, including topical eye drops and intravitreal injections, are limited by poor bioavailability and short residence time, necessitating frequent dosing to manage the disease. Intravitreal injections can also lead to serious ocular complications. Biodegradable nano-based drug delivery systems (DDSs) have emerged as a potential solution to these limitations, offering longer residence time in ocular tissues and better penetration through ocular barriers. Furthermore, the biodegradable polymers used to create these systems are nanosized, reducing the risk of toxicity and adverse reactions.

  • ocular surface disease
  • retinal disease
  • nanosystems for ocular drug delivery
  • nanocarriers
  • biodegradable polymers
  • ocular drug delivery system
  • hydrogels
  • ocular inserts
  • exosomes

1. Posterior Segment Diseases

For delivering treatments to the posterior segment of the eye, bioavailability becomes a challenge using a systemic or topical route. Most systemic medications, whether administered orally or intravenously, have difficulty crossing the blood–retinal barrier (BRB), requiring a high-dose administration that can result in systemic side effects. Topical eye drops also have limitations due to the multiple ocular barriers that impede the medication’s path from the ocular surface to the posterior segment, as discussed in a previous section of this article. Intravitreal injections are more invasive and can lead to potential sight-threatening complications such as endophthalmitis or retinal detachment. Additionally, they have a short retention time and require multiple visits to the ophthalmologist for administration in a sterile condition, which decreases patient compliance [1]. To ameliorate these issues, there has been extensive research done within nanomedicine to improve drug delivery to the posterior eye in a targeted, prolonged manner.

2. Retinitis Pigmentosa

Retinitis pigmentosa (RP) is a group of inherited disorders that affect the retina. It is caused by various genetic mutations, leading to the degeneration of the photoreceptor cells, primarily rods rather than cones, and subsequent progressive vision loss beginning with night blindness and peripheral vision loss. As RP is a genetic condition with over 3000 known mutations that target specific systems or proteins, which are affected by multiple mutations, it is an effective approach to maximizing the therapeutic effect in a large patient population. Successful nano-based DDSs of therapies, which have shown success in this front, thus hold great promise. Several studies have instead focused on targeting the posterior eye to prevent retinal degeneration, such as preventing photoreceptor death and promoting its survival. This is typically accomplished by administering neuroprotective agents to retinal cells. These agents have neurotrophic, anti-apoptotic, or antioxidant properties aimed at reducing retinal inflammation, decreasing oxidative stress, and promoting repair of damaged neurons and cells [2].
Arranz–Romera et al. used PLGA microspheres to co-deliver the growth-derived neurotrophic factor (GDNF) to promote neuronal survival, with tauroursodeoxycholic acid (TUDCA), a substance shown to have anti-apoptotic, antioxidant, anti-inflammatory, and cytoprotective attributes in retinal degeneration models [3] The biodegradable nature of this microsphere allowed for a sustained, erosion-driven controlled drug release in the target tissue at effective concentrations. Through the optimization of drug loading, they were able to improve TUDCA entrapment while reducing the initial burst effect of GDNF. They observed a sustained release for at least 91 days in vitro, an essential component for RP since it requires long-acting drug responses. One benefit of nano-based formulations is the possibility of small-scale modifications that have a significant impact on the final behaviour of the DDSs and drugs. In this study, the addition of vitamin E during microsphere formulations allowed for a greater stability of GDNF during the emulsion, translating to improved GDNF function and prolonged release in vitro. Furthermore, the use of the water-soluble ethanol (EtOH) as a co-solvent-affected DDS solidification and microsphere porosity and structure, contributing to improved encapsulation efficiency of both GDNF and TUDCA. The external morphology of microparticles, modified through the addition of EtOH and other substances, affects the release profile of their encapsulated proteins [4]. Finally, combination therapy holds its own benefits, and in this experiment, it was observed that the presence of amphiphilic TUDCA modulated the release of hydrophilic GDNF. Another substance found to attenuate retinal degeneration is ML240, which inhibits the valosin-containing protein (VCP), a potential therapeutic target for autosomal-dominant RP [5]. To improve solubility and thereby maximize ML240’s therapeutic potential, Sen et al. used methoxy-poly (ethylene glycol) (mPEG)-cholane and mPEG-cholesterol-based nanoparticles that self-assemble to encapsulate the drug and improve its retention time [6]. The formulations prolonged the drug release over 10 days, and neuroprotection, particularly photoreceptor protection, was observed for up to 21 days in retinal explants with decreased inflammatory microglial responses in an ex vivo rat model. It was also observed that the formulations were safe and well-tolerated in in vivo wild-type rat eyes. However, this may not translate to rats or humans with RP as there are secondary insults and biological changes that are not present in wild-type counterparts. The study nevertheless highlights the significant role of nano-based DDSs for making accessible therapeutic targets that have shown an initial promise but are limited by their delivery and behaviour in vivo without support. Furthermore, they observed small particle sizes of mPEG-loaded nanoparticles, ranging from 32 to 55 nm, which is optimal for corneal penetration, absorption, reduced eye irritation, and patient compliance as this requires smaller needles. However, as with the above study, in vivo work is required to concretely establish therapeutic success as many initially promising therapies fail to instigate the desired effect in the complicated in vivo system. It should also be noted that neither study directly assessed the biodegradability of its proposed DDS. While both PLGA and PEG are biocompatible and degradable, it is worth exploring the biodegradability, and subsequent long-term effects of the degraded components, for specific formulations. Prioritizing patient comfort, Platania et al. developed a novel topical formulation of myriocin-loaded nanostructured lipid carriers (Myr-NLCs) in the form of eye-drops [7]. They observed that this system considerably decreased retinal sphingolipid levels in rabbit eyes, showing potential in the treatment of RP by inhibiting ceramide synthesis. The researchers observed that the Myr-NLC formulation is well-tolerated after delivery and indicated effective levels of myriocin in the posterior eye. In previous work, myriocin has shown promise in lowering retinal ceremide levels in RP mouse models when loaded in solid lipid nanocarriers (SLNs) [8]. This current work went one step further to highlight the superiority of NLCs over SLNs, particularly for drug solubility and, thus, loading. SLNs face challenges with long-term storage as there is a high chance of drug expulsion that can be overcome with NLCs, allowing for possible large-scale production if clinical success is achieved [1]. However, it is currently unclear how well these NLCs translate to in vivo efficacy. In particular, myriocin has limited stability at temperatures above 0 °C and, despite the increased stability afforded by the NLC system, it is unclear how the drug will respond at physiological temperature.
Due to the limited number of studies, and the high heterogeneity in the type and formulation of the DDSs and the active substances assessed, it is currently unclear which nano-based DDSs are most effective for RP. The longest sustained release, with a reduced initial burst release, was observed with the use of PLGA microspheres. However, whether longer release time necessarily correlates to drug efficacy and disease treatment is unclear. Regardless, it can be concluded that DDSs, which successfully enhance residence time and the stability of potential therapeutic targets that have been previously limited in their use and modulate neuroprotective effects in the retina, are likely to show the most promise in clinical applications. Overall, all studies mentioned above conducted preliminary ex vivo, in vitro, and in vivo experiments. Therefore, there is a need for in-vivo models on bigger rodents with similar anatomy to the human eye to further elucidate the therapeutic efficacy in a way that can be clinically translatable for RP.

3. Age-Related Macular Degeneration and Choroidal Neovascularization

Age-related macular degeneration (AMD) is a prevalent eye disorder that affects individuals over the age of 50 and is a major contributor to vision loss and blindness among the elderly. The condition affects the macula and results in difficulties with tasks such as reading and facial recognition. AMD can be classified into two types: dry and wet. The dry form is the most common type and progresses gradually over time. The wet form, while less common, is more severe and results from the growth of abnormal blood vessels under the macula, which then leak fluid and blood, leading to a rapid decline in vision. There are various delivery targets for AMD, including reducing inflammation and drusen formations, improving RPE survival, inhibiting angiogenesis, as well as treating choroidal neovascularization (CNV) found in wet-AMD. In a clinical setting, the treatment for AMD depends on its severity and type. Dry AMD can be monitored and managed with nutritional supplements, while wet AMD typically requires regular intravitreal injections of anti-VEGF drugs [9][10].
Anti-VEGF therapy has been one of the most common therapies for treating wet-AMD and CNV, and nano-based DDS systems to improve its delivery will be extensively reviewed in the next sections. However, one-third of patients respond poorly to anti-VEGF based treatments, and there are potential vision-threatening complications such as endophthalmitis or retinal detachment. Intravitreal injections of anti-VEGF also heavily rely on a patient’s compliance [11][12]. Therefore, there is a need for optimizing therapies targeting the inflammation, degeneration, and development of the neovascularisation.
There have been efforts in creating biomimetic nano-based DDSs to improve targeted delivery to CNV lesion sites in the eyes of AMD patients. Zhang et al. used mesenchymal stem cells (MSCs) to carry PLGA nanoparticles loaded with HIF-1α siRNA. Inhibiting HIF-1α can reduce a variety of pro-angiogenic factors working upstream of VEGF [13].
Given that hypoxia plays a major role in the pathogenesis of CNV, the study was conducted within a hypoxic environment. MSCs were able to target CNV lesion sites in this environment with the biodegradable nanoparticles improving the drug-carrying capacity and sustained release. Drug delivery through stem cell loading reached clinical trials in several cases, including apoptotic-inducing factors and oncolytic viruses, holding promise for MSC-guided delivery in retinal disorders [14]. Here, a compounded benefit is observed in which combination therapy overcomes the individual barriers to each component. siRNA alone is prone to RNAse enzymatic degradation, but encapsulation in PLGA NPs has proven protective for siRNA. Likewise, MSCs alone have poor drug carrier capacity due to poor drug loading, which can be ameliorated with the engineering of MSCs with NPs, enhancing drug loading and therapeutic efficacy. The PLGA NPs-loaded HIF siRNA effectively decreases expression of HIF-1α for 7 days in retinal pigment epithelial (RPE) cells. However, no significant difference was observed in the proliferation, apoptosis, or migration of RPEs when compared to control groups, suggesting that more work is needed to characterize how well MSC-guided delivery translates to physiological and functional improvement. Overall, this formulation requires further optimization and safety testing on animals to ensure a therapeutic benefit for AMD and CNV.
To treat CNV intravenously, Xia et al. provide another biomimetic DDS using macrophages to disguise PLGA nanoparticles loaded with rapamycin [9]. Rapamycin is an mTOR inhibitor that is known to suppress inflammation, enhance the dysregulated autophagy observed in AMD, and act upstream to VEGF-mediated inhibition of angiogenesis. Although a promising therapeutic drug for AMD, rapamycin’s low water solubility and poor accumulation at lesion sites have historically limited its use. Using the knowledge that macrophages are generally recruited to areas of RPE atrophy and CNV lesions, Xia et al. applied this to deliver PLGA-rapamycin nanoparticles intravenously in a laser-induced CNV mouse model. PLGA, as a hydrophobic drug carrier, opens the door to several potential drugs with limited water solubility despite an initial promise. The drug successfully traversed the impaired BRB, improved the bioavailability of rapamycin, and, along with anti-angiogenic effects, contributed to suppressed neovascularization. Rapamycin delivery also suppressed inflammation and enhanced autophagy both in vitro and in vivo in a CNV mouse model. Xia et al. carefully parsed out the mechanisms of action of macrophage-guided drug delivery and subsequent impact on the retinal microenvironment successfully, and characterized both in vitro and in vivo behaviour, paving the way for future clinical work to characterize the use of this formulation more effectively in humans. Using biomimetic carriers could, therefore, provide an alternative way to improve posterior ocular delivery. Rapamycin was also delivered intravitreally using synthetic high-density lipoprotein (sHDL) nanoparticles in a study by Mei et al. [10]. They particularly focused on a treatment for dry AMD, using rapamycin to suppress inflammation through the inhibition of NF-κB, as well as enhance autophagy, and using sHDL to also reduce lipid deposition, contributing to drusen formation. This DDS altogether provided a non-toxic, synergistic, anti-inflammatory effect and improved the bioavailability and distribution of rapamycin to the RPE layer following intravitreal administration in rats, with as much as a 125-fold increase in drug aqueous concentration. Combined with the observed benefits of macrophage-guided rapamycin delivery, it can be said that rapamycin is a promising drug for AMD and CNV, both because of its influence on VEGF production as well as the general effects on apoptosis, autophagy, and inflammation. This study also highlights the benefits of combined therapy, as sHDL itself had protective effects through the removal of excess cholesterol alongside its role as the nanocarrier. It also circles back to the influence of nanocarriers in effectively delivering hydrophobic drugs in largely hydrophilic environments, such as the ocular environment. However, it should be noted that neither study exploring rapamycin efficacy has explored the longevity of their formulations and the effects of long-term delivery of rapamycin in the posterior eye segment. Further studies using disease animal models are also needed to validate therapeutic efficacy and modify these therapies for clinical translation. Moreover, there are adverse side effects associated with frequent intravitreal injections.
Oxidative stress and the production of reactive oxidative species (ROS) have also been implicated in the pathophysiology and progression of AMD, thus targeting ROS production to initiate antioxidative effects. To explore this, Nguyen et al. intravitreally co-delivered resveratrol and metformin using poly(ε-caprolactone) (PCL) nanoparticles as a potential therapy for wet AMD [15]. Combined with metformin’s anti-angiogenic effects, resveratrol has been noted to provide antioxidant and anti-inflammatory effects. Due to the multifaceted effects of ROS-initiated RPE damage, therapies that can simultaneously target several components at once are highly desirable. The advantages of PCL, including its biodegradability, are mentioned, where PCL is not only considered more biocompatible in the RPE regions, but its degraded by-products are less acidic when compared to PLGA and PLA, which result in the build-up of lactic acid, avoiding unnecessary associated inflammation. It is also FDA-approved, thus easing progression in clinical trials. The polymer was further modified with cell-penetrating peptides (CPPs) to significantly improve retinal permeability. A sustained release for up to 56 days, as well as therapeutic effects, were observed in a rat model of AMD. This study provides a foundation for future long-term efficacy and safety studies. Another co-delivery system was suggested by Lai et al. for berberine hydrochloride and chrysophanol, which possesses potent antioxidant, anti-angiogenic, and anti-inflammatory properties [16]. These drugs have demonstrated potential in the treatment of AMD in animal studies. Previously limited in their application due to poor stability and bioavailability, Lai et al. proposed using polyamidoamine dendrimers (PAMAM) and liposomes to effectively deliver berberine hydrochloride and chrysophanol to the retina. PAMAM acts as an external coating for the compound-loaded liposomes due to its high water-binding ability and low toxicity. In comparison to uncoated compound liposomes, this coated DDS revealed a negative zeta potential, which is preferred for drug delivery to the retina, and significantly improved encapsulation efficiency, demonstrating that PAMAM coating enhanced drug loading. Results show considerable cellular permeability and increased bio-adhesion on corneal epithelial cells. PAMAM-liposome systems (P-CBLs) also substantially improved berberine hydrochloride bioavailability. Further, no side effects were observed on rabbit ocular surface structure after the administration of P-CBLs. While the drugs exhibited stability for 7 h in vivo, the study did not assess the release profiles of the drugs in the posterior segment of the eye, leaving questions regarding the functionality of this DDS in AMD. Regardless, the P-CBL system displays a potential use for treating AMD and, potentially, other ocular diseases.
Oxidative stress and the production of reactive oxidative species (ROS) have also been implicated in the pathophysiology and progression of AMD. Thus, targeting ROS production to initiate antioxidative effects. To explore this, Nguyen et al. intravitreally co-delivered resveratrol and metformin using poly(ε-caprolactone) (PCL) nanoparticles as a potential therapy for wet AMD [15]. Combined with metformin’s anti-angiogenic effects, resveratrol has been noted to provide antioxidant and anti-inflammatory effects. Due to the multifaceted effects of ROS-initiated RPE damage, therapies that can simultaneously target several components at once are highly desirable. The advantages of PCL, including its biodegradability, are mentioned, where PCL is not only considered more biocompatible in the RPE regions, but its degraded by-products are less acidic when compared to PLGA and PLA, which result in build-up of lactic acid, avoiding unnecessary associated inflammation. It is also FDA-approved, thus easing progression in clinical trials. The polymer was further modified with cell-penetrating peptides (CPPs) to significantly improve retinal permeability. A sustained release for up to 56 days, as well as therapeutic effects, were observed in a rat model of AMD. This study provides a foundation for future long-term efficacy and safety studies. Another co-delivery system was suggested by Lai et al. for berberine hydrochloride and chrysophanol, which possesses potent antioxidant, anti-angiogenic, and anti-inflammatory properties [16]. These drugs have demonstrated potential in the treatment of AMD in animal studies. Previously limited in their application due to poor stability and bioavailability, Lai et al. proposed using polyamidoamine dendrimers (PAMAM) and liposomes to effectively deliver berberine hydrochloride and chrysophanol to the retina. PAMAM acts as an external coating for the compound-loaded liposomes due to its high water-binding ability and low toxicity. In comparison to uncoated compound liposomes, this coated DDS revealed a negative zeta potential, which is preferred for drug delivery to the retina and significantly improved encapsulation efficiency, demonstrating that PAMAM coating enhanced drug loading. Results show considerable cellular permeability and increased bio-adhesion on corneal epithelial cells. PAMAM-liposome systems (P-CBLs) also substantially improved berberine hydrochloride bioavailability. Further, no side effects were observed on the rabbit ocular surface structure after the administration of P-CBLs. While the drugs exhibited stability for 7 h in vivo, the study did not assess the release profiles of the drugs in the posterior segment of the eye, leaving questions regarding the functionality of this DDS in AMD. Regardless, the P-CBL system displays a potential use for treating AMD and, potentially, other ocular diseases.

4. Diabetic Retinopathy

Diabetic retinopathy is a chronic ocular condition affecting diabetic patients. The condition results from damage to the blood vessels in the retina and can progress over time. There are two main stages of diabetic retinopathy: non-proliferative diabetic retinopathy (NPDR) and proliferative diabetic retinopathy (PDR). NPDR is characterized by increased vascular permeability and capillary occlusion, and can lead to the formation of microaneurysms, dot and blot hemorrhages, cotton wool spots, and hard exudates. PDR occurs in advanced stages of diabetic retinopathy due to continued damage to the retinal blood vessels, leading to significant retinal ischemia. The ischemic retinal tissue releases pro-angiogenic factors, including the vascular endothelial growth factor (VEGF), which stimulates the production of new and abnormal blood vessels. These neo-vessels can lead to various vision-threatening complications, such as a neovascularization of the disc and retina causing vitreous hemorrhage and tractional retinal detachment, and neovascularization of the iris and angle resulting in glaucoma. The management of Proliferative Diabetic Retinopathy primarily focuses on reducing the production of VEGF by ischemic tissue through laser photocoagulation or intravitreal anti-VEGF injections.
Antioxidants, anti-inflammatory agents, and neurotrophic factors are considered promising options to treat the neuronal and vascular abnormalities that progress with diabetic retinopathy (DR) [17]. Nano-carriers have been proposed to improve the targeting of the diabetic retina. Due to their high biocompatibility, PLGA-based nanoparticles have been used to improve the therapeutic efficacy of drugs that are currently limited due to inefficient delivery routes. For example, Zeng et al. used PLGA nanoparticles to deliver Interleukin-12 (IL-12), a cytokine that can diminish the levels of matrix metalloproteinase-9 (MMP-9) and VEGF-A, both of which are known to affect the severity of diabetic retinopathy [18]. Previously limited due to it being prone to rapid degradation, when IL-12 was carried by PLGA nanoparticles (IL-12-PNP), it had an appreciable drug encapsulation efficiency (~34.7%) and prolonged drug release. IL-12-PNP exhibited better inhibition against VEGF-A and MMP-9 expression in diabetic retinopathic mouse retina and rat endothelial cells. Moreover, this treatment resulted in significantly decreased retinal damage in a DR mouse model with increased thickness and reduced neovascularization. Similarly, Romeo et al. proposed to deliver melatonin with PLGA-PEG Lipid-polymer hybrid nanoparticles (LPHN) [19]. Melatonin offers various neuroprotective strategies suitable for treating this DR. However, at high doses, it may compromise retina morphology and functioning. The DDS developed in this study targeted the retina without unnecessary high dosages to deliver melatonin. Using a biodegradable polymer, they found no signs of cytotoxicity or ocular irritation in vivo and confirmed neuroprotective and antioxidant effects on a model of glucose-induced diabetic retinopathy on Human Retinal Endothelial Cells (HREC). They also observed high encapsulation efficacy (79.8%) using this hybrid model, suggesting its superiority to a PLGA only nanoparticle. In previous work, the neuroprotective effects of melatonin have been observed only after prolonged exposure of greater than 72 h, necessitating a stable, sustained release DDS for its ocular delivery. Romeo et al. successfully observed a prolonged and sustained release for up to 8 days compared to a rapid burst release of free melatonin.
Another example of a lipid-modified nanoparticle system is a study by Zingale et al., where they used nanostructured lipid carriers (NLCs) to deliver diosmin, a flavonoid known for its anti-inflammatory, cytoprotective, and antioxidant effects, especially in high glucose environments [20]. They were able to achieve a high encapsulation efficiency, and the DDS was found safe and well-tolerated in vitro. However, a common issue observed with using lipid-based nanocarriers is the need to use surfactants for their preparation that may cause irritation and a sensitizing action [21]. Further studies are being conducted to confirm the clinically relevant anti-inflammatory effects of diosmin NLCs. As mentioned above, NLCs have the advantage of minimal toxicity as it can be manufactured without the requirement of toxic organic solvents [22]. They can also be stored stably for long periods, as Zingale et al. observed stability under different storage conditions for up to 60 days. NLCs further possess the versatility of being applied as topical eye drops as demonstrated here and also by Platania et al., which greatly increases patient compliance [6]. What’s currently unclear and garners further exploration is the release profiles of drug-loaded NLCs, to better assess how often administration is required.
Other types of biodegradable nanoparticles have also been assessed for optimizing treatments for diabetic retinopathy. Radwan et al. investigated an alternative non-invasive delivery with an anti-VEGF factor, apatinib, encapsulated into bovine serum albumin nanoparticles, which are coated with hyaluronic acid [23]. With a relatively high entrapment efficiency (~69%), these apatinib-loaded nanoparticles (Apa-HA-BSA-NPs) illustrated a sustained biphasic release rate with an initial burst, appreciable mucoadhesion, and no cytotoxicity were detected on rabbit corneal epithelial cells. This 2021 study indicated improved retinal thickness and lessen retinal microstructural and ultrastructural changes in Apa–HA–BSA–NP-treated eyes. Moreover, the authors observed better retinal accumulation through this topical treatment while avoiding ocular complications resulting from frequent intravitreal injections. As aforementioned in the AMD section, using PCL as a biodegradable polymer for nanoparticle systems has many advantages [15]. For diabetic retinopathy, Mahaling et al. developed nanoparticles with a hydrophobic polycaprolactone (PCL) core and a hydrophilic Pluronic® F68 shell, containing triamcinolone acetonide [24]. TA has demonstrated efficacy in both NPDR and PDR, attributed to its anti-inflammatory, anti-angiogenic, and neuroprotective properties. Likewise, NPs containing PCL and PF68 have previously demonstrated strong bioavailability in retina during topical administration [25]. In a DR rat model, a topical administration of these nanoparticles resulted in significant structural improvements, particularly retinal thickness and vascular health, as well as functional improvements. The authors found diminished retinal inflammation, decreased glial cell hyperplasia, and reduced microvascular complications. These findings demonstrate the potential of a triamcinolone acetonide-loaded nanoparticle delivery system in the treatment of diabetic retinopathy. Topical administration has observed significant success in DR animal models, opening the door to non-invasive, patient self-administered delivery routes. This overcomes several challenges of intravitreal administration, including intraocular bleeding, increased intraocular pressure, endophthalmitis, and discomfort.

5. Diabetic Macular Edema (DME)

Diabetic macular edema (DME) is a common complication in diabetic retinopathy where fluid accumulates in the macula causing rapidly progressive decrease in visual acuity. It occurs due to increased permeability and inflammation in the retinal vessels [17].
Other than intravitreal anti-VEGF injection and topical NSAIDs, intravitreal triamcinolone acetonide (TA) can sometimes be used to reduce associated inflammation with DME. However, intravitreal triamcinolone is associated with excessively high rates of complications, such as IOP elevation and cataract formation. Navarro–Partida et al. provided a topical route for delivering TA by loading it on liposomes [26]. This was a feasibility study, where they first found TA-loaded liposomes to be safe and tolerable in healthy patients through a Phase 1 clinical assay. They further presented a sustained therapeutic effect of reduced central fovea thickness (CFT) in DME patients through an open-label, non-randomized study. Further studies are required to confirm the long-term safety and therapeutic efficacy, such as ensuring TA at high concentrations does not adversely affect intraocular morphology and function [27]. To improve the biodegradability and mucoadhesion of liposomes, Khalil et al. used chitosan-coated liposomes to deliver TA to the posterior segment [28]. This enhanced bioavailability and prolonged the release of TA in their in vivo models. Although their efficiency of drug release was done on a CNV rat model, the authors recommend this DDS for any posterior segment disease, particularly highlighting DME, proliferative diabetic retinopathy, and CNV related to AMD. Further in vivo studies are required to validate the therapeutic efficacy of this DDS, ensuring its clinical significance. Initial clinical success with TA topical administration in lipid-based nanomaterial has further supported both the superiority of lipid-based DDSs and topical administration in ocular drug delivery. Khalil et al. further demonstrate the flexibility afforded by nano-based DDS, as base constructs, such as liposomes, can be modified to improve retention time, permeability, encapsulation efficiency, and personalize treatment to the drug, disease, and area of interest.

This entry is adapted from the peer-reviewed paper 10.3390/pharmaceutics15041094

References

  1. 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.
  2. Cuenca, N.; Fernández-Sánchez, L.; Campello, L.; Maneu, V.; De la Villa, P.; Lax, P.; Pinilla, I. Cellular Responses Following Retinal Injuries and Therapeutic Approaches for Neurodegenerative Diseases. Prog. Retin. Eye Res. 2014, 43, 17–75.
  3. Arranz-Romera, A.; Esteban-Pérez, S.; Molina-Martínez, I.T.; Bravo-Osuna, I.; Herrero-Vanrell, R. Co-Delivery of Glial Cell-Derived Neurotrophic Factor (GDNF) and Tauroursodeoxycholic Acid (TUDCA) from PLGA Microspheres: Potential Combination Therapy for Retinal Diseases. Drug Deliv. Transl. Res. 2021, 11, 566–580.
  4. Martín-Sabroso, C.; Fraguas-Sánchez, A.I.; Aparicio-Blanco, J.; Cano-Abad, M.F.; Torres-Suárez, A.I. Critical Attributes of Formulation and of Elaboration Process of PLGA-Protein Microparticles. Int. J. Pharm. 2015, 480, 27–36.
  5. Sen, M.; Bassetto, M.; Poulhes, F.; Zelphati, O.; Ueffing, M.; Arango-Gonzalez, B. Efficient Ocular Delivery of VCP SiRNA via Reverse Magnetofection in RHO P23H Rodent Retina Explants. Pharmaceutics 2021, 13, 225.
  6. Sen, M.; Al-Amin, M.; Kicková, E.; Sadeghi, A.; Puranen, J.; Urtti, A.; Caliceti, P.; Salmaso, S.; Arango-Gonzalez, B.; Ueffing, M. Retinal Neuroprotection by Controlled Release of a VCP Inhibitor from Self-Assembled Nanoparticles. J. Control. Release 2021, 339, 307–320.
  7. Platania, C.B.M.; Dei Cas, M.; Cianciolo, S.; Fidilio, A.; Lazzara, F.; Paroni, R.; Pignatello, R.; Strettoi, E.; Ghidoni, R.; Drago, F.; et al. Novel Ophthalmic Formulation of Myriocin: Implications in Retinitis Pigmentosa. Drug Deliv. 2019, 26, 237–243.
  8. Strettoi, E.; Gargini, C.; Novelli, E.; Sala, G.; Piano, I.; Gasco, P.; Ghidoni, R. Inhibition of Ceramide Biosynthesis Preserves Photoreceptor Structure and Function in a Mouse Model of Retinitis Pigmentosa. Proc. Natl. Acad. Sci. USA 2010, 107, 18706–18711.
  9. Xia, W.; Li, C.; Chen, Q.; Huang, J.; Zhao, Z.; Liu, P.; Xu, K.; Li, L.; Hu, F.; Zhang, S.; et al. Intravenous Route to Choroidal Neovascularization by Macrophage-Disguised Nanocarriers for MTOR Modulation. Acta Pharm. Sin. B 2022, 12, 2506–2521.
  10. Mei, L.; Yu, M.; Liu, Y.; Weh, E.; Pawar, M.; Li, L.; Besirli, C.G.; Schwendeman, A.A. Synthetic High-Density Lipoprotein Nanoparticles Delivering Rapamycin for the Treatment of Age-Related Macular Degeneration. Nanomedicine 2022, 44, 102571.
  11. Martin, D.F.; Maguire, M.G.; Fine, S.L.; Ying, G.; Jaffe, G.J.; Grunwald, J.E.; Toth, C.; Redford, M.; Ferris, F.L. Ranibizumab and Bevacizumab for Treatment of Neovascular Age-Related Macular Degeneration: Two-Year Results. Ophthalmology 2020, 127, S135–S145.
  12. Feng, L.; Ju, M.; Lee, K.Y.V.; Mackey, A.; Evangelista, M.; Iwata, D.; Adamson, P.; Lashkari, K.; Foxton, R.; Shima, D.; et al. A Proinflammatory Function of Toll-Like Receptor 2 in the Retinal Pigment Epithelium as a Novel Target for Reducing Choroidal Neovascularization in Age-Related Macular Degeneration. Am. J. Pathol. 2017, 187, 2208–2221.
  13. Zhang, L.; Yan, J.J.; Wang, H.Y.; Li, M.Q.; Wang, X.X.; Fan, L.; Wang, Y.S. A Trojan Horse Biomimetic Delivery System Using Mesenchymal Stem Cells for HIF-1α SiRNA-Loaded Nanoparticles on Retinal Pigment Epithelial Cells under Hypoxia Environment. Int. J. Ophthalmol. 2022, 15, 1743–1751.
  14. Kimbrel, E.A.; Lanza, R. Next-Generation Stem Cells—Ushering in a New Era of Cell-Based Therapies. Nat. Rev. Drug Discov. 2020, 19, 463–479.
  15. Nguyen, D.D.; Luo, L.J.; Yang, C.J.; Lai, J.Y. Highly Retina-Permeating and Long-Acting Resveratrol/Metformin Nanotherapeutics for Enhanced Treatment of Macular Degeneration. ACS Nano 2023, 17, 168–183.
  16. Lai, S.; Wei, Y.; Wu, Q.; Zhou, K.; Liu, T.; Zhang, Y.; Jiang, N.; Xiao, W.; Chen, J.; Liu, Q.; et al. Liposomes for Effective Drug Delivery to the Ocular Posterior Chamber. J. Nanobiotechnol. 2019, 17, 64.
  17. Oshitari, T. Neurovascular Impairment and Therapeutic Strategies in Diabetic Retinopathy. Int. J. Environ. Res. Public Health 2021, 19, 439.
  18. Zeng, L.; Ma, W.; Shi, L.; Chen, X.; Wu, R.; Zhang, Y.; Chen, H.; Chen, H. Poly(Lactic-Co-Glycolic Acid) Nanoparticle-Mediated Interleukin-12 Delivery for the Treatment of Diabetic Retinopathy. Int. J. Nanomed. 2019, 14, 6357–6369.
  19. Romeo, A.; Bonaccorso, A.; Carbone, C.; Lupo, G.; Daniela Anfuso, C.; Giurdanella, G.; Caggia, C.; Randazzo, C.; Russo, N.; Luca Romano, G.; et al. Melatonin Loaded Hybrid Nanomedicine: DoE Approach, Optimization and in Vitro Study on Diabetic Retinopathy Model. Int. J. Pharm. 2022, 627, 122195.
  20. 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.
  21. 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.
  22. Müller, R.H.; Shegokar, R.; Keck, C.M. 20 Years of Lipid Nanoparticles (SLN and NLC): Present State of Development and Industrial Applications. Curr. Drug Discov. Technol. 2011, 8, 207–227.
  23. Radwan, S.E.S.; El-Kamel, A.; Zaki, E.I.; Burgalassi, S.; Zucchetti, E.; El-Moslemany, R.M. Hyaluronic-Coated Albumin Nanoparticles for the Non-Invasive Delivery of Apatinib in Diabetic Retinopathy. Int. J. Nanomed. 2021, 16, 4481–4494.
  24. Mahaling, B.; Srinivasarao, D.A.; Raghu, G.; Kasam, R.K.; Bhanuprakash Reddy, G.; Katti, D.S. A Non-Invasive Nanoparticle Mediated Delivery of Triamcinolone Acetonide Ameliorates Diabetic Retinopathy in Rats. Nanoscale 2018, 10, 16485–16498.
  25. Tang, B.C.; Dawson, M.; Lai, S.K.; Wang, Y.Y.; Suk, J.S.; Yang, M.; Zeitlin, P.; Boyle, M.P.; Fu, J.; Hanes, J. Biodegradable Polymer Nanoparticles That Rapidly Penetrate the Human Mucus Barrier. Proc. Natl. Acad. Sci. USA 2009, 106, 19268–19273.
  26. Navarro-Partida, J.; Altamirano-Vallejo, J.C.; la Rosa, A.G.D.; Armendariz-Borunda, J.; Castro-Castaneda, C.R.; Santos, A. Safety and Tolerability of Topical Ophthalmic Triamcinolone Acetonide-Loaded Liposomes Formulation and Evaluation of Its Biologic Activity in Patients with Diabetic Macular Edema. Pharmaceutics 2021, 13, 322.
  27. Chan, C.K.M.; Fan, D.S.P.; Chan, W.M.; Lai, W.W.; Lee, V.Y.W.; Lam, D.S.C. Ocular-Hypertensive Response and Corneal Endothelial Changes after Intravitreal Triamcinolone Injections in Chinese Subjects: A 6-Month Follow-up Study. Eye 2005, 19, 625–630.
  28. Khalil, M.; Hashmi, U.; Riaz, R.; Rukh Abbas, S. Chitosan Coated Liposomes (CCL) Containing Triamcinolone Acetonide for Sustained Delivery: A Potential Topical Treatment for Posterior Segment Diseases. Int. J. Biol. Macromol. 2020, 143, 483–491.
More
This entry is offline, you can click here to edit this entry!
Video Production Service