Nano-based DDS for Posterior Segment Diseases: Comparison
Please note this is a comparison between Version 4 by Rita Xu and Version 3 by Catherine Yang.

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.

This review provides a comprehensive overview of the latest advances in biodegradable nano-based DDSs for treating posterior segment diseases. By examining current therapeutic challenges and exploring various types of biodegradable nanocarriers, we aim to highlight the potential of these systems to enhance treatment outcomes. Our review includes pre-clinical and clinical studies published between 2017 and 2022, demonstrating the rapid evolution of nano-based DDSs. As biodegradable materials continue to advance, and our understanding of ocular pharmacology improves, nano-based DDSs hold great promise for overcoming obstacles encountered in clinical practice. 

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

The delivery of treatments to the posterior segment of the eye presents a challenge in achieving adequate bioavailability through systemic or topical routes. Systemic medications, whether administered orally or intravenously, struggle to cross the blood–retinal barrier (BRB), necessitating high-dose administration that can result in systemic side effects. Topical eye drops have limitations due to the multiple ocular barriers impeding the medication's path from the ocular surface to the posterior segment, as previously discussed. Intravitreal injections are invasive and carry potential sight-threatening complications such as endophthalmitis or retinal detachment. They require a sterile administration and need to be injected frequently during multiple ophthalmologist visits that contributes to decreasing patient compliance. Nanomedicine has been extensively researched to overcome these issues and improve drug delivery to the posterior eye in a targeted and prolonged manner, as summarized in Table 1.

 

2. Retinitis Pigmentosa

Retinitis Pigmentosa

Retinitis pigmentosa (RP) is a genetic disorder that causes progressive vision loss, starting with night blindness and peripheral vision loss due to the degeneration of photoreceptor cells, primarily rods. With over 3000 known mutations targeting specific systems or proteins, effective therapies for a large patient population are needed. Successful nano-based DDSs hold great promise for targeted and maximal therapeutic effect. In this regard, several studies have focused on administering neuroprotective agents to prevent retinal degeneration and promote the survival of retinal cells by reducing inflammation, decreasing oxidative stress, and promoting repair of damaged cells [70][1].

Arranz-Romera et al. used PLGA microspheres to co-deliver GDNF and TUDCA for promoting neuronal survival in retinal degeneration models [71][2]. These biodegradable microspheres provided a sustained, erosion-driven controlled release of effective drug concentrations for at least 91 days in vitro. Optimization of drug loading and the addition of vitamin E during microsphere formulations and water-soluble ethanol as a co-solvent significantly improved encapsulation efficiency of GDNF and TUDCA [72][3]. Furthermore, the external morphology of the microspheres was found to affect the release profile of encapsulated proteins. Sen et al. developed self-assembling nanoparticles based on mPEG-cholane and mPEG-cholesterol to improve the solubility and therapeutic potential of ML240, an inhibitor of valosin-containing protein (VCP) [74][4]. In retinal explants, the nanoparticles showed photoreceptor protection for up to 21 days in an ex vivo rat model, with in vivo studies demonstrating their safety and tolerability in wild-type rat eyes. However, it remains to be seen whether these results are translatable to rats or humans with RP. Platania et al. developed a topical formulation of myriocin-loaded nanostructured lipid carriers (Myr-NLCs) in the form of eye-drops [75][5]. The Myr-NLC formulation was well-tolerated and decreased retinal sphingolipid levels in rabbit eyes, showing potential for treating RP by inhibiting ceramide synthesis. Nonetheless, the performance of these NLCs in vivo is uncertain, especially given that myriocin has limited stability at physiological temperature. While the studies highlight the significant role of nano-based drug delivery systems in making therapeutic targets accessible, the biodegradability of specific formulations and their effects on the body must be explored further.

Currently, it is unclear which nano-based DDSs are most effective for RP due to the limited number of studies and the high heterogeneity in the type and formulation of the DDSs and active substances assessed. However, PLGA microspheres have shown the longest sustained release with a reduced initial burst release. Nonetheless, it remains unclear whether longer release time necessarily correlates with drug efficacy and disease treatment. Successful DDSs enhance drug residency time, improve stability of potential therapeutic targets, and modulate neuroprotective effects in the retina, leading to more promising clinical applications. While preliminary ex vivo, in vitro, and in vivo experiments have been conducted, there is a need for larger in vivo models, such as rodents with similar eye anatomy to humans, to further investigate therapeutic efficacy for RP that can be translated to clinical settings.

 

3. Age-Related Macular Degeneration and Choroidal Neovascularization

Age-Related Macular Degeneration and Choroidal Neovascularization

Age-related macular degeneration (AMD) is a common eye disorder that affects people over the age of 50 and is a significant contributor to vision loss in the elderly. There are two types of AMD: dry and wet. The dry form progresses gradually over time, while the wet form is less common but more severe and results from the growth of abnormal blood vessels under the macula. These vessels leak fluid and blood, leading to a rapid decline in vision. AMD can be targeted in various ways, such as reducing inflammation and drusen formation, improving RPE survival, inhibiting angiogenesis, and treating choroidal neovascularization (CNV) found in wet-AMD. Management of AMD depends on its severity and type. Nutritional supplements can be used to manage dry AMD, while regular intravitreal injections of anti-VEGF drugs are typically required for wet AMD [77, 78][6][7].

In the treatment of wet AMD and CNV, anti-VEGF therapy has been commonly used, and nano-based DDS systems have been developed to enhance its delivery [CITE]. However, a significant proportion of patients exhibit a poor response to anti-VEGF treatments, and the therapy may lead to severe complications such as endophthalmitis and retinal detachment. Additionally, the effectiveness of anti-VEGF treatment is dependent on patient compliance [CITE][8][9]. To address these limitations, it is important to optimize therapies targeting inflammation, degeneration, and the development of neovascularization.

Efforts have been made to create biomimetic nano-based DDSs for targeted delivery to CNV lesion sites in the eyes of AMD patients. For instance, Zhang et al. utilized mesenchymal stem cells (MSCs) to carry PLGA nanoparticles containing HIF-1α siRNA to reduce a variety of pro-angiogenic factors upstream of VEGF [81][10].

To improve targeted delivery to CNV lesion sites, Zhang et al. used mesenchymal stem cells (MSCs) to carry PLGA nanoparticles loaded with HIF-1α siRNA in a hypoxic environment. The biodegradable nanoparticles improved drug-carrying capacity and sustained release, making MSC-guided delivery promising for retinal disorders. Combining siRNA with PLGA NPs proved effective in decreasing expression of HIF-1α for 7 days in RPE cells. However, more work is needed to characterize the physiological and functional improvement, and the formulation requires further optimization and safety testing on animals to ensure therapeutic benefit for AMD and CNV. Nonetheless, this restudyearch demonstrates the compounded benefit of combination therapy, which overcomes individual barriers to each component, such as the protective effect of PLGA NPs on siRNA prone to enzymatic degradation and the enhanced drug loading and efficacy of MSCs engineered with NPs.

In the treatment of CNV, biomimetic DDSs have shown promise in improving targeted delivery to the lesion sites in the eyes of AMD patients. Xia et al. developed a DDS using macrophages to carry PLGA nanoparticles loaded with rapamycin intravenously. Rapamycin is known to suppress inflammation and enhance dysregulated autophagy observed in AMD, while acting upstream to VEGF-mediated inhibition of angiogenesis. Despite its promise, rapamycin's low water solubility and poor accumulation at lesion sites have historically limited its use. Xia et al. used the knowledge that macrophages are recruited to areas of RPE atrophy and CNV lesions to successfully deliver PLGA-rapamycin nanoparticles intravenously in a laser-induced CNV mouse model, improving bioavailability, suppressing neovascularization, inflammation and enhancing autophagy both in vitro and in vivo in a CNV mouse model. Similarly, Mei et al. used synthetic high-density lipoprotein (sHDL) nanoparticles to deliver rapamycin intravitreally, providing a non-toxic, synergistic, anti-inflammatory effect, reducing lipid deposition, and improving bioavailability and distribution of rapamycin to the RPE layer. Combined with the observed benefits of macrophage-guided rapamycin delivery, rapamycin shows promise for treating AMD and CNV. The use of nanocarriers is critical in delivering hydrophobic drugs in largely hydrophilic environments such as the ocular environment. However, neither study 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 needed to validate therapeutic efficacy and modify these therapies for clinical translation. Moreover, adverse side effects associated with frequent intravitreal injections should also be noted.

Oxidative stress and reactive oxidative species (ROS) have been linked to AMD, making ROS production an attractive target for antioxidant therapies. Nguyen et al. developed a potential therapy for wet AMD by co-delivering resveratrol and metformin using poly(ε-caprolactone) (PCL) nanoparticles [83][11]. Resveratrol provides antioxidant and anti-inflammatory effects, while metformin has anti-angiogenic properties. As ROS cause multifaceted RPE damage, targeting several components at once is desirable. PCL, a biodegradable polymer, is more biocompatible in the RPE regions than PLA and PLGA, and its degraded by-products are less acidic, avoiding inflammation. Cell-penetrating peptides (CPPs) improved retinal permeability, and sustained release for up to 56 days, and therapeutic effects were observed in a rat model of AMD. Lai et al. suggested a co-delivery system for berberine hydrochloride and chrysophanol, with potent antioxidant, anti-angiogenic, and anti-inflammatory properties [54][12]. These drugs have potential in the treatment of AMD. However, their poor stability and bioavailability were addressed using polyamidoamine dendrimers (PAMAM) and liposomes to effectively deliver the drugs to the retina. PAMAM-coated DDS revealed a negative zeta potential, preferred for drug delivery to the retina, and enhanced drug loading. PAMAM-liposome systems (P-CBLs) improved berberine hydrochloride bioavailability, with no side effects observed on the ocular surface structure of rabbits. Although the study did not evaluate the release profiles of the drugs in the posterior segment of the eye, the P-CBL system shows potential for treating AMD and other ocular diseases.

Oxidative stress and reactive oxidative species (ROS) are involved in the pathophysiology and progression of age-related macular degeneration (AMD). Targeting ROS production to initiate antioxidative effects is a potential therapeutic strategy for AMD. Nguyen et al. investigated the intravitreal co-delivery of resveratrol and metformin using poly(ε-caprolactone) (PCL) nanoparticles to simultaneously target multiple components of AMD [83][11]. Resveratrol provides antioxidant and anti-inflammatory effects, while metformin has anti-angiogenic effects. PCL is biocompatible in the RPE regions and degrades into less acidic by-products compared to other polymers. The polymer was further modified with cell-penetrating peptides (CPPs) to significantly improve retinal permeability. The sustained release of the therapy for up to 56 days and therapeutic effects were observed in a rat model of AMD. Lai et al. suggested a co-delivery system for berberine hydrochloride and chrysophanol using polyamidoamine dendrimers (PAMAM) and liposomes [54][12]. PAMAM acts as an external coating for the compound-loaded liposomes to enhance drug loading, cellular permeability, and bio-adhesion on corneal epithelial cells. The PAMAM-liposome system substantially improved berberine hydrochloride bioavailability. No side effects were observed on the rabbit ocular surface structure after the administration of P-CBLs. Although the release profiles of the drugs in the posterior segment of the eye were not assessed, the P-CBL system has potential for treating AMD and other ocular diseases.

 

4. Diabetic Retinopathy

Diabetic Retinopathy

Diabetic retinopathy is a chronic condition in diabetic patients that results from damage to the blood vessels in the retina. The condition progresses over time and has two stages: non-proliferative diabetic retinopathy (NPDR) and proliferative diabetic retinopathy (PDR). NPDR is characterized by increased vascular permeability, capillary occlusion, and the formation of microaneurysms, hemorrhages, cotton wool spots, and hard exudates. PDR, which occurs in advanced stages of diabetic retinopathy, leads to retinal ischemia and the release of pro-angiogenic factors such as VEGF, stimulating the formation of new abnormal blood vessels. These vessels can cause various vision-threatening complications such as vitreous hemorrhage and tractional retinal detachment, as well as glaucoma. Treatment of PDR primarily focuses on reducing the production of VEGF through laser photocoagulation or intravitreal anti-VEGF injections.

The treatment of diabetic retinopathy (DR) may involve the use of antioxidants, anti-inflammatory agents, and neurotrophic factors [84][13]. Nano-carriers, such as PLGA-based nanoparticles, have been suggested to improve drug delivery to the diabetic retina. For instance, Zeng et al. used PLGA nanoparticles to deliver Interleukin-12 (IL-12), which has been shown to reduce the levels of matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF-A), both of which contribute to DR severity [85][14]. IL-12-PNP exhibited better inhibition against VEGF-A and MMP-9 expression, resulting in significantly reduced 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) [86][15], which demonstrated neuroprotective and antioxidant effects on a model of glucose-induced DR on human retinal endothelial cells. The use of a biodegradable polymer in this restudyearch showed high encapsulation efficacy (79.8%) and no signs of cytotoxicity or ocular irritation in vivo. Romeo et al. observed a prolonged and sustained release for up to 8 days compared to a rapid burst release of free melatonin, which is necessary for its ocular delivery. These studies highlight the potential of nano-carriers in improving the therapeutic efficacy of drugs limited by inefficient delivery routes.

Zingale et al. used nanostructured lipid carriers (NLCs) to deliver diosmin, a flavonoid known for its anti-inflammatory, cytoprotective, and antioxidant effects in high glucose environments [87][16]. This delivery system achieved high encapsulation efficiency and was found safe and well-tolerated in vitro. However, surfactants are commonly used in the preparation of lipid-based nanocarriers, which may cause irritation and sensitizing action [88][17]. Further studies are being conducted to confirm the clinically relevant anti-inflammatory effects of diosmin NLCs. NLCs have the advantage of minimal toxicity and can be stored stably for long periods [89][18]. They can also be applied as topical eye drops, as demonstrated by Platania et al. [74][4], which greatly increases patient compliance. However, further exploration is needed to better assess the release profiles of drug loaded NLCs and determine how often administration is required.

Various types of biodegradable nanoparticles have been studied for the treatment of diabetic retinopathy. One study by Radwan et al. used bovine serum albumin nanoparticles coated with hyaluronic acid to deliver the anti-VEGF factor apatinib, resulting in sustained biphasic release with high mucoadhesion and improved retinal thickness and microstructure [90][19]. Another study by Mahaling et al. used nanoparticles with a hydrophobic polycaprolactone core and hydrophilic Pluronic® F68 shell to deliver triamcinolone acetonide, demonstrating significant structural and functional improvements in a DR rat model [91][20]. These findings indicate the potential of non-invasive, patient self-administered nanoparticle-based delivery systems for the treatment of diabetic retinopathy, overcoming challenges associated with intravitreal administration. The use of PCL as a biodegradable polymer for nanoparticle systems has been particularly advantageous in DR treatments, given its strong bioavailability in the retina during topical administration [92][21].

 

5. Diabetic Macular Edema (DME)

Diabetic Macular Edema (DME)

Diabetic macular edema (DME) is a frequent complication of diabetic retinopathy, characterized by the accumulation of fluid in the macula, resulting in a rapid decline in visual acuity. This is caused by increased permeability and inflammation in the retinal vessels [84][13].

In addition to intravitreal anti-VEGF injection and topical NSAIDs, intravitreal triamcinolone acetonide (TA) can be used to reduce inflammation associated with diabetic macular edema (DME), but it can lead to complications such as IOP elevation and cataract formation. To overcome these issues, liposomes have been used for topical delivery of TA. Navarro-Partida et al. found TA-loaded liposomes to be safe and tolerable in healthy patients through a Phase 1 clinical assay and reported a sustained therapeutic effect in DME patients in an open-label, non-randomized study. However, further studies are needed to confirm long-term safety and therapeutic efficacy. Chitosan-coated liposomes have been used by Khalil et al. to improve the biodegradability and mucoadhesion of liposomes, enhancing bioavailability and prolonging the release of TA in in vivo models. Although their efficiency of drug release was demonstrated on a CNV rat model, the authors recommend this DDS for any posterior segment disease, particularly DME, proliferative diabetic retinopathy, and CNV related to AMD. Lipid-based nanomaterials show initial clinical success with TA topical administration, demonstrating the superiority of lipid-based DDSs and the flexibility afforded by nano-based DDSs to improve retention time, permeability, encapsulation efficiency, and personalize treatment to the drug, disease, and area of interest.

 

[1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][73][74][75][76][77][78][79][80][81][82][83][84][85][86][87][88][89][90][91][92][93][94][95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110][111][112][113][114][115][116][117][118][119][120][121][122][123][124][125][126][127][128][129][130][131][132][133][134][135][136][137][138][139][140][141][142][143][144][145][146][147][148][149][150][151][152][153][154][155][156][157][158][159][160][161][162][163][164][165][166][167][168][169][170][171][172][173][174][175][176][177][178][179][180][181][182][183][184][185][186][187][188][189][190][191][192][193][194][195][196][197][198][199]

References

  1. Urtti, A. Challenges and Obstacles of Ocular Pharmacokinetics and Drug Delivery. Adv. Drug Deliv. Rev. 2006, 58, 1131–1135. [Google Scholar] [CrossRef]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.
  2. 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]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.
  3. Addo, R.T. Ocular Drug Delivery: Advances, Challenges and Applications. In Ocular Drug Delivery: Advances, Challenges and Applications; Springer: Berlin/Heidelberg, Germany, 2016; pp. 1–185. [Google Scholar] [CrossRef]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.
  4. Chang, A.Y.; Purt, B. Biochemistry, Tear Film. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]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.
  5. 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]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.
  6. Wu, K.Y.; Kulbay, M.; Tanasescu, C.; Jiao, B.; Nguyen, B.H.; Tran, S.D. An Overview of the Dry Eye Disease in Sjögren’s Syndrome Using Our Current Molecular Understanding. Int. J. Mol. Sci. 2023, 24, 1580. [Google Scholar] [CrossRef] [PubMed]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.
  7. Goel, M.; Picciani, R.G.; Lee, R.K.; Bhattacharya, S.K. Aqueous Humor Dynamics: A Review. Open Ophthalmol. J. 2010, 4, 52–59. [Google Scholar] [CrossRef] [PubMed]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.
  8. Chen, M.-S.; Hou, P.-K.; Tai, T.-Y.; Lin, B.J. Blood-Ocular Barriers. Tzu Chi Med. J. 2008, 20, 25–34. [Google Scholar] [CrossRef]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.
  9. Cunha-Vaz, J.; Bernardes, R.; Lobo, C. Blood-Retinal Barrier. Eur. J. Ophthalmol. 2011, 21, 3–9. [Google Scholar] [CrossRef] [PubMed]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.
  10. Bertens, C.J.F.; Gijs, M.; van den Biggelaar, F.J.H.M.; Nuijts, R.M.M.A. Topical Drug Delivery Devices: A Review. Exp. Eye Res. 2018, 168, 149–160. [Google Scholar] [CrossRef] [PubMed]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.
  11. Singh Malik, D.; Mital, N.; Kaur, G. Topical Drug Delivery Systems: A Patent Review. Expert Opin. Ther. Pat. 2016, 26, 213–228. [Google Scholar] [CrossRef]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.
  12. Farkouh, A.; Frigo, P.; Czejka, M. Systemic Side Effects of Eye Drops: A Pharmacokinetic Perspective. Clin. Ophthalmol. 2016, 10, 2433–2441. [Google Scholar] [CrossRef]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.
  13. Wu, W.; He, Z.; Zhang, Z.; Yu, X.; Song, Z.; Li, X. Intravitreal Injection of Rapamycin-Loaded Polymeric Micelles for Inhibition of Ocular Inflammation in Rat Model. Int. J. Pharm. 2016, 513, 238–246. [Google Scholar] [CrossRef] [PubMed]Oshitari, T. Neurovascular Impairment and Therapeutic Strategies in Diabetic Retinopathy. Int. J. Environ. Res. Public Health 2021, 19, 439.
  14. Abrishami, M.; Shariati, M.M.; Malaekeh-Nikouei, B.; Tajani, A.S.; Mahmoudi, A.; Abrishami, M.; Khameneh, B. Preparation and in Vivo Evaluation of Nanoliposomes Containing Vancomycin after Intravitreal Injection in Albino Rabbits. Iran. J. Basic Med. Sci. 2020, 23, 551–555. [Google Scholar] [CrossRef] [PubMed]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.
  15. García-Caballero, C.; Prieto-Calvo, E.; Checa-Casalengua, P.; García-Martín, E.; Polo-Llorens, V.; García-Feijoo, J.; Molina-Martínez, I.T.; Bravo-Osuna, I.; Herrero-Vanrell, R. Six Month Delivery of GDNF from PLGA/Vitamin E Biodegradable Microspheres after Intravitreal Injection in Rabbits. Eur. J. Pharm. Sci. 2017, 103, 19–26. [Google Scholar] [CrossRef]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.
  16. Park, J.G.; Callaway, N.F.; Ludwig, C.A.; Mahajan, V.B. Intravitreal Methotrexate and Fluocinolone Acetonide Implantation for Vogt-Koyanagi-Harada Uveitis. Am. J. Ophthalmol. Case Rep. 2020, 19, 100859. [Google Scholar] [CrossRef] [PubMed]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.
  17. Glendenning, A.; Crews, K.; Sturdivant, J.; deLong, M.A.; Kopczynski, C.; Lin, C.-W. Sustained Release, Biodegradable PEA Implants for Intravitreal Delivery of the ROCK/PKC Inhibitor AR-13503. Investig. Ophthalmol. Vis. Sci. 2018, 59, 5672. [Google Scholar]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.
  18. Blazaki, S.; Pachis, K.; Tzatzarakis, M.; Tsilimbaris, M.; Antimisiaris, S.G. Novel Liposome Aggregate Platform (LAP) System for Sustained Retention of Drugs in the Posterior Ocular Segment Following Intravitreal Injection. Int. J. Pharm. 2020, 576, 118987. [Google Scholar] [CrossRef]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.
  19. Agban, Y.; Thakur, S.S.; Mugisho, O.O.; Rupenthal, I.D. Depot Formulations to Sustain Periocular Drug Delivery to the Posterior Eye Segment. Drug Discov. Today 2019, 24, 1458–1469. [Google Scholar] [CrossRef]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.
  20. Radhakrishnan, K.; Vincent, A.; Joseph, R.R.; Moreno, M.; Dickescheid, A.; Agrawal, R.; Venkatraman, S. Hollow Microcapsules as Periocular Drug Depot for Sustained Release of Anti-VEGF Protein. Pharmaceutics 2019, 11, 330. [Google Scholar] [CrossRef]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.
  21. Yiu, G.; Chung, S.H.; Mollhoff, I.N.; Nguyen, U.T.; Thomasy, S.M.; Yoo, J.; Taraborelli, D.; Noronha, G. Suprachoroidal and Subretinal Injections of AAV Using Transscleral Microneedles for Retinal Gene Delivery in Nonhuman Primates. Mol. Ther. Methods Clin. Dev. 2020, 16, 179–191. [Google Scholar] [CrossRef]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.
  22. Weijtens, O.; Feron, E.J.; Schoemaker, R.C.; Cohen, A.F.; Lentjes, E.G.W.M.; Romijn, F.P.H.T.M.; Van Meurs, J.C. High Concentration of Dexamethasone in Aqueous and Vitreous after Subconjunctival Injection. Am. J. Ophthalmol. 1999, 128, 192–197. [Google Scholar] [CrossRef]
  23. Salama, H.A.; Ghorab, M.; Mahmoud, A.A.; Abdel Hady, M. PLGA Nanoparticles as Subconjunctival Injection for Management of Glaucoma. AAPS PharmSciTech 2017, 18, 2517–2528. [Google Scholar] [CrossRef] [PubMed]
  24. Fu, J.; Sun, F.; Liu, W.; Liu, Y.; Gedam, M.; Hu, Q.; Fridley, C.; Quigley, H.A.; Hanes, J.; Pitha, I. Subconjunctival Delivery of Dorzolamide-Loaded Poly(Ether-Anhydride) Microparticles Produces Sustained Lowering of Intraocular Pressure in Rabbits. Mol. Pharm. 2016, 13, 2987–2995. [Google Scholar] [CrossRef] [PubMed]
  25. Cuming, R.S.; Abarca, E.M.; Duran, S.; Wooldridge, A.A.; Stewart, A.J.; Ravis, W.; Babu, R.J.; Lin, Y.J.; Hathcock, T. Development of a Sustained-Release Voriconazole-Containing Thermogel for Subconjunctival Injection in Horses. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2746–2754. [Google Scholar] [CrossRef] [PubMed]
  26. Martínez-Carrasco, R.; Sánchez-Abarca, L.I.; Nieto-Gómez, C.; Martín García, E.; Sánchez-Guijo, F.; Argüeso, P.; Aijón, J.; Hernández-Galilea, E.; Velasco, A. Subconjunctival Injection of Mesenchymal Stromal Cells Protects the Cornea in an Experimental Model of GVHD. Ocul. Surf. 2019, 17, 285–294. [Google Scholar] [CrossRef] [PubMed]
  27. Lindholm, J.M.; Taipale, C.; Ylinen, P.; Tuuminen, R. Perioperative Subconjunctival Triamcinolone Acetonide Injection for Prevention of Inflammation and Macular Oedema after Cataract Surgery. Acta Ophthalmol. 2020, 98, 36–42. [Google Scholar] [CrossRef]
  28. Schneider, M.E.; Milstein, D.E.; Oyakawa, R.T.; Ober, R.R.; Campo, R. Ocular Perforation from a Retrobulbar Injection. Am. J. Ophthalmol. 1988, 106, 35–40. [Google Scholar] [CrossRef]
  29. Safi, M.; Ang, M.J.; Patel, P.; Silkiss, R.Z. Rhino-Orbital-Cerebral Mucormycosis (ROCM) and Associated Cerebritis Treated with Adjuvant Retrobulbar Amphotericin B. Am. J. Ophthalmol. Case Rep. 2020, 19, 100771. [Google Scholar] [CrossRef]
  30. Arbisser, L.B. Safety of Intracameral Moxifloxacin for Prophylaxis of Endophthalmitis after Cataract Surgery. J. Cataract. Refract. Surg. 2008, 34, 1114–1120. [Google Scholar] [CrossRef]
  31. Christopher, K.; Chauhan, A. Contact Lens Based Drug Delivery to the Posterior Segment Via Iontophoresis in Cadaver Rabbit Eyes. Pharm. Res. 2019, 36. [Google Scholar] [CrossRef]
  32. Jung, J.H.; Chiang, B.; Grossniklaus, H.E.; Prausnitz, M.R. Ocular Drug Delivery Targeted by Iontophoresis in the Suprachoroidal Space Using a Microneedle. J. Control. Release 2018, 277, 14–22. [Google Scholar] [CrossRef]
  33. See, G.L.; Sagesaka, A.; Torizuka, H.; Todo, H.; Sugibayashi, K. Iontophoresis-Aided Drug Delivery into the Eyeball via Eyelid Skin. J. Drug Deliv. Sci. Technol. 2018, 47, 380–385. [Google Scholar] [CrossRef]
  34. 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] [PubMed]
  35. Habot-Wilner, Z.; Noronha, G.; Wykoff, C.C. Suprachoroidally Injected Pharmacological Agents for the Treatment of Chorio-Retinal Diseases: A Targeted Approach. Acta Ophthalmol. 2019, 97, 460–472. [Google Scholar] [CrossRef]
  36. Allyn, M.M.; Luo, R.H.; Hellwarth, E.B.; Swindle-Reilly, K.E. Considerations for Polymers Used in Ocular Drug Delivery. Front. Med. 2022, 8, 787644. [Google Scholar] [CrossRef]
  37. Bodratti, A.M.; Alexandridis, P. Amphiphilic Block Copolymers in Drug Delivery: Advances in Formulation Structure and Performance. Expert. Opin. Drug Deliv. 2018, 15, 1085–1104. [Google Scholar] [CrossRef] [PubMed]
  38. Liu, W.; Borrell, M.A.; Venerus, D.C.; Mieler, W.F.; Kang-Mieler, J.J. Characterization of Biodegradable Microsphere-Hydrogel Ocular Drug Delivery System for Controlled and Extended Release of Ranibizumab. Transl. Vis. Sci. Technol. 2019, 8, 12. [Google Scholar] [CrossRef]
  39. Zhang, X.; Wei, D.; Xu, Y.; Zhu, Q. Hyaluronic Acid in Ocular Drug Delivery. Carbohydr. Polym. 2021, 264, 118006. [Google Scholar] [CrossRef]
  40. Orasugh, J.T.; Dutta, S.; Das, D.; Nath, J.; Pal, C.; Chattopadhyay, D. Utilization of Cellulose Nanocrystals (CNC) Biopolymer Nanocomposites in Ophthalmic Drug Delivery System (ODDS). J. Nanotechnol. Res. 2019, 1, 75–87. [Google Scholar]
  41. Gupta, B.; Mishra, V.; Gharat, S.; Momin, M.; Omri, A. Cellulosic Polymers for Enhancing Drug Bioavailability in Ocular Drug Delivery Systems. Pharmaceuticals 2021, 14, 1201. [Google Scholar] [CrossRef]
  42. Tavakolian, M.; Jafari, S.M.; van de Ven, T.G.M. A Review on Surface-Functionalized Cellulosic Nanostructures as Biocompatible Antibacterial Materials. Nanomicro Lett. 2020, 12, 73. [Google Scholar] [CrossRef]
  43. Junnuthula, V.; Boroujeni, A.S.; Cao, S.; Tavakoli, S.; Ridolfo, R.; Toropainen, E.; Ruponen, M.; van Hest, J.C.M.; Urtti, A. Intravitreal Polymeric Nanocarriers with Long Ocular Retention and Targeted Delivery to the Retina and Optic Nerve Head Region. Pharmaceutics 2021, 13, 445. [Google Scholar] [CrossRef] [PubMed]
  44. Kianersi, S.; Solouk, A.; Saber-Samandari, S.; Keshel, S.H.; Pasbakhsh, P. Alginate Nanoparticles as Ocular Drug Delivery Carriers. J. Drug Deliv. Sci. Technol. 2021, 66, 102889. [Google Scholar] [CrossRef]
  45. Wong, F.S.Y.; Tsang, K.K.; Chu, A.M.W.; Chan, B.P.; Yao, K.M.; Lo, A.C.Y. Injectable Cell-Encapsulating Composite Alginate-Collagen Platform with Inducible Termination Switch for Safer Ocular Drug Delivery. Biomaterials 2019, 201, 53–67. [Google Scholar] [CrossRef] [PubMed]
  46. Jiang, G.; Jia, H.; Qiu, J.; Mo, Z.; Wen, Y.; Zhang, Y.; Wen, Y.; Xie, Q.; Ban, J.; Lu, Z.; et al. PLGA Nanoparticle Platform for Trans-Ocular Barrier to Enhance Drug Delivery: A Comparative Study Based on the Application of Oligosaccharides in the Outer Membrane of Carriers. Int. J. Nanomed. 2020, 15, 9373–9387. [Google Scholar] [CrossRef] [PubMed]
  47. Swetledge, S.; Carter, R.; Stout, R.; Astete, C.E.; Jung, J.P.; Sabliov, C.M. Stability and Ocular Biodistribution of Topically Administered PLGA Nanoparticles. Sci. Rep. 2021, 11, 12270. [Google Scholar] [CrossRef]
  48. Weng, Y.H.; Ma, X.W.; Che, J.; Li, C.; Liu, J.; Chen, S.Z.; Wang, Y.Q.; Gan, Y.L.; Chen, H.; Hu, Z.B.; et al. Nanomicelle-Assisted Targeted Ocular Delivery with Enhanced Antiinflammatory Efficacy In Vivo. Adv. Sci. 2017, 5, 1700455. [Google Scholar] [CrossRef]
  49. Dai, L.; Li, X.; Yao, M.; Niu, P.; Yuan, X.; Li, K.; Chen, M.; Fu, Z.; Duan, X.; Liu, H.; et al. Programmable Prodrug Micelle with Size-Shrinkage and Charge-Reversal for Chemotherapy-Improved IDO Immunotherapy. Biomaterials 2020, 241, 119901. [Google Scholar] [CrossRef]
  50. Hwang, D.; Ramsey, J.D.; Kabanov, A.V. Polymeric Micelles for the Delivery of Poorly Soluble Drugs: From Nanoformulation to Clinical Approval. Adv. Drug Deliv. Rev. 2020, 156, 80–118. [Google Scholar] [CrossRef] [PubMed]
  51. Sun, H.; Meng, F.; Cheng, R.; Deng, C.; Zhong, Z. Reduction-Responsive Polymeric Micelles and Vesicles for Triggered Intracellular Drug Release. Antioxid. Redox Signal. 2014, 21, 755–767. [Google Scholar] [CrossRef] [PubMed]
  52. Yu, Y.; Chen, D.; Li, Y.; Yang, W.; Tu, J.; Shen, Y. Improving the Topical Ocular Pharmacokinetics of Lyophilized Cyclosporine A-Loaded Micelles: Formulation, in Vitro and in Vivo Studies. Drug Deliv. 2018, 25, 888–899. [Google Scholar] [CrossRef]
  53. 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] [PubMed]
  54. 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. [Google Scholar] [CrossRef] [PubMed]
  55. Abdi, F.; Arkan, E.; Mansouri, K.; Shekarbeygi, Z.; Barzegari, E. Interactions of Bevacizumab with Chitosan Biopolymer Nanoparticles: Molecular Modeling and Spectroscopic Study. J. Mol. Liq. 2021, 339, 116655. [Google Scholar] [CrossRef]
  56. Ishida, T.; Harashima, H.; Kiwada, H. Liposome Clearance. Biosci. Rep. 2002, 22, 197–224. [Google Scholar] [CrossRef] [PubMed]
  57. Jin, X.; Zhu, L.; Xue, B.; Zhu, X.; Yan, D. Supramolecular Nanoscale Drug-Delivery System with Ordered Structure. Natl. Sci. Rev. 2019, 6, 1128–1137. [Google Scholar] [CrossRef] [PubMed]
  58. Ji, T.; Kohane, D.S. Nanoscale Systems for Local Drug Delivery. Nano Today 2019, 28, 100765. [Google Scholar] [CrossRef]
  59. Di, J.; Gao, X.; Du, Y.; Zhang, H.; Gao, J.; Zheng, A. Size, Shape, Charge and “Stealthy” Surface: Carrier Properties Affect the Drug Circulation Time in Vivo. Asian J. Pharm. Sci. 2021, 16, 444–458. [Google Scholar] [CrossRef]
  60. Rebibo, L.; Tam, C.; Sun, Y.; Shoshani, E.; Badihi, A.; Nassar, T.; Benita, S. Topical Tacrolimus Nanocapsules Eye Drops for Therapeutic Effect Enhancement in Both Anterior and Posterior Ocular Inflammation Models. J. Control. Release 2021, 333, 283–297. [Google Scholar] [CrossRef]
  61. Duan, Y.; Dhar, A.; Patel, C.; Khimani, M.; Neogi, S.; Sharma, P.; Siva Kumar, N.; 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]
  62. Lancina, M.G.; Yang, H. Dendrimers for Ocular Drug Delivery. Can. J. Chem. 2017, 95, 897–902. [Google Scholar] [CrossRef]
  63. Lin, D.; Lei, L.; Shi, S.; Li, X. Stimulus-Responsive Hydrogel for Ophthalmic Drug Delivery. Macromol. Biosci. 2019, 19, e1900001. [Google Scholar] [CrossRef] [PubMed]
  64. Lynch, C.R.; Kondiah, P.P.D.; Choonara, Y.E.; du Toit, L.C.; Ally, N.; Pillay, V. Hydrogel Biomaterials for Application in Ocular Drug Delivery. Front. Bioeng. Biotechnol. 2020, 8, 228. [Google Scholar] [CrossRef] [PubMed]
  65. Nagai, N.; Otake, H. Novel Drug Delivery Systems for the Management of Dry Eye. Adv. Drug Deliv. Rev. 2022, 191, 114582. [Google Scholar] [CrossRef] [PubMed]
  66. Jacob, S.; Nair, A.B.; Shah, J. Emerging Role of Nanosuspensions in Drug Delivery Systems. Biomater. Res. 2020, 24, 3. [Google Scholar] [CrossRef]
  67. Xie, J.; Luo, Y.; Liu, Y.; Ma, Y.; Yue, P.; Yang, M. Novel Redispersible Nanosuspensions Stabilized by Co-Processed Nanocrystalline Cellulose-Sodium Carboxymethyl Starch for Enhancing Dissolution and Oral Bioavailability of Baicalin. Int. J. Nanomed. 2019, 14, 353–369. [Google Scholar] [CrossRef]
  68. Gade, S.S.; Pentlavalli, S.; Mishra, D.; Vora, L.K.; Waite, D.; Alvarez-Lorenzo, C.I.; Herrero Vanrell, M.R.; Laverty, G.; Larraneta, E.; Donnelly, R.F.; et al. Injectable Depot Forming Thermoresponsive Hydrogel for Sustained Intrascleral Delivery of Sunitinib Using Hollow Microneedles. J. Ocul. Pharmacol. Ther. 2022, 38, 433–448. [Google Scholar] [CrossRef]
  69. 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]
  70. 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. [Google Scholar] [CrossRef]
  71. 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. [Google Scholar] [CrossRef]
  72. 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. [Google Scholar] [CrossRef]
  73. 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. [Google Scholar] [CrossRef] [PubMed]
  74. 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. [Google Scholar] [CrossRef] [PubMed]
  75. 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. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar] [CrossRef] [PubMed]
  77. 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. [Google Scholar] [CrossRef] [PubMed]
  78. 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. [Google Scholar] [CrossRef]
  79. 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. [Google Scholar] [CrossRef]
  80. 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. [Google Scholar] [CrossRef]
  81. 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. [Google Scholar] [CrossRef]
  82. 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. [Google Scholar] [CrossRef]
  83. 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. [Google Scholar] [CrossRef]
  84. Oshitari, T. Neurovascular Impairment and Therapeutic Strategies in Diabetic Retinopathy. Int. J. Environ. Res. Public Health 2021, 19, 439. [Google Scholar] [CrossRef]
  85. 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. [Google Scholar] [CrossRef]
  86. 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. [Google Scholar] [CrossRef]
  87. 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]
  88. 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]
  89. 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. [Google Scholar] [CrossRef] [PubMed]
  90. 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. [Google Scholar] [CrossRef]
  91. 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. [Google Scholar] [CrossRef] [PubMed]
  92. 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. [Google Scholar] [CrossRef] [PubMed]
  93. 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. [Google Scholar] [CrossRef] [PubMed]
  94. 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. [Google Scholar] [CrossRef] [PubMed]
  95. 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. [Google Scholar] [CrossRef]
  96. Alambiaga-Caravaca, A.M.; Domenech-Monsell, I.M.; Sebastián-Morelló, M.; Calatayud-Pascual, M.A.; Merino, V.; Rodilla, V.; López-Castellano, A. Development, Characterization, and Ex Vivo Evaluation of an Insert for the Ocular Administration of Progesterone. Int. J. Pharm. 2021, 606, 120921. [Google Scholar] [CrossRef] [PubMed]
  97. Yadav, M.; Schiavone, N.; Guzman-Aranguez, A.; Giansanti, F.; Papucci, L.; Perez de Lara, M.J.; Singh, M.; Kaur, I.P. Correction to: Atorvastatin-Loaded Solid Lipid Nanoparticles as Eye Drops: Proposed Treatment Option for Age-Related Macular Degeneration (AMD). Drug Deliv. Transl. Res. 2020, 10, 1531. [Google Scholar] [CrossRef]
  98. Mitra, R.N.; Gao, R.; Zheng, M.; Wu, M.J.; Voinov, M.A.; Smirnov, A.I.; Smirnova, T.I.; Wang, K.; Chavala, S.; Han, Z. Glycol Chitosan Engineered Autoregenerative Antioxidant Significantly Attenuates Pathological Damages in Models of Age-Related Macular Degeneration. ACS Nano 2017, 11, 4669–4685. [Google Scholar] [CrossRef]
  99. Nagai, N.; Daigaku, R.; Motoyama, R.; Kaji, H.; Abe, T. Release of Ranibizumab Using a Porous Poly(Dimethylsiloxane) Capsule Suppressed Laser-Induced Choroidal Neovascularization via the Transscleral Route. J. Mater. Sci. Mater. Med. 2022, 34, 5. [Google Scholar] [CrossRef] [PubMed]
  100. Qiu, F.; Meng, T.; Chen, Q.; Zhou, K.; Shao, Y.; Matlock, G.; Ma, X.; Wu, W.; Du, Y.; Wang, X.; et al. Fenofibrate-Loaded Biodegradable Nanoparticles for the Treatment of Experimental Diabetic Retinopathy and Neovascular Age-Related Macular Degeneration. Mol. Pharm. 2019, 16, 1958–1970. [Google Scholar] [CrossRef]
  101. Sun, Q.; Shen, Y.; Su, L.; Xu, X. Inhibition of Pathological Retinal Neovascularization by a Small Peptide Derived from Human Tissue-Type Plasminogen Kringle 2. Front. Pharmacol. 2020, 10, 1639. [Google Scholar] [CrossRef]
  102. Ishibazawa, A.; Nagaoka, T.; Yokota, H.; Takahashi, A.; Omae, T.; Song, Y.S.; Takahashi, T.; Yoshida, A. Characteristics of Retinal Neovascularization in Proliferative Diabetic Retinopathy Imaged by Optical Coherence Tomography Angiography. Investig. Ophthalmol. Vis. Sci. 2016, 57, 6247–6255. [Google Scholar] [CrossRef]
  103. Sacconi, R.; Fragiotta, S.; Sarraf, D.; Sadda, S.V.R.; Freund, K.B.; Parravano, M.; Corradetti, G.; Cabral, D.; Capuano, V.; Miere, A.; et al. Towards a Better Understanding of Non-Exudative Choroidal and Macular Neovascularization. Prog. Retin. Eye Res. 2023, 92, 101113. [Google Scholar] [CrossRef] [PubMed]
  104. Fernandes, A.R.; dos Santos, T.; Granja, P.L.; Sanchez-Lopez, E.; Garcia, M.L.; Silva, A.M.; Souto, E.B. Permeability, Anti-Inflammatory and Anti-VEGF Profiles of Steroidal-Loaded Cationic Nanoemulsions in Retinal Pigment Epithelial Cells under Oxidative Stress. Int. J. Pharm. 2022, 617, 121615. [Google Scholar] [CrossRef] [PubMed]
  105. García-Estrada, P.; García-Bon, M.A.; López-Naranjo, E.J.; Basaldúa-Pérez, D.N.; Santos, A.; Navarro-Partida, J. Polymeric Implants for the Treatment of Intraocular Eye Diseases: Trends in Biodegradable and Non-Biodegradable Materials. Pharmaceutics 2021, 13, 701. [Google Scholar] [CrossRef] [PubMed]
  106. Gil-Martínez, M.; Santos-Ramos, P.; Fernández-Rodríguez, M.; Abraldes, M.J.; Rodríguez-Cid, M.J.; Santiago-Varela, M.; Fernández-Ferreiro, A.; Gómez-Ulla, F. Pharmacological Advances in the Treatment of Age-Related Macular Degeneration. Curr. Med. Chem. 2020, 27, 583–598. [Google Scholar] [CrossRef]
  107. Ferrara, N.; Adamis, A.P. Ten Years of Anti-Vascular Endothelial Growth Factor Therapy. Nat. Rev. Drug Discov. 2016, 15, 385–403. [Google Scholar] [CrossRef]
  108. Klettner, A.; Recber, M.; Roider, J. Comparison of the Efficacy of Aflibercept, Ranibizumab, and Bevacizumab in an RPE/Choroid Organ Culture. Graefes Arch. Clin. Exp. Ophthalmol. 2014, 252, 1593–1598. [Google Scholar] [CrossRef] [PubMed]
  109. Oo, C.; Kalbag, S.S. Leveraging the Attributes of Biologics and Small Molecules, and Releasing the Bottlenecks: A New Wave of Revolution in Drug Development. Expert. Rev. Clin. Pharmacol. 2016, 9, 747–749. [Google Scholar] [CrossRef]
  110. Li, J.; Mooney, D.J. Designing Hydrogels for Controlled Drug Delivery. Nat. Rev. Mater. 2016, 1, 16071. [Google Scholar] [CrossRef] [PubMed]
  111. Galante, R.; Pinto, T.J.A.; Colaço, R.; Serro, A.P. Sterilization of Hydrogels for Biomedical Applications: A Review. J. Biomed. Mater. Res. B Appl. Biomater. 2018, 106, 2472–2492. [Google Scholar] [CrossRef]
  112. Bordbar-Khiabani, A.; Gasik, M. Smart Hydrogels for Advanced Drug Delivery Systems. Int. J. Mol. Sci. 2022, 23, 3665. [Google Scholar] [CrossRef]
  113. Osswald, C.R.; Kang-Mieler, J.J. Controlled and Extended In Vitro Release of Bioactive Anti-Vascular Endothelial Growth Factors from a Microsphere-Hydrogel Drug Delivery System. Curr. Eye Res. 2016, 41, 1216–1222. [Google Scholar] [CrossRef] [PubMed]
  114. Osswald, C.R.; Guthrie, M.J.; Avila, A.; Valio, J.A.; Mieler, W.F.; Kang-Mieler, J.J. In Vivo Efficacy of an Injectable Microsphere-Hydrogel Ocular Drug Delivery System. Curr. Eye Res. 2017, 42, 1293–1301. [Google Scholar] [CrossRef] [PubMed]
  115. Hu, C.C.; Chiu, Y.C.; Chaw, J.R.; Chen, C.F.; Liu, H.W. Thermo-Responsive Hydrogel as an Anti-VEGF Drug Delivery System to Inhibit Retinal Angiogenesis in Rex Rabbits. Technol. Health Care 2019, 27, S153–S163. [Google Scholar] [CrossRef]
  116. Xue, K.; Zhao, X.; Zhang, Z.; Qiu, B.; Tan, Q.S.W.; Ong, K.H.; Liu, Z.; Parikh, B.H.; Barathi, V.A.; Yu, W.; et al. Sustained Delivery of Anti-VEGFs from Thermogel Depots Inhibits Angiogenesis without the Need for Multiple Injections. Biomater. Sci. 2019, 7, 4603–4614. [Google Scholar] [CrossRef]
  117. Liu, W.; Lee, B.S.; Mieler, W.F.; Kang-Mieler, J.J. Biodegradable Microsphere-Hydrogel Ocular Drug Delivery System for Controlled and Extended Release of Bioactive Aflibercept In Vitro. Curr. Eye Res. 2019, 44, 264–274. [Google Scholar] [CrossRef]
  118. Liu, W.; Tawakol, A.P.; Rudeen, K.M.; Mieler, W.F.; Kang-Mieler, J.J. Treatment Efficacy and Biocompatibility of a Biodegradable Aflibercept-Loaded Microsphere-Hydrogel Drug Delivery System. Transl. Vis. Sci. Technol. 2020, 9, 13. [Google Scholar] [CrossRef]
  119. Fan, W.; Li, S.; Tao, J.; Yu, C.; Sun, M.; Xie, Z.; Wu, X.; Ge, L.; Wu, Y.; Liu, Y. Anti-Vascular Endothelial Growth Factor Drug Conbercept-Loaded Peptide Hydrogel Reduced Angiogenesis in the Neovascular Age-Related Macular Degeneration. J. Biomed. Nanotechnol. 2022, 18, 277–287. [Google Scholar] [CrossRef] [PubMed]
  120. Li, J.; Tian, Q.; Sun, H.; Zhang, Y.; Yang, X.; Kaur, P.; Wang, R.; Fang, Y.; Yan, H.; Du, X.; et al. A Novel, Liposome-Loaded, Injectable Hydrogel for Enhanced Treatment of Choroidal Neovascularization by Sub-Tenon’s Injection. Mater. Today Nano 2022, 20, 100264. [Google Scholar] [CrossRef]
  121. Tanetsugu, Y.; Tagami, T.; Terukina, T.; Ogawa, T.; Ohta, M.; Ozeki, T. Development of a Sustainable Release System for a Ranibizumab Biosimilar Using Poly(Lactic-Co-Glycolic Acid) Biodegradable Polymer-Based Microparticles as a Platform. Biol. Pharm. Bull. 2017, 40, 145–150. [Google Scholar] [CrossRef]
  122. Sousa, F.; Cruz, A.; Fonte, P.; Pinto, I.M.; Neves-Petersen, M.T.; Sarmento, B. A New Paradigm for Antiangiogenic Therapy through Controlled Release of Bevacizumab from PLGA Nanoparticles. Sci. Rep. 2017, 7, 3736. [Google Scholar] [CrossRef]
  123. Sousa, F.; Cruz, A.; Pinto, I.M.; Sarmento, B. Nanoparticles Provide Long-Term Stability of Bevacizumab Preserving Its Antiangiogenic Activity. Acta Biomater. 2018, 78, 285–295. [Google Scholar] [CrossRef]
  124. Zhang, X.P.; Sun, J.G.; Yao, J.; Shan, K.; Liu, B.H.; Yao, M.D.; Ge, H.M.; Jiang, Q.; Zhao, C.; Yan, B. Effect of Nanoencapsulation Using Poly (Lactide-Co-Glycolide) (PLGA) on Anti-Angiogenic Activity of Bevacizumab for Ocular Angiogenesis Therapy. Biomed. Pharmacother. 2018, 107, 1056–1063. [Google Scholar] [CrossRef] [PubMed]
  125. Kelly, S.J.; Hirani, A.; Shahidadpury, V.; Solanki, A.; Halasz, K.; Gupta, S.V.; Madow, B.; Sutariya, V. Aflibercept Nanoformulation Inhibits VEGF Expression in Ocular In Vitro Model: A Preliminary Report. Biomedicines 2018, 6, 92. [Google Scholar] [CrossRef] [PubMed]
  126. Yan, J.; Peng, X.; Cai, Y.; Cong, W. Development of Facile Drug Delivery Platform of Ranibizumab Fabricated PLGA-PEGylated Magnetic Nanoparticles for Age-Related Macular Degeneration Therapy. J. Photochem. Photobiol. B 2018, 183, 133–136. [Google Scholar] [CrossRef] [PubMed]
  127. Liu, J.; Zhang, X.; Li, G.; Xu, F.; Li, S.; Teng, L.; Li, Y.; Sun, F. Anti-Angiogenic Activity Of Bevacizumab-Bearing Dexamethasone-Loaded PLGA Nanoparticles For Potential Intravitreal Applications. Int. J. Nanomed. 2019, 14, 8819–8834. [Google Scholar] [CrossRef]
  128. Heljak, M.K.; Swieszkowski, W. In Silico Model of Bevacizumab Sustained Release from Intravitreal Administrated PLGA Drug-Loaded Microspheres. Mater. Lett. 2021, 307, 131080. [Google Scholar] [CrossRef]
  129. Liu, J.; Li, S.; Li, G.; Li, X.; Yu, C.; Fu, Z.; Li, X.; Teng, L.; Li, Y.; Sun, F. Highly Bioactive, Bevacizumab-Loaded, Sustained-Release PLGA/PCADK Microspheres for Intravitreal Therapy in Ocular Diseases. Int. J. Pharm. 2019, 563, 228–236. [Google Scholar] [CrossRef]
  130. Tsujinaka, H.; Fu, J.; Shen, J.; Yu, Y.; Hafiz, Z.; Kays, J.; McKenzie, D.; Cardona, D.; Culp, D.; Peterson, W.; et al. Sustained Treatment of Retinal Vascular Diseases with Self-Aggregating Sunitinib Microparticles. Nat. Commun. 2020, 11, 694. [Google Scholar] [CrossRef] [PubMed]
  131. Luo, L.; Yang, J.; Oh, Y.; Hartsock, M.J.; Xia, S.; Kim, Y.C.; Ding, Z.; Meng, T.; Eberhart, C.G.; Ensign, L.M.; et al. Controlled Release of Corticosteroid with Biodegradable Nanoparticles for Treating Experimental Autoimmune Uveitis. J. Control. Release 2019, 296, 68–80. [Google Scholar] [CrossRef] [PubMed]
  132. Cai, W.; Chen, Q.; Shen, T.; Yang, Q.; Hu, W.; Zhao, P.; Yu, J. Intravenous Anti-VEGF Agents with RGD Peptide-Targeted Core Cross-Linked Star (CCS) Polymers Modified with Indocyanine Green for Imaging and Treatment of Laser-Induced Choroidal Neovascularization. Biomater. Sci. 2020, 8, 4481–4491. [Google Scholar] [CrossRef]
  133. Luis de Redín, I.; Boiero, C.; Martínez-Ohárriz, M.C.; Agüeros, M.; Ramos, R.; Peñuelas, I.; Allemandi, D.; Llabot, J.M.; Irache, J.M. Human Serum Albumin Nanoparticles for Ocular Delivery of Bevacizumab. Int. J. Pharm. 2018, 541, 214–223. [Google Scholar] [CrossRef]
  134. Luis de Redín, I.; Boiero, C.; Recalde, S.; Agüeros, M.; Allemandi, D.; Llabot, J.M.; García-Layana, A.; Irache, J.M. In Vivo Effect of Bevacizumab-Loaded Albumin Nanoparticles in the Treatment of Corneal Neovascularization. Exp. Eye Res. 2019, 185, 107697. [Google Scholar] [CrossRef]
  135. Llabot, J.M.; Luis de Redin, I.; Agüeros, M.; Dávila Caballero, M.J.; Boiero, C.; Irache, J.M.; Allemandi, D. In Vitro Characterization of New Stabilizing Albumin Nanoparticles as a Potential Topical Drug Delivery System in the Treatment of Corneal Neovascularization (CNV). J. Drug Deliv. Sci. Technol. 2019, 52, 379–385. [Google Scholar] [CrossRef]
  136. Pandit, J.; Sultana, Y.; Aqil, M. Chitosan-Coated PLGA Nanoparticles of Bevacizumab as Novel Drug Delivery to Target Retina: Optimization, Characterization, and in Vitro Toxicity Evaluation. Artif. Cells Nanomed. Biotechnol. 2017, 45, 1397–1407. [Google Scholar] [CrossRef]
  137. Ugurlu, N.; Aşık, M.D.; Çakmak, H.B.; Tuncer, S.; Turk, M.; Çagıl, N.; Denkbas, E.B. Transscleral Delivery of Bevacizumab-Loaded Chitosan Nanoparticles. J. Biomed. Nanotechnol. 2019, 15, 830–838. [Google Scholar] [CrossRef]
  138. Savin, C.L.; Popa, M.; Delaite, C.; Costuleanu, M.; Costin, D.; Peptu, C.A. Chitosan Grafted-Poly(Ethylene Glycol) Methacrylate Nanoparticles as Carrier for Controlled Release of Bevacizumab. Mater. Sci. Eng. C Mater. Biol. Appl. 2019, 98, 843–860. [Google Scholar] [CrossRef] [PubMed]
  139. Jiang, P.; Chaparro, F.J.; Cuddington, C.T.; Palmer, A.F.; Ohr, M.P.; Lannutti, J.J.; Swindle-Reilly, K.E. Injectable Biodegradable Bi-Layered Capsule for Sustained Delivery of Bevacizumab in Treating Wet Age-Related Macular Degeneration. J. Control. Release 2020, 320, 442–456. [Google Scholar] [CrossRef] [PubMed]
  140. Jiang, P.; Jacobs, K.M.; Ohr, M.P.; Swindle-Reilly, K.E. Chitosan-Polycaprolactone Core-Shell Microparticles for Sustained Delivery of Bevacizumab. Mol. Pharm. 2020, 17, 2570–2584. [Google Scholar] [CrossRef]
  141. Chaharband, F.; Daftarian, N.; Kanavi, M.R.; Varshochian, R.; Hajiramezanali, M.; Norouzi, P.; Arefian, E.; Atyabi, F.; Dinarvand, R. Trimethyl Chitosan-Hyaluronic Acid Nano-Polyplexes for Intravitreal VEGFR-2 SiRNA Delivery: Formulation and in Vivo Efficacy Evaluation. Nanomedicine 2020, 26, 102181. [Google Scholar] [CrossRef]
  142. Formica, M.L.; Legeay, S.; Bejaud, J.; Montich, G.G.; Ullio Gamboa, G.V.; Benoit, J.P.; Palma, S.D. Novel Hybrid Lipid Nanocapsules Loaded with a Therapeutic Monoclonal Antibody—Bevacizumab—And Triamcinolone Acetonide for Combined Therapy in Neovascular Ocular Pathologies. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 119, 111398. [Google Scholar] [CrossRef]
  143. Mu, H.; Wang, Y.; Chu, Y.; Jiang, Y.; Hua, H.; Chu, L.; Wang, K.; Wang, A.; Liu, W.; Li, Y.; et al. Multivesicular Liposomes for Sustained Release of Bevacizumab in Treating Laser-Induced Choroidal Neovascularization. Drug Deliv. 2018, 25, 1372–1383. [Google Scholar] [CrossRef] [PubMed]
  144. Karumanchi, D.K.; Skrypai, Y.; Thomas, A.; Gaillard, E.R. Rational Design of Liposomes for Sustained Release Drug Delivery of Bevacizumab to Treat Ocular Angiogenesis. J. Drug Deliv. Sci. Technol. 2018, 47, 275–282. [Google Scholar] [CrossRef]
  145. de Cristo Soares Alves, A.; Lavayen, V.; Figueiró, F.; Dallemole, D.R.; de Fraga Dias, A.; Cé, R.; Battastini, A.M.O.; Guterres, S.S.; Pohlmann, A.R. Chitosan-Coated Lipid-Core Nanocapsules Functionalized with Gold-III and Bevacizumab Induced In Vitro Cytotoxicity against C6 Cell Line and In Vivo Potent Antiangiogenic Activity. Pharm. Res. 2020, 37, 91. [Google Scholar] [CrossRef] [PubMed]
  146. Jiang, P.; Choi, A.; Swindle-Reilly, K.E. Controlled Release of Anti-VEGF by Redox-Responsive Polydopamine Nanoparticles. Nanoscale 2020, 12, 17298–17311. [Google Scholar] [CrossRef] [PubMed]
  147. Webber, A.L.; Sharwood, P. Practical Use and Prescription of Ocular Medications in Children and Infants. Clin. Exp. Optom. 2021, 104, 385–395. [Google Scholar] [CrossRef]
  148. Sheybani, N.D.; Yang, H. Pediatric Ocular Nanomedicines: Challenges and Opportunities. Chin. Chem. Lett. 2017, 28, 1817–1821. [Google Scholar] [CrossRef]
  149. Sunita, M.; Manisha, S.; Sanjeev, M.K.; Ravi, K.S.; Aarzoo, J.; Ajai, A. Anatomical and Clinical Characteristics of Paediatric and Adult Eyes. Natl. J. Clin. Anat. 2021, 10, 5. [Google Scholar] [CrossRef]
  150. Mudgil, P.; Borchman, D.; Ramasubramanian, A. Insights into Tear Film Stability from Babies and Young Adults: A Study of Human Meibum Lipid Conformation and Rheology. Int. J. Mol. Sci. 2018, 19, 3502. [Google Scholar] [CrossRef]
  151. Chidi-Egboka, N.C.; Briggs, N.E.; Jalbert, I.; Golebiowski, B. The Ocular Surface in Children: A Review of Current Knowledge and Meta-Analysis of Tear Film Stability and Tear Secretion in Children. Ocul. Surf. 2019, 17, 28–39. [Google Scholar] [CrossRef]
  152. Mutlu, F.M.; Sarici, S.U. Treatment of Retinopathy of Prematurity: A Review of Conventional and Promising New Therapeutic Options. Int. J. Ophthalmol. 2013, 6, 228–236. [Google Scholar] [CrossRef]
  153. Mukherjee, P.; Bhattacharya, R.; Wang, P.; Wang, L.; Basu, S.; Nagy, J.A.; Atala, A.; Mukhopadhyay, D.; Soker, S. Antiangiogenic Properties of Gold Nanoparticles. Clin. Cancer Res. 2005, 11, 3530–3534. [Google Scholar] [CrossRef]
  154. Saeed, B.A.; Lim, V.; Yusof, N.A.; Khor, K.Z.; Rahman, H.S.; Samad, N.A. Antiangiogenic Properties of Nanoparticles: A Systematic Review. Int. J. Nanomed. 2019, 14, 5135–5146. [Google Scholar] [CrossRef]
  155. Kim, J.H.; Kim, M.H.; Jo, D.H.; Yu, Y.S.; Lee, T.G.; Kim, J.H. The Inhibition of Retinal Neovascularization by Gold Nanoparticles via Suppression of VEGFR-2 Activation. Biomaterials 2011, 32, 1865–1871. [Google Scholar] [CrossRef] [PubMed]
  156. Kolosnjaj-Tabi, J.; Volatron, J.; Gazeau, F. Basic Principles of in Vivo Distribution, Toxicity, and Degradation of Prospective Inorganic Nanoparticles for Imaging. In Design and Applications of Nanoparticles in Biomedical Imaging; Springer: Cham, Switzerland, 2016; pp. 9–41. [Google Scholar] [CrossRef]
  157. Radomska, A.; Leszczyszyn, J.; Radomski, M.W. The Nanopharmacology and Nanotoxicology of Nanomaterials: New Opportunities and Challenges. Adv. Clin. Exp. Med. 2016, 25, 151–162. [Google Scholar] [CrossRef] [PubMed]
  158. Higbee-Dempsey, E.M.; Amirshaghaghi, A.; Case, M.J.; Bouché, M.; Kim, J.; Cormode, D.P.; Tsourkas, A. Biodegradable Gold Nanoclusters with Improved Excretion Due to PH-Triggered Hydrophobic-to-Hydrophilic Transition. J. Am. Chem. Soc. 2020, 142, 7783–7794. [Google Scholar] [CrossRef] [PubMed]
  159. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering Precision Nanoparticles for Drug Delivery. Nat. Rev. Drug Discov. 2020, 20, 101–124. [Google Scholar] [CrossRef] [PubMed]
  160. Bohley, M.; Dillinger, A.E.; Schweda, F.; Ohlmann, A.; Braunger, B.M.; Tamm, E.R.; Goepferich, A. A Single Intravenous Injection of Cyclosporin A-Loaded Lipid Nanocapsules Prevents Retinopathy of Prematurity. Sci. Adv. 2022, 8. [Google Scholar] [CrossRef]
  161. Li, Q.; Weng, J.; Wong, S.N.; Thomas Lee, W.Y.; Chow, S.F. Nanoparticulate Drug Delivery to the Retina. Mol. Pharm. 2021, 18, 506–521. [Google Scholar] [CrossRef]
  162. Bertrand, N.; Leroux, J.C. The Journey of a Drug-Carrier in the Body: An Anatomo-Physiological Perspective. J. Control. Release 2012, 161, 152–163. [Google Scholar] [CrossRef]
  163. Hagigit, T.; Abdulrazik, M.; Valamanesh, F.; Behar-Cohen, F.; Benita, S. Ocular Antisense Oligonucleotide Delivery by Cationic Nanoemulsion for Improved Treatment of Ocular Neovascularization: An in-Vivo Study in Rats and Mice. J. Control. Release 2012, 160, 225–231. [Google Scholar] [CrossRef]
  164. Hagigit, T.; Nassar, T.; Behar-Cohen, F.; Lambert, G.; Benita, S. The Influence of Cationic Lipid Type on In-Vitro Release Kinetic Profiles of Antisense Oligonucleotide from Cationic Nanoemulsions. Eur. J. Pharm. Biopharm. 2008, 70, 248–259. [Google Scholar] [CrossRef]
  165. Hagigit, T.; Abdulrazik, M.; Orucov, F.; Valamanesh, F.; Lambert, M.; Lambert, G.; Behar-Cohen, F.; Benita, S. Topical and Intravitreous Administration of Cationic Nanoemulsions to Deliver Antisense Oligonucleotides Directed towards VEGF KDR Receptors to the Eye. J. Control. Release 2010, 145, 297–305. [Google Scholar] [CrossRef] [PubMed]
  166. Xu, W.; Wu, Y.; Hu, Z.; Sun, L.; Dou, G.; Zhang, Z.; Wang, H.; Guo, C.; Wang, Y. Exosomes from Microglia Attenuate Photoreceptor Injury and Neovascularization in an Animal Model of Retinopathy of Prematurity. Mol. Ther. Nucleic Acids 2019, 16, 778–790. [Google Scholar] [CrossRef] [PubMed]
  167. Moisseiev, E.; Anderson, J.D.; Oltjen, S.; Goswami, M.; Zawadzki, R.J.; Nolta, J.A.; Park, S.S. Protective Effect of Intravitreal Administration of Exosomes Derived from Mesenchymal Stem Cells on Retinal Ischemia. Curr. Eye Res. 2017, 42, 1358–1367. [Google Scholar] [CrossRef] [PubMed]
  168. Ebneter, A.; Kokona, D.; Schneider, N.; Zinkernagel, M.S. Microglia Activation and Recruitment of Circulating Macrophages During Ischemic Experimental Branch Retinal Vein Occlusion. Investig. Ophthalmol. Vis. Sci. 2017, 58, 944–953. [Google Scholar] [CrossRef]
  169. Deliyanti, D.; Talia, D.M.; Zhu, T.; Maxwell, M.J.; Agrotis, A.; Jerome, J.R.; Hargreaves, E.M.; Gerondakis, S.; Hibbs, M.L.; Mackay, F.; et al. Foxp3+ Tregs Are Recruited to the Retina to Repair Pathological Angiogenesis. Nat. Commun. 2017, 8, 748. [Google Scholar] [CrossRef]
  170. Muffat, J.; Li, Y.; Yuan, B.; Mitalipova, M.; Omer, A.; Corcoran, S.; Bakiasi, G.; Tsai, L.H.; Aubourg, P.; Ransohoff, R.M.; et al. Efficient Derivation of Microglia-like Cells from Human Pluripotent Stem Cells. Nat. Med. 2016, 22, 1358–1367. [Google Scholar] [CrossRef]
  171. Wang, Z.; Liu, A.; Zhang, H.; Wang, M.; Tang, Q.; Huang, Y.; Wang, L. Inhibition of Retinal Neovascularization by VEGF SiRNA Delivered via Bioreducible Lipid-like Nanoparticles. Graefes Arch. Clin. Exp. Ophthalmol. 2020, 258, 2407–2418. [Google Scholar] [CrossRef]
  172. Singerman, L. Combination Therapy Using the Small Interfering RNA Bevasiranib. Retina 2009, 29, S49–S50. [Google Scholar] [CrossRef]
  173. Jiang, Y.; Huo, S.; Hardie, J.; Liang, X.J.; Rotello, V.M. Progress and Perspective of Inorganic Nanoparticle-Based SiRNA Delivery Systems. Expert Opin. Drug Deliv. 2016, 13, 547–559. [Google Scholar] [CrossRef]
  174. Huang, K.; Lin, Z.; Ge, Y.; Chen, X.; Pan, Y.; Lv, Z.; Sun, X.; Yu, H.; Chen, J.; Yao, Q. Immunomodulation of MiRNA-223-Based Nanoplatform for Targeted Therapy in Retinopathy of Prematurity. J. Control. Release 2022, 350, 789–802. [Google Scholar] [CrossRef] [PubMed]
  175. Sung, M.S.; Moon, M.J.; Thomas, R.G.; Kim, S.Y.; Lee, J.S.; Jeong, Y.Y.; Park, I.K.; Park, S.W. Intravitreal Injection of Liposomes Loaded with a Histone Deacetylase Inhibitor Promotes Retinal Ganglion Cell Survival in a Mouse Model of Optic Nerve Crush. Int. J. Mol. Sci. 2020, 21, 9297. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, T.; Li, Y.; Guo, M.; Dong, X.; Liao, M.; Du, M.; Wang, X.; Yin, H.; Yan, H. Exosome-Mediated Delivery of the Neuroprotective Peptide PACAP38 Promotes Retinal Ganglion Cell Survival and Axon Regeneration in Rats With Traumatic Optic Neuropathy. Front. Cell Dev. Biol. 2021, 9, 659783. [Google Scholar] [CrossRef] [PubMed]
  177. Mezu-Ndubuisi, O.J.; Wang, Y.; Schoephoerster, J.; Falero-Perez, J.; Zaitoun, I.S.; Sheibani, N.; Gong, S. Intravitreal Delivery of VEGF-A165-Loaded PLGA Microparticles Reduces Retinal Vaso-Obliteration in an In Vivo Mouse Model of Retinopathy of Prematurity. Curr. Eye Res. 2019, 44, 275–286. [Google Scholar] [CrossRef] [PubMed]
  178. Hong, E.H.; Shin, Y.U.; Cho, H. Retinopathy of Prematurity: A Review of Epidemiology and Current Treatment Strategies. Clin. Exp. Pediatr. 2022, 65, 115–126. [Google Scholar] [CrossRef]
  179. Desjarlais, M.; Rivera, J.C.; Lahaie, I.; Cagnone, G.; Wirt, M.; Omri, S.; Chemtob, S. MicroRNA Expression Profile in Retina and Choroid in Oxygen-Induced Retinopathy Model. PLoS ONE 2019, 14, e0218282. [Google Scholar] [CrossRef]
  180. Lee, S.S.; Hughes, P.; Ross, A.D.; Robinson, M.R. Biodegradable Implants for Sustained Drug Release in the Eye. Pharm. Res. 2010, 27, 2043–2053. [Google Scholar] [CrossRef]
  181. Castro-Navarro, V.; Cervera-Taulet, E.; Navarro-Palop, C.; Monferrer-Adsuara, C.; Hernández-Bel, L.; Montero-Hernández, J. Intravitreal Dexamethasone Implant Ozurdex® in Naïve and Refractory Patients with Different Subtypes of Diabetic Macular Edema. BMC Ophthalmol. 2019, 19, 15. [Google Scholar] [CrossRef]
  182. Kuno, N.; Fujii, S. Biodegradable Intraocular Therapies for Retinal Disorders: Progress to Date. Drugs Aging 2010, 27, 117–134. [Google Scholar] [CrossRef]
  183. 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]
  184. Wong, J.G.; Chang, A.; Guymer, R.H.; Wickremasinghe, S.; Reilly, E.; Bell, N.; Vantipalli, S.; Moshfeghi, A.A.; Goldstein, M.H. Phase 1 Study of an Intravitreal Axitinib Hydrogel-Based Implant for the Treatment of Neovascular Age-Related Macular Degeneration (NAMD). Investig. Ophthalmol. Vis. Sci. 2021, 62, 218. [Google Scholar]
  185. 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] [PubMed]
  186. Vinores, S.A. Pegaptanib in the Treatment of Wet, Age-Related Macular Degeneration. Int. J. Nanomed. 2006, 1, 263–268. [Google Scholar]
  187. Yang, M.; Peterson, W.M.; Yu, Y.; Kays, J.; Cardona, D.; Culp, D.; Gilger, B.C.; Cleland, J. GB-102 for Wet AMD: A Novel Injectable Formulation That Safely Delivers Active Levels of Sunitinib to the Retina and RPE/Choroid for Over Four Months. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5037. [Google Scholar]
  188. Cheng, Y.; Burda, C. 2.01—Nanoparticles for Photodynamic Therapy. In Comprehensive Nanoscience and Technology; Andrews, D.L., Scholes, G.D., Wiederrecht, G.P., Eds.; Academic Press: Amsterdam, The Netherlands, 2011; pp. 1–28. ISBN 978-0-12-374396-1. [Google Scholar]
  189. Rodrigues, G.A.; Lutz, D.; Shen, J.; Yuan, X.; Shen, H.; Cunningham, J.; Rivers, H.M. Topical Drug Delivery to the Posterior Segment of the Eye: Addressing the Challenge of Preclinical to Clinical Translation. Pharm. Res. 2018, 35, 245. [Google Scholar] [CrossRef] [PubMed]
  190. Doukas, J.; Mahesh, S.; Umeda, N.; Kachi, S.; Akiyama, H.; Yokoi, K.; Cao, J.; Chen, Z.; Dellamary, L.; Tam, B.; et al. Topical Administration of a Multi-Targeted Kinase Inhibitor Suppresses Choroidal Neovascularization and Retinal Edema. J. Cell. Physiol. 2008, 216, 29–37. [Google Scholar] [CrossRef]
  191. Yafai, Y.; Yang, X.M.; Niemeyer, M.; Nishiwaki, A.; Lange, J.; Wiedemann, P.; King, A.G.; Yasukawa, T.; Eichler, W. Anti-Angiogenic Effects of the Receptor Tyrosine Kinase Inhibitor, Pazopanib, on Choroidal Neovascularization in Rats. Eur. J. Pharmacol. 2011, 666, 12–18. [Google Scholar] [CrossRef]
  192. Adams, C.M.; Anderson, K.; Artman, G.; Bizec, J.C.; Cepeda, R.; Elliott, J.; Fassbender, E.; Ghosh, M.; Hanks, S.; Hardegger, L.A.; et al. The Discovery of N-(1-Methyl-5-(Trifluoromethyl)-1H-Pyrazol-3-Yl)-5-((6- ((Methylamino)Methyl)Pyrimidin-4-Yl)Oxy)-1H-Indole-1-Carboxamide (Acrizanib), a VEGFR-2 Inhibitor Specifically Designed for Topical Ocular Delivery, as a Therapy for Neovascular Age-Related Macular Degeneration. J. Med. Chem. 2018, 61, 1622–1635. [Google Scholar] [CrossRef] [PubMed]
  193. Zernii, E.Y.; Baksheeva, V.E.; Iomdina, E.N.; Averina, O.A.; Permyakov, S.E.; Philippov, P.P.; Zamyatnin, A.A.; Senin, I.I. Rabbit Models of Ocular Diseases: New Relevance for Classical Approaches. CNS Neurol. Disord. Drug Targets 2016, 15, 267–291. [Google Scholar] [CrossRef]
  194. Owen, G.R.; Brooks, A.C.; James, O.; Robertson, S.M. A Novel in Vivo Rabbit Model That Mimics Human Dosing to Determine the Distribution of Antibiotics in Ocular Tissues. J. Ocul. Pharmacol. Ther. 2007, 23, 335–342. [Google Scholar] [CrossRef]
  195. Shen, J.; Durairaj, C.; Lin, T.; Liu, Y.; Burke, J. Ocular Pharmacokinetics of Intravitreally Administered Brimonidine and Dexamethasone in Animal Models with and without Blood-Retinal Barrier Breakdown. Investig. Ophthalmol. Vis. Sci. 2014, 55, 1056–1066. [Google Scholar] [CrossRef]
  196. Kang-Mieler, J.J.; Rudeen, K.M.; Liu, W.; Mieler, W.F. Advances in Ocular Drug Delivery Systems. Eye 2020, 34, 1371–1379. [Google Scholar] [CrossRef] [PubMed]
  197. Paunovska, K.; Loughrey, D.; Dahlman, J.E. Drug Delivery Systems for RNA Therapeutics. Nat. Rev. Genet. 2022, 23, 265–280. [Google Scholar] [CrossRef] [PubMed]
  198. Jahangirian, H.; Lemraski, E.G.; Webster, T.J.; Rafiee-Moghaddam, R.; Abdollahi, Y. A Review of Drug Delivery Systems Based on Nanotechnology and Green Chemistry: Green Nanomedicine. Int. J. Nanomed. 2017, 12, 2957–2978. [Google Scholar] [CrossRef] [PubMed]
  199. Sung, Y.K.; Kim, S.W. Recent Advances in Polymeric Drug Delivery Systems. Biomater. Res. 2020, 24, 12. [Google Scholar] [CrossRef] [PubMed]
More
Video Production Service