Next Article in Journal
The Influence of Aminoalcohols on ZnO Films’ Structure
Next Article in Special Issue
rhEGF-Loaded Hydrogel in the Treatment of Chronic Wounds in Patients with Diabetes: Clinical Cases
Previous Article in Journal
Central Composite Design (CCD) for the Optimisation of Ethosomal Gel Formulation of Punica granatum Extract: In Vitro and In Vivo Evaluations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glaucoma Treatment and Hydrogel: Current Insights and State of the Art

Department of Ophthalmology, University of Turin, Via Cherasco 23, 10126 Turin, Italy
*
Author to whom correspondence should be addressed.
Gels 2022, 8(8), 510; https://doi.org/10.3390/gels8080510
Submission received: 14 July 2022 / Revised: 9 August 2022 / Accepted: 13 August 2022 / Published: 17 August 2022
(This article belongs to the Special Issue Advanced Hydrogels for Regenerative Medicine and Tissue Engineering)

Abstract

:
Aqueous gels formulated using hydrophilic polymers (hydrogels) and those based on stimuli-responsive polymers (in situ gelling or gel-forming systems) attract increasing interest in the treatment of several eye diseases. Their chemical structure enables them to incorporate various ophthalmic medications, achieving their optimal therapeutic doses and providing more clinically relevant time courses (weeks or months as opposed to hours and days), which will inevitably reduce dose frequency, thereby improving patient compliance and clinical outcomes. Due to its chronic course, the treatment of glaucoma may benefit from applying gel technologies as drug-delivering systems and as antifibrotic treatment during and after surgery. Therefore, our purpose is to review current applications of ophthalmic gelling systems with particular emphasis on glaucoma.

1. Introduction

A progressive loss of the visual field and eventually blindness characterize glaucoma, an optic neuropathy caused by the destruction of ganglion cells [1]. Glaucoma is the second most common cause of blindness worldwide, with a growing number of people affected [2,3]. Glaucoma can dramatically compromise individuals’ habits and quality of life (QoL), determining a progressive and irreversible visual impairment and, potentially, blindness [4].
Intraocular pressure (IOP) reduction is currently the mainstay of glaucoma therapy, and it may be obtained with drugs, laser, or surgical therapy [5,6,7]. The first-line treatment is presently based on topical drug treatment.
Multiple observational studies and randomized clinical trials have confirmed that antiglaucoma medications effectively lower IOP at various disease stages [7,8,9,10,11]. Beta-blockers, topical carbonic anhydrase inhibitors, cholinergics, prostaglandins/prostamides, alpha2-agonists, and Rho-kinase inhibitors are only a few medication families used alone or in combination to treat glaucoma. Though medical therapy is a crucial asset for patients and ophthalmologists, it can have significant drawbacks. For example, adverse effects (ocular inflammation, itching, and visual disturbances) reduce adherence, thereby diminishing the therapeutic efficacy. In addition, it is challenging for patients and caregivers to administer eye drops; administration is frequently suboptimal. For these reasons, the adherence to glaucoma medical therapy is often poor, with an early interruption of the treatment [12,13]. Moreover, topical treatments may harm conjunctival health, reducing the surgery’s effectiveness [12,13].
Furthermore, the eyes have several metabolic, static, and dynamic barriers that increase the difficulty of topical drug administration. The conjunctiva, sclera, and cornea are the static obstructions that medications must get through to work pharmacologically on the eye. These barriers are challenging because of their various characteristics (polarity, hydrophilicity, surface charge, etc.).
On the contrary, dynamic barriers are the rapid clearance of the drops from the eye through nasolacrimal drainage and conjunctival vessels. Moreover, metabolic enzymes of the ocular surface can deactivate drugs [14]. The sum of these mechanisms drastically reduces the pharmaceuticals’ absorption due to the lower residence time on the ocular surface. Reduced effectiveness of the treatment leads to frequent changes in the therapy and, thus, to poorer patient compliance.
Conventional ophthalmic dosages, formulated as eye drops (solutions or suspensions), or ointments, are preferable to distribute medications to the ocular surface and anterior eye segment. Their comparatively low production costs, non-invasiveness, and ease of use offer clear benefits. [15,16].
Their side effects and low bioavailability may reduce optimal drug concentration at the target site (Figure 1). To avoid the failure of the medical treatment, several researchers have developed various drug delivery systems to enhance the residence time on the ocular surface [17,18,19]. One of the most frequently adopted strategies is medical gels [20].
Another exciting field of gel application is to prevent scarring after filtering glaucoma surgery.
This review will focus on drug delivery and adjunctives in glaucoma surgery.

Polymers and Hydrogel in Ocular Drugs Delivery

Hydrogels are a network of biologic and synthetic hydrophilic polymers able to absorb aqueous fluids. The extensive array of customizability and the high hydrophilicity render hydrogel the ideal compound for ophthalmic applications, ranging from vitreous substitutes to drug delivery [21]. Several authors proposed hydrogel as a drug carrier, incorporated with other polymers, to treat glaucoma. Among these molecules, synthetic polymeric biomaterials are a group of compounds based on chemically-derived monomers and represent one of the most reported options for delivering ocular pharmaceuticals [21]. To this class belong polyethylene oxides, polyvinyl alcohol (PVA), polyesters, polymethacrylates, polyolefins, and dendrimers [22].
On the contrary, biopolymers are based on naturally derived monomers and are characterized by high biocompatibility and fast degradation [23]. The most frequently reported biological polymers used for ocular drug delivery are chitosan, cellulose, hyaluronic acid, carboxymethyl cellulose, and gelatin. Another promising field in ocular delivery systems is micro- and nanotechnology, such as formulations of hydrogel combined with micro- and nanoparticles, liposomes, and micelles [22]. This drug delivery strategy combines the hydrogel’s high water content with the small size (from 1 to 1000 nm) of these particles, offering a more targeted delivery and a sustained release of the medications [23].
Among the medical gels, the most investigated are the gel-forming systems. Their properties make them able to undergo phase transition after environmental stimulus following their application in the conjunctival cul-de-sac and thus, transform themselves into a viscoelastic gel from the original liquid dosage [24] because of pH change, temperature modulation, or ion triggers [25,26,27,28,29,30]. Because they do not need organic solvents or copolymerization catalysts to cause gel formation, in situ polymeric gelling systems have drawn significant attention.

2. Drug Delivery

The use of topical medications represents the primary approach to treating glaucoma. Several therapeutic classes, α-2 agonists, β-blockers, carbonic anhydrase inhibitors, prostaglandin analogous, and cholinergic agents, were developed in the form of eye drops for IOP regulation [31].
A correct administration of eye drops and proper therapy can prevent an IOP elevation and, thus, the progression of the glaucoma-induced damages.
Despite that, as stated above, self-administration of topical therapy can be challenging for an elderly population. Moreover, ophthalmic solutions are often related to local side effects such as ocular burning and stinging [32,33,34,35]. These conditions may lead to poorer therapy compliance and, thus, can cause a worse treatment outcome [36,37]. Therefore, improving patient adherence and compliance while facilitating the use of glaucoma drugs is a top public health priority [38,39].
Furthermore, drugs’ efficacy may be decreased due to their rapid clearance (low residence time), which results in a lower bioavailability [17,40]. For these reasons, a proper delivery system may help improve medications’ effectiveness, safety, and duration.
Then, several electronic devices, transdermal and ocular inserts, and mechanical drug delivery systems were developed [41].

2.1. Contact Lens

Due to their location near the cornea, contact lenses (CL) have some unique advantages for delivering drugs [42]. First of all, the limitation opposed by the CL to the tear film action results in a drug residence time of more than 30 min [43,44] compared to 5 min only for eye drops alone [45]. The enhanced residence time leads to significant increases in bioavailability, possibly as large as 50% [46].
Notably, hydrogels are the main constituents of CL; thus, their high-water content and favorable properties render them highly compatible with human tissues [47,48,49]. Indeed, even if the water content of the lens reaches 99%, the oxygen permeability will not exceed the theoretical value of about 40 DK/t, which affects the user’s comfort during a prolonged period [50].
For these reasons, CL can be used to treat ocular diseases such as glaucoma [51] due to their ability to extend the release of a preloaded drug to several days or even months [52].
These drugs can be loaded into lenses by soaking [53,54], molecular imprinting [55], microemulsion, or through nanoparticles [56,57].
Current methods can produce lenses of suitable thickness, water content, and optical properties [58,59]. Moreover, it was shown that the application time did not affect ocular tissues, and no ocular adverse effects were observed in any case [60].
Furthermore, therapeutic CL can reduce drug doses and thus side effects [61] and can be particularly beneficial for applications in elderly patients having trouble adhering to repeated dosage regimens [62,63]. CL can be made of hydroxyethyl methacrylate (HEMA), Methacrylic acid (MAA) [55], poly(vinyl alcohol) (PVA) [64], and N-vinyl-2-pyrrolidone (NVP), Poly(2-hydroxyethyl methacrylate) (pHEMA) [65]. Moreover, some authors, such as Gulsen et al. [66,67,68] and Kapoor et al. [69,70], have proposed CL incorporated with nanoparticles, such as liposomes, micelles, and microemulsions [71,72,73,74,75,76,77,78,79]. Jung et al. focused on soaking commercial lenses in a solution of timolol-Propoxylated Glyceryl Triacylate (PGT) nanoparticles or timolol- Ethylene Glycol Dimethacrylate (EGDMA) nanoparticles. Drug release studies in a diffusion cell showed an extended release for about 2–4 weeks [80].
Maulvi et al., in 2016, formulated a nanoparticle-loaded ring implant placed between partially polymerized hydrogel contact lenses to provide an extended release of Timolol Maleate (TM) at therapeutic rates without affecting the optical or physical properties of CL. In vivo studies showed a tear fluid release for more than 192 h [81] (Figure 2).
Subsequently, Maulvi et al., in 2021, proposed to use graphene oxide (GO) loaded into silicone hydrogel CL through a polymerization process to improve the Bimatoprost release. GO improved the swelling properties of the lenses, the transmittance, and the bioavailability [82].
Ciolino et al. created Latanoprost-eluting contact lenses by encapsulating latanoprost-PLGA (poly[lactic-co-glycolic acid]) between two layers of pHEMA hydrogel by ultraviolet light polymerization. In vitro and in vivo studies reported an extended release of up to one month [83].
Sekar et al. showed the ability of the vitamin-E-integrated polymeric hydrogel to prolong prostaglandins (Bimatoprost) release for more than ten days [84], while increasing stability and limiting diffusion during storage [85,86]. Indeed, vitamin-E incorporation into the hydrogel matrix has been reported to form nano-sized barriers shaped as ellipsoids, retarding drug diffusion through the polymer matrix [84].
Nicolson et al. found out that hydrogel lenses based on hydrophilic monomers such as 2- HEMA and NVP did not succeed in absorbing the minimum oxygen requirement for eyes under closed eyelid conditions [87].
Silicone hydrogel CL offers advantages over other types of lenses in drug delivery because of their oxygen transmissibility and their potential for hydrophobic drug delivery [88].
Nguyen-Phuong-Dung et al. showed that higher content of hydrophilic polymers increased water uptake ability and improved hydrophilicity of silicone hydrogel lenses. However, the oxygen permeability is linked with the quantity of polydimethylsiloxane (PDMS): the permeability decreases with the decrease of PDMS content. In addition, these silicone hydrogel lenses exhibited relatively good optical transparency, anti-protein deposition and appeared non-cytotoxic [89].
Chen et al. and Jones et al. explained that PDMS exhibits a lower water uptake ability, poor wettability, and high lipid adsorption despite having unique high oxygen transmissibility and superior resistance to tearing for contact lens application [87,90].
One advantage of the hydrophobic drug released from CL is that it could more readily diffuse through corneal cells’ tight lipid junctions and cell membranes due to its hydrophobic character.
Furthermore, different studies have found that drugs with hydrophobic character (ex. Latanoprost) can be loaded quickly (in 4 min) into Hydrogel contact lenses using non-aqueous solvents [91].
One of the most critical problems of drug delivery through CL remains the difficulty of incorporating more than one drug. A high degree of cross-link hydrogel is needed in this case, but this process might alter physical properties or oxygen permeability [92,93,94,95] (Table 1).

2.2. Hydrogel as an Ocular Drug Delivery System for Glaucoma Treatment

As stated above, the essential properties of hydrogels are their ability to absorb water due to their hydrophilicity and excellent biocompatibility, making them ideal for drug delivery applications [96]. Several authors proposed hydrogel as a drug carrier incorporated with other polymers such as chitosan, cellulose derivatives, gelatin, PVA, or poly lactic-co-glycolic acid for the treatment of glaucoma [96,97,98,99]. Other polymer-based hydrogels evaluated for ophthalmic administration are stimulus-responsive gel [100,101,102,103,104]. These delivery strategies have been investigated for several ophthalmic drugs commonly used in the treatment of glaucoma, such as TM, brimonidine tartrate (BT), pilocarpine, and latanoprost, or combinations with each other [105,106,107,108] (Table 2).

2.2.1. Pilocarpine

Pilocarpine is a muscarinic acetylcholine M3 agonist, causing miosis and increasing aqueous humor outflow [109]. In 2007, Natu et al. developed, as drug carriers, hydrogels loaded with pilocarpine hydrochloride by soaking in an aqueous solution containing the drug [110]. In vitro evaluation showed a percentage of released drugs that varied between 29.2% and 99.2% in 8 h [110]. Subsequently, Chou et al. developed pilocarpine-loaded antioxidant-functionalized biodegradable thermogels for intracameral administration of antiglaucoma medications [107]. The antioxidant function is due to the chemical grafting of antioxidant gallic acid onto biodegradable gelatin [107]. In vivo study on a rabbit model showed a mean IOP reduction of 5 mmHg for 28 days [107].
Lai and co-workers proposed a new biodegradable in situ gelling delivery system for the intracameral administration of the pilocarpine [111]. The preparation of the copolymeric carriers was achieved from gelatin-g-poly(N-isopropyl-acrylamide). All prepared copolymeric carriers exhibit a relevant intraocular pressure-lowering and an excellent miotic effect [111]. Afterward, Nguyen and colleagues prepared injectable biodegradable thermogels coloaded with pilocarpine and ascorbic acid [112]. The synthesized gel obtained the double effect of lowering elevated IOP and reducing stromal collagen degradation in inflammation-induced glaucoma. In vitro results indicated a sustained release for 80 days [112]. In 2020, Luo et al. reported a new biodegradable and injectable thermogel, coloaded with pilocarpine and RGFP966 to exert antioxidant activities and sustained drug delivery for treating glaucomatous nerve damage [113]. RGFP966 is an inhibitor of histone deacetylases and, consequently, plays a critical role in regulating retinal ganglion cell atrophy and is becoming a primary target for treating neurodegeneration [113]. Their results indicate a long-active release function of both drugs [113].

2.2.2. Timolol Maleate

TM is a small hydrophilic molecule (432 Da) regarded as the “gold standard” treatment for glaucoma [114,115]. Indeed, the intraocular pressure (IOP)-lowering potential of this β-receptor antagonist has been reported to be between 20 and 25% of the initial values [116]. Incorporating viscosifying agents can avoid some limitations of topical administration, such as extensive drug loss due to the turnover of lacrimal drainage. Several authors proposed hydrogel-based formulation for TM application [117]. In 2011 Zhang et al. investigated a novel TM liposomal-hydrogel to improve drug permeability and extend residence time in the precorneal region [118]. In an in vivo study on 12 rabbits, it lowered IOP for six h before rising to its initial value [118]. Later, in 2012 Holden et al. proposed a polyamidoamine dendrimer hydrogel linked with polyethylene glycol (PEG)-acrylate chains for BT and TM delivery [119]. This dendrimer hydrogel increased the solubility of brimonidine and sustained the in vitro release of both drugs over 56–72 h [119].
Yang et al. developed a hybrid dendrimer hydrogel/poly(lactic-co-glycolic acid) nanoparticle platform to efficiently deliver BT and TM to the eye and gradually release the drug [103]. IOP was tested in albino rabbits; its maximum decrease was 29.5% less than the initial values, and its duration was four days. Subsequently, Kulkarni et al. [120] proposed a controlled-release ocular film of TM using a natural hydrogel derived from the seeds of Tamarindus indica. In three rabbits, they obtained a controlled drug release for 24 h in vivo. A self-assembling elastin-like hydrogel was experimented by Fernańdez-Colino and co-workers. In vivo testing revealed a hypotensive effect lasting more than an eighth [114]. In 2017 Karavasili et al. tested self-assembling peptides Ac-(RADA)4-CONH2 and Ac-(IEIK)3I-CONH2, which form hydrogels in physiological conditions, as carriers for ocular delivery of TM [121]. Ac-(RADA)-CONH was demonstrated to significantly enhance the bioavailability of TM, achieving effective IOP reduction for up to 24 h [121].
Dubey and colleagues prepared a stimuli-sensitive hydrogel with Carbopol (poly(acrylic acid)) [122]. TM and BT were used in this formulation, propyl methylcellulose as a viscosity enhancer, and ethylenediaminetetraacetic acid (EDTA) as a chelating agent. This delivery system achieved a 100% cumulative release of the drugs in 8 h and a higher IOP reduction efficiency in the in vivo studies [122]. In the same way, the use of carbopol was reported by Singh et al. along with hydroxyethyl cellulose for TM delivery [123]. The in vitro results showed a continuous release of the drug over eight hours and a gradual decrease of IOP if compared to marketed eye drops [123]. Subsequently, El-Feky et al. developed an oxidized sucrose crosslinker used in the formulation of chitosan-gelatin hydrogel for the sustained release of TM to control ocular hypertension [108]. They obtained a hypotensive effect that lasted more than eight hours [108].
Pakzad et al. synthesized a chitosan with Glycidyltrimethylammonium chloride (GTMAC) to prepare N-(2-hydroxy-3-trimethylammonium) propyl chitosan chloride (HTCC) [124]. HTCC increased mucoadhesive capacity, solubility in water, and antibacterial properties at physiological pH. A hydrogel was prepared with HTCC and glycerolphosphate. The studies showed an extended-release of TM for up to 80 h [124]. An in situ gel forming self-assembling peptide, ac-(RADA)4-CONH2, was evaluated as a carrier for the ocular co-delivery of TM and BT by Taka et al. [105]. A complete release of both drugs was obtained within eight hours. A 5.4-fold and 2.8-fold higher corneal permeability was achieved for BT and TM, respectively [105]. Patel et al. synthesized a combined pH and ion-sensitive in situ hydrogel using Carbopol and gellan gum, which compared with TM’s marketed eye drop formulation [104]. Furthermore, they evaluated the efficacy of benzododecenium bromide as a preservative and a corneal permeability enhancer [104]. The novel formulations reported better corneal permeability and fewer side effects than the marketed formulation [104].
In 2021, Wang et al. tested the delivery efficacy of BT and TM loaded into dendrimer gel particles of various sizes: nano-in-nano dendrimer hydrogel particles of 200 nm (nDHP) and two micronized DHPs—μDHP3 (3 μm), and μDHP10 of (9 μm) [125]. They evaluated nDHP superiority in cytocompatibility, corneal permeability, degradability, and drug release kinetics. Indeed, μDHP10 and μDHP3 induced a cytotoxicity 2.8-fold higher than nDHP, while they had a faster degradation time. Moreover, nDHP enabled 4.1 μg of BT to permeate through the cornea, while μDHP10 and μDHP3 were able to permeate, respectively, 2.4 μg and 3.5 μg. Finally, nDHP showed a longer release time [125]. The mean IOP lowering after treatment was 18.68 ± 1.35 mmHg [125]. Another research proposed a new multilayered drug delivery hydrogel inspired by a lollipop structure. The final polymer was a multilayered sodium alginate-chitosan hydrogel ball decorated by zinc oxide-modified biochar, encapsulating TM and levofloxacin inside the different layers. The results showed that in vitro release of TM can be sustained for longer than two weeks [126].

2.2.3. Brimonidine Tartrate

Brimonidine tartrate exerts its effects in the eye due to its high a2- adrenoceptor affinity, which is considered a standard reference compound for the treatment of glaucoma [127]. As stated above, several papers reported hydrogel-based delivery strategies evaluated for the combination of BT and TM [99,105,119,122,126]. In 2017 Fedorchak et al. proposed an innovative drug delivery system composed of a thermo-responsive hydrogel carrier and BT-loaded poly(lactic-co-glycolic) acid microspheres [128]. Their results suggest in vivo efficacy for over 28 days from a single drop instillation [128]. Another paper reported a mildly cross-linked dendrimer hydrogel synthesized through the addition of a polyamidoamine (PAMAM) dendrimer and polyethylene glycol diacrylate (PEG-DA) [129], resulting in a 48 h drug release and an enhanced corneal permeation [129]. Subsequently, Wang et al. developed a branched polyrotaxane hydrogel made of 4-arm polyethylene glycol (4-PEG) and a-cyclodextrin (a-CD) [130]. BT was loaded on the resulting a-CD/4-PEG hydrogel, which underwent a reversible gel-sol transition in response to shear stress change [130]. A controlled release of 24 h was obtained. In 2019 Bellotti and co-workers reported a pNIPAAm-based thermoresponsive hydrogel for BT delivery [131]. They manipulated gelation kinetics by modifying the poly(ethylene glycol) content, thus obtaining a suitable viscosity for administration as an eye drop and resisting various climatic conditions without being eliminated [131].

2.2.4. Latanoprost

Latanoprost is an ester prodrug analog of prostaglandin and is the first-line treatment in patients suffering from glaucoma because it reduces IOP by increasing uveoscleral outflow [132]. However, its daily administration is correlated with local side effects such as conjunctival hyperemia and dry eye syndrome [97]. Several polymers have been developed as drug carriers to prolong Latanoprost permanence on the ocular surface and minimize the possibility of therapy adherence failure.
In 2014, Cheng and co-workers proposed an injectable thermosensitive chitosan/gelatin/glycerol phosphate (C/G/GP) hydrogel as a sustained-release system of latanoprost for glaucoma treatment [98]. In vivo evaluation, performed on ten albino rabbits, showed a decrease of IOP of 9.2% within eight days and then a permanence within normal limits for the next 31 days [98]. Hsiao et al. prepared a chitosan-based thermogel to improve glaucoma therapy [106]. In vivo examination, conducted with subconjunctival injections in six rabbits, revealed excellent biocompatibility and a lowering of IOP for 40 days [106].
In the same way, Cheng et al. formulated a thermosensitive chitosan/gelatin for the sustained release of latanoprost as a topical eye drop to control ocular hypertension [133]. In vivo results of a rabbit model confirmed IOP was significantly decreased within seven days [133]. In 2019, another thermosensitive hydrogel containing latanoprost and curcumin-loaded nanoparticles was developed [97]. Indeed, curcumin possesses antioxidant and anti-inflammation properties and reduces oxidative stress on trabecular meshwork [97]. In vitro drug release evaluation revealed that both latanoprost and curcumin-loaded nanoparticles maintained a sustained release for seven days [97].

2.2.5. Other Drugs

Epinephrine was applied in glaucoma treatment to reduce intraocular pressure by decreasing aqueous formation and increasing the outflow facility [134]. Hsiue et al. developed two preparations of ophthalmic drops for controlled release based on the thermosensitivity of poly-N-isopropyl acrylamide (PNIPAAm) [135]. The first formulation contained a linear chain of PNIPAAm, and the second had a mixture of linear PNIPAAm and cross-linked PNIPAAm nanoparticles. After in vivo examination of the rabbits, the authors reported that the formulation containing a linear PNIPAAm and nanoparticles maintained a more prolonged IOP decrease (32 h) but showed a weaker effect. In contrast, the linear PNIPAAm, had a shorter duration but a more substantial IOP decrease (−7.2 mmHg) [135]. In the same way, Prasannan and colleagues structured a thermosensitive PNIPAAm-based hydrogel loaded with epinephrine [136].
Atenolol is a β1 adrenoceptor blocker for the treatment of glaucoma. A niosomal hydrogel containing atenolol was proposed by Abuhashim et al. in 2014 [137]. Their formulation was an eye drop solution that showed promising results after in vivo examination as it significantly decreased the IOP and showed a prolonged effect up to 8 h [137].
Table 2. Studies on drug delivery hydrogel.
Table 2. Studies on drug delivery hydrogel.
AuthorYearDrugGroupPolymer NameIOP Decrease (Mean Value)DurationAdministration
Bellotti et al. [131]2019Brimonidine tartrateα2 agonistpNIPAAm hydrogels (PEG)/ Eye drop
Fedorchak et al. [128]2017Brimonidine tartrateα2 agonistPoly(lactic-co-glycolic) acid microspheres microspheres
incorporated into the pNIPAAM gel
/28 daysEye drop
Wang et al. [129]2017Brimonidine tartrateα2 agonistLinked dendrimer hydrogel via addition of polyamidoamine
(PAMAM) dendrimer G5 and polyethylene glycol
diacrylate (PEG)
/48 hAC filling
Wang et al. [130]2018Brimonidine tartrateα2 agonist(a-CD/4-PEG hydrogels) hydrogel made of 4-arm polyethylene
glycol (4-PEG) and a-cyclodextrin (a-CD)
/24 h/
Dubey et al. [122]2014Timolol maleate- brimonidine tartrateβ-blockers-
α2 agonist
Stimuli-sensitive hydrogel with Carbopol (poly(acrylic acid)14 mmHg (mean IOP after treatment)8 hEye drop
Holden et al. [119]2012Brimonidine-timolol maleateα2 agonist-
β-blockers
Polyamidoamine dendrimer hydrogel linked with
polyethylene glycol (PEG)-acrylate chains
/6–72 h/
Taka et al. [105]2020Timolol maleate-brimonidine tartrateβ-blockers-
α2 agonist
Self-assembling peptide ac-(RADA)4-CONH2/8 h/
Wang et al. [125]2021Brimonidine tartrate-and timolol maleateα2 agonist
β-blockers
Nano-in-nano dendrimer hydrogel particles −200 nm (nDHP)18.68 ± 1.35 mmHg (mean
IOP after treatment)
/Eye suspension
Yang et al. [99]2013Brimonidine-timolol maleateα2 agonist-
β-blockers
Hybrid dendrimer hydrogel/poly(lactic-co-glycolic acid)
nanoparticle platform
29.5%4 days/
Cheng et al. [133]2016LatanoprostProstaglandinThermosensitive chitosan/gelatin/7 daysEye drop
Cheng et al. [97]2019LatanoprostProstaglandinThermosensitive hydrogel containing latanoprost and curcumin-loaded nanoparticles/7 daysEye drop
Cheng et al. [98]2014LatanoprostProstaglandinThermosensitive chitosan/gelatin/glycerol phosphate (C/G/GP) hydrogel2.4 mmHg (9.2%)31 daysSubconjunctival injection
Hsiao et al. [106]2014LatanoprostProstaglandinAmphiphilic chitosan-based thermogelling10 mmHg39 daysSubconjunctival injection
Abu Hashim et al. [137]20140.5% atenololβ1 adrenoceptor blockerNiosomal Hydrogel containing atenolol/8 hEye drop
Hsiue et al. [135]2002epinephrineCatecholamineThermosensitive poly-N-isopropylacrylamide (PNIPAAm)8.9 mmHg (maximum)24 hEye drop
Prasannan et al. [136]2014epinephrineCatecholaminePAAc-g-PNIPAAm (PNIPAAm)//Eye drop
Chou et al. [107]2017pilocarpineCholinergicPilocarpine-loaded gallic acid (GA)-grafted gelatin-g-poly(N-isopropylacrylamide) (GN)5 mm Hg28 days/
Lai et al. [111]2013pilocarpineCholinergicCarboxyl- terminated PNIPAAm/12 h/
Luo et al. [113]2020pilocarpine and RGFP966Cholinergic4-hydroxy-3,5-dimethoxybenzoic acid (p-DMB)-modified
chitosan-g-poly(N-isopropylacrylamide)
/70 days/
Natu et al. [110]2007Pilocarpine hydrochlorideCholinergicLinking reaction of gelatin in N,N-(3dimethylaminopropyl)-N′-ethyl carbodiimide and
N-hydroxy succinimide
/8 h/
Nguyen et al. [112]2019Pilocarpine and ascorbic acidCholinergicPAMAM dendrimers bearing amine surface groups (-NH2)
linked with gelatin hydrogel and poly(N-isopropyl acrylamide)
/84 days/
El-Feky et al. [108]2018Timolol Maleateβ-blockersChitosan-gelatin hydrogel linked with oxidized sucrose/8 hEye drop
Esteban-Pérez et al. [117]2020Timolol maleateβ-blockersGelatin nanoparticles in a hydroxypropyl
methylcellulose viscous solution
4.33 ± 0.308 hEye drop
Fernandez-Colino et al. [114]2017Timolol maleateβ-blockersSelf-assembling elastin-like (EL)
and silk-elastin-like hydrogels
/8 hEye drop
Karavasili et al. [121]2017Timolol maleateβ-blockersSelf-assembling peptides Ac-(RADA)4-CONH2
and Ac-(IEIK)3I-CONH2
/24 hEye drop
Kulkarni et al. [120]2016Timolol maleateβ-blockersNatural hydrogel from Tamarindus indica/24 hEye drop
Pakzad et al. [124]2020Timolol maleateβ-blockersN-(2-hydroxy-3-trimethylammonium) propyl chitosan
chloride glycerophosphate (HTCC/GP)
/1 week/
Wang et al. [126]2021Timolol maleate (TM) and levofloxacinβ-blockersMultilayered sodium alginate-chitosan (SA-CS) hydrogel ball (HB) decorated by zinc oxide-modified biochar (ZnO-BC)
(‘lollipop inspired’)
/2 weeks/
Zhang et al. [118]2011Timolol maleateβ-blockersLiposomal-hydrogel/6 hEye drop

3. Hydrogel Formulation after Glaucoma Surgery

3.1. Anti-Scarring Hydrogel

Glaucoma filtration surgery is currently the most effective treatment for glaucoma unresponsive to medical therapy. Filtering surgery lowers IOP by forming a fistula between the anterior chamber and the subconjunctival space, favoring the drainage of aqueous humor in a filtering bleb. However, traditional trabeculectomy and filtering MIGS fail in a significant percentage of patients, up to 30–50%, mainly because of fibrosis in the subconjunctival filtering bleb [138,139].
The reparative process, especially in the first two weeks after surgery, involves the release of a great variety of proinflammatory cytokines and glycoproteins of the extracellular matrix. The latter stimulates the migration and proliferation of fibroblasts in the Tenon’s capsule and the subconjunctival space leading to the formation of a scar [140].
For this reason, glaucoma filtration surgery involves using antifibrotic drugs applied intraoperatively for a variable time (generally between 2 and 5 min). The most used drugs are mitomycin C (MMC) and 5-fluorouracil (5-FU), but cyclosporine A or antibodies against vascular endothelial growth factor (VEGF) such as Bevacizumab are also used. However, MMC and 5-FU, in particular, have a non-specific mechanism of action for which, in addition to inhibiting fibroblastic proliferation, they also induce cell death in the surrounding tissues giving rise to potential complications (postoperative hypotonia, bleb leaks, epithelial and endothelial corneal toxicity, thinning up to the rupture of the bleb with the consequent risk of endophthalmitis) [141,142,143,144]. Furthermore, a single application of these drugs may not be sufficient to limit the fibrotic and connective tissue reaction that develops even in the long term after the surgery [145].
Therefore, in recent years delivery systems have been studied to allow a controlled and prolonged release of drugs over time to limit the formation of postoperative scar, keep the IOP low, and guarantee the persistence of the filtering bleb while reducing toxicity. As stated above, nanoparticles-based hydrogels and stimuli-responsive hydrogels, for their natural properties, are the best candidates to play the role of drug carrier [146,147]. Their simplicity of synthesis has the advantage of precisely controlling the concentration of the incorporated drug, which is mixed in the initial aqueous solution, and of not denaturing peptides or bioactive proteins within them [148,149]. The porous structure of hydrogels allows them to transport many therapeutic molecules, both hydrophilic and hydrophobic, such as proteins, DNA, and RNA.
The hydrogels are degraded slowly, in a variable way, between two and three weeks. This characteristic makes them ideal for transporting and releasing anti-scarring drugs that can act with a stable and effective concentration precisely in the critical period of scar tissues [150].
Table 3 lists the primary studies on using anti-scarring hydrogels in glaucoma filtration surgery.
Some studies focus on the intrinsic hydrogel antifibrotic activity. In the works of Liang et al. [151] and Martin et al. [152], these biomaterials, if applied to the intervention site, form covalent bonds with the sclera and significantly reduce scar formation. Hydrogels minimize inflammation and hinder fibroblasts’ adhesion to scleral tissue, as confirmed by the reduction of connective tissue growth factor (CTGF), a peptide promoting the formation of fibrosis and scarring in ocular tissues [153]. Chen et al. [154] investigated in vitro hydrogels containing different concentrations of the arginine-glycine-aspartic acid (RGD) sequence (0.25; 0.5 and 1%). This sequence induces competition with proinflammatory proteins of the extracellular membrane for binding to integrins, cellular receptors that promote migration, and the proliferation of fibroblasts without cytotoxicity. The authors could demonstrate that 1% wt Fmoc-FFGGRGD self-assembly peptide hydrogel could inhibit the expression of β1-integrin, FAK, and Akt in Tenon’s capsule fibroblasts, which play an essential role in fibrogenesis and scar formation.
The majority of authors used hydrogels as a drug delivery system: they are implanted and provide for a localized and prolonged release of the drug they contain. Nagata et al. [155], Xi et al. [156], and Kojima et al. [157] studied MMC-loaded hydrogels. They demonstrated that hydrogel causes a prolonged and controlled release of MMC, reducing its toxicity while simultaneously reducing the formation of fibrosis and ensuring a prolonged persistence of the bleb. Yang et al. [158], Peng et al. [159], and Han et al. [150] used hydrogels to release Bevacizumab, a synthetic monoclonal antibody directed against VEGF. This drug, widely used in proliferative diabetic retinopathy, age-related macular degeneration, and neovascular glaucoma, limits the formation of fibrosis and scar after glaucoma filtration surgery and reduces IOP. VEGF is a crucial molecule in the wound healing process: it is not only an essential promoter of angiogenesis but also a direct mediator of the migration and proliferation of fibroblasts and inflammatory cells [160].
Other authors investigated the release of less commonly used drugs in glaucoma filtration surgery to reduce toxicity. Kojima et al. [161] studied the use of a chymase inhibitor. Chymase is a protease in the granules of mast cells that induces the accumulation of neutrophils, eosinophils, and other inflammatory cells and promotes cell growth of fibroblasts through the up-regulation of transforming growth factor (TGF-β) [162,163]. Using the chymase inhibitors could prevent scar formation and cause fewer complications than the current antimetabolites. Maeda et al. [164] directly associated a TGF-β antibody with the hydrogel. TGF-β is the main factor stimulating the conjunctival scar following trabeculectomy [165]. Sun et al. [166] used Cyclosporine A, while Qiao et al. [167,168] used heparin, an anticoagulant that also can limit the proliferation of fibroblasts. Chun et al. [169] used the hydrogel as a vector of small interfering RNA (siRNA). This promising therapy acts through an epigenetic silencing strategy, deactivating the genes that code for proteins promoting fibrosis and scar formation.
Another area of investigation is the administration site of the hydrogel. Most studies involve a subconjunctival injection in correspondence with the filtering bleb, and other authors suggest applying the hydrogel under the scleral flap created during the surgery [170]. A further strategy is the anterior chamber implant, as proposed by Peng et al. [159] and Han et al. [150]. The intracameral injection allows a controlled and stable release of the drug in the anterior chamber and, at the same time, in the filtering bleb through the drainage of aqueous humor determined by the surgery, without further manipulation of the conjunctiva [171].
All the studies reported favorable data regarding the reduction of fibrosis and scar formation and low toxicity. Hydrogels could therefore represent an excellent potential to increase the long-term success of these interventions.

3.2. Management of Others Post or Intraoperative Complications

One of the most frequent complications in the early postoperative period after glaucoma surgery is the leakage from the conjunctival limbal incision or the filtration blebs [172]. This leakage can lead to severe complications such as hypotony, choroidal effusion, suprachoroidal hemorrhages, loss of the anterior chamber, endophthalmitis, and bleb failure [173,174]. The leakage occurrence as a short-term complication after glaucoma surgery is becoming more frequent because of the increased application of mitomycin-C to delay wound healing [175]. Nagata et al. proposed a PEG-Based Synthetic Hydrogel as a sealant after glaucoma surgery to inhibit bleb leaking [155]. On a rabbit model, they observed fewer lymphocytic infiltrations without inflammatory effects or conjunctival toxicity [155].
Calladine et al. proposed an intraoperative implant of methacrylic hydrogel, applicable during deep sclerectomy, to maintain the intrascleral space essential for proper filtration after this surgery [176]. Their polymer showed good intrascleral biocompatibility, while no case of hypotony was reported [176].

4. Conclusions

The global cost of sight loss is estimated to be over US$3 trillion annually. Glaucoma is the second-leading cause of irreversible visual loss and is mainly treated with eye drops. Although the drug delivery formulations represent a promising alternative to conventional treatment, there are still limitations related to the performances of these ocular delivery systems. Poor adherence and/or persistence with topically applied eye drops mainly results from the need for multiple daily applications to obtain its intended therapeutic effect. New 3D-printed hydrogels, ophthalmic gels, medical devices, and nanogels designed to deliver ophthalmic gels have the potential to mitigate glaucoma-related comorbidities. Although numerous studies have demonstrated the great potential of hydrogel-based treatments, future research should continue to investigate their use in vivo and conduct clinical trials to advance the clinical application of this technology.
Furthermore, modifications of traditional surgery and the introduction of new devices to shunt aqueous humor subconjunctivally significantly reduced the early postoperative complications related to hypotony but are still facing a significant failure rate due to fibrosis of the bleb. The development of longer-releasing antifibrotic agents that tackle the different phases of the scarring process would allow for a longer-term efficacy of surgery and may potentially allow an earlier surgical approach with better control of IOP throughout the day.

Author Contributions

A.M.F., C.N., P.C. and G.B. had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: A.M.F., C.N., P.C. and G.B. Acquisition: analysis, or interpretation of data: All authors. Drafting of the manuscript: All authors. Critical revision of the manuscript for important intellectual content: All authors. Study supervision: A.M.F., P.C. and G.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Biggerstaff, K.S.; Lin, A. Glaucoma and Quality of Life. Int. Ophthalmol. Clin. 2018, 58, 11–22. [Google Scholar] [CrossRef] [PubMed]
  2. Quigley, H.A.; Broman, A.T. The number of people with glaucoma worldwide in 2010 and 2020. Br. J. Ophthalmol. 2006, 90, 262–267. [Google Scholar] [CrossRef] [PubMed]
  3. Tham, Y.-C.; Li, X.; Wong, T.Y.; Quigley, H.A.; Aung, T.; Cheng, C.-Y. Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology 2014, 121, 2081–2090. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, Y.; Alnwisi, S.; Ke, M. The impact of mild, moderate, and severe visual field loss in glaucoma on patients’ quality of life measured via the Glaucoma Quality of Life-15 Questionnaire: A meta-analysis. Medicine 2017, 96, e8019. [Google Scholar] [CrossRef]
  5. Ting, N.; Li, Y.J.; Ng, J. Different strategies and cost-effectiveness in the treatment of primary open angle glaucoma. Clin. Outcomes Res. 2014, 6, 523–530. [Google Scholar] [CrossRef]
  6. European Glaucoma Society. European Glaucoma Society Terminology and Guidelines for Glaucoma, 4th Edition—Chapter 3: Treatment principles and options Supported by the EGS Foundation: Part 1: Foreword; Introduction; Glossary; Chapter 3 Treatment principles and options. Br. J. Ophthalmol. 2017, 101, 130–195. [Google Scholar] [CrossRef]
  7. Prum, B.E.; Rosenberg, L.F.; Gedde, S.J.; Mansberger, S.L.; Stein, J.D.; Moroi, S.E.; Herndon, L.W.; Lim, M.C.; Williams, R.D. Primary Open-Angle Glaucoma Preferred Practice Pattern(®) Guidelines. Ophthalmology 2016, 123, P41–P111. [Google Scholar] [CrossRef]
  8. Kass, M.A.; Heuer, D.K.; Higginbotham, E.J.; Johnson, C.A.; Keltner, J.L.; Miller, J.P.; Parrish, R.K.; Wilson, M.R.; Gordon, M.O.; Ocular Hypertension Treatment Study Group. The Ocular Hypertension Treatment Study: A randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch. Ophthalmol. 2002, 120, 701–730. [Google Scholar] [CrossRef]
  9. The AGIS Investigators. The Advanced Glaucoma Intervention Study (AGIS): 7. The relationship between control of intraocular pressure and visual field deterioration. Am. J. Ophthalmol. 2000, 130, 429–440. [Google Scholar] [CrossRef]
  10. Collaborative Normal-Tension Glaucoma Study Group. The effectiveness of intraocular pressure reduction in the treatment of normal-tension glaucoma. Am. J. Ophthalmol. 1998, 126, 498–505. [Google Scholar] [CrossRef]
  11. Heijl, A.; Leske, M.C.; Bengtsson, B.; Hyman, L.; Bengtsson, B.; Hussein, M. Reduction of intraocular pressure and glaucoma progression: Results from the Early Manifest Glaucoma Trial. Arch. Ophthalmol. 2002, 120, 1268–1279. [Google Scholar] [CrossRef] [PubMed]
  12. Nordstrom, B.L.; Friedman, D.S.; Mozaffari, E.; Quigley, H.A.; Walker, A.M. Persistence and adherence with topical glaucoma therapy. Am. J. Ophthalmol. 2005, 140, 598–606. [Google Scholar] [CrossRef] [PubMed]
  13. Belhassen, M.; Laforest, L.; Licaj, I.; Van Ganse, É. Early adherence to antiglaucoma therapy: An observational study. Therapie 2016, 71, 491–499. [Google Scholar] [CrossRef] [PubMed]
  14. Thrimawithana, T.R.; Young, S.; Bunt, C.R.; Green, C.; Alany, R.G. Drug delivery to the posterior segment of the eye. Drug Discov. Today 2011, 16, 270–277. [Google Scholar] [CrossRef] [PubMed]
  15. Lang, J.C. Ocular drug delivery conventional ocular formulations. Adv. Drug Deliv. Rev. 1995, 16, 39–43. [Google Scholar] [CrossRef]
  16. Bourlais, C.L.; Acar, L.; Zia, H.; Sado, P.A.; Needham, T.; Leverge, R. Ophthalmic drug delivery systems--recent advances. Prog. Retin. Eye Res. 1998, 17, 33–58. [Google Scholar] [CrossRef]
  17. Patel, A.; Cholkar, K.; Agrahari, V.; Mitra, A.K. Ocular drug delivery systems: An overview. World J. Pharm. 2013, 2, 47–64. [Google Scholar] [CrossRef]
  18. Elsaid, N.; Jackson, T.L.; Gunic, M.; Somavarapu, S. Positively charged amphiphilic chitosan derivative for the transscleral delivery of rapamycin. Investig. Ophthalmol. Vis. Sci. 2012, 53, 8105–8111. [Google Scholar] [CrossRef]
  19. Elsaid, N.; Somavarapu, S.; Jackson, T.L. Cholesterol-poly(ethylene) glycol nanocarriers for the transscleral delivery of sirolimus. Exp. Eye Res. 2014, 121, 121–129. [Google Scholar] [CrossRef]
  20. Wichterle, O.; Lím, D. Hydrophilic Gels for Biological Use. Nature 1960, 185, 117–118. [Google Scholar] [CrossRef]
  21. 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] [PubMed]
  22. Akulo, K.A.; Adali, T.; Moyo, M.T.G.; Bodamyali, T. Intravitreal Injectable Hydrogels for Sustained Drug Delivery in Glaucoma Treatment and Therapy. Polymers 2022, 14, 2359. [Google Scholar] [CrossRef] [PubMed]
  23. 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]
  24. Kumar, S.; Haglund, B.O.; Himmelstein, K.J. In situ-forming gels for ophthalmic drug delivery. J. Ocul. Pharm. 1994, 10, 47–56. [Google Scholar] [CrossRef] [PubMed]
  25. Kumar, A.; Srivastava, A.; Galaev, I.Y.; Mattiasson, B. Smart polymers: Physical forms and bioengineering applications. Prog. Polym. Sci. 2007, 32, 1205–1237. [Google Scholar] [CrossRef]
  26. Ma, W.; Xu, H.; Nie, S.; Pan, W. Temperature-responsive, Pluronic-g-poly(acrylic acid) copolymers in situ gels for ophthalmic drug delivery: Rheology, in vitro drug release, and in vivo resident property. Drug Dev. Ind. Pharm. 2008, 34, 258–266. [Google Scholar] [CrossRef]
  27. Makwana, S.B.; Patel, V.A.; Parmar, S.J. Development and characterization of in-situ gel for ophthalmic formulation containing ciprofloxacin hydrochloride. Results Pharma Sci. 2016, 6, 1–6. [Google Scholar] [CrossRef]
  28. Gan, L.; Gan, Y.; Zhu, C.; Zhang, X.; Zhu, J. Novel microemulsion in situ electrolyte-triggered gelling system for ophthalmic delivery of lipophilic cyclosporine A: In vitro and in vivo results. Int. J. Pharm. 2009, 365, 143–149. [Google Scholar] [CrossRef]
  29. Qiu, Y.; Park, K. Environment-sensitive hydrogels for drug delivery. Adv. Drug Deliv. Rev. 2001, 53, 321–339. [Google Scholar] [CrossRef]
  30. Blanchard, J. Controlled Drug Delivery: Challenges and Strategies Edited by Kinam Park (Purdue University). American Chemical Society: Washington, DC. 1997. xvii + 629 pp. $145.95. ISBN 0-8412-3418-3. J. Am. Chem. Soc. 1998, 120, 4554–4555. [Google Scholar] [CrossRef]
  31. Schnyder, A.; Huwyler, J. Drug transport to brain with targeted liposomes. NeuroRx 2005, 2, 99–107. [Google Scholar] [CrossRef] [PubMed]
  32. Winfield, A.J.; Jessiman, D.; Williams, A.; Esakowitz, L. A study of the causes of non-compliance by patients prescribed eyedrops. Br. J. Ophthalmol. 1990, 74, 477–480. [Google Scholar] [CrossRef] [PubMed]
  33. Ghate, D.; Edelhauser, H.F. Barriers to glaucoma drug delivery. J. Glaucoma 2008, 17, 147–156. [Google Scholar] [CrossRef] [PubMed]
  34. Hennessy, A.L.; Katz, J.; Covert, D.; Kelly, C.A.; Suan, E.P.; Speicher, M.A.; Sund, N.J.; Robin, A.L. A video study of drop instillation in both glaucoma and retina patients with visual impairment. Am. J. Ophthalmol. 2011, 152, 982–988. [Google Scholar] [CrossRef]
  35. Stone, J.L.; Robin, A.L.; Novack, G.D.; Covert, D.W.; Cagle, G.D. An objective evaluation of eyedrop instillation in patients with glaucoma. Arch. Ophthalmol. 2009, 127, 732–736. [Google Scholar] [CrossRef]
  36. Sleath, B.; Blalock, S.; Covert, D.; Stone, J.L.; Skinner, A.C.; Muir, K.; Robin, A.L. The relationship between glaucoma medication adherence, eye drop technique, and visual field defect severity. Ophthalmology 2011, 118, 2398–2402. [Google Scholar] [CrossRef]
  37. Morse, A.R. Improving Medication Adherence to Reduce Vision Loss in Patients with Glaucoma: Low Hanging Fruit? Ophthalmology 2015, 122, 1280–1282. [Google Scholar] [CrossRef]
  38. Quigley, H.A. Glaucoma. Lancet 2011, 377, 1367–1377. [Google Scholar] [CrossRef]
  39. Schwartz, G.F.; Quigley, H.A. Adherence and persistence with glaucoma therapy. Surv. Ophthalmol. 2008, 53 (Suppl. S1), S57–S68. [Google Scholar] [CrossRef]
  40. Novack, G.D. Ophthalmic drug delivery: Development and regulatory considerations. Clin. Pharm. Ther. 2009, 85, 539–543. [Google Scholar] [CrossRef]
  41. Gupta, S.K.; Galpalli, N.D.; Agrawal, S.S.; Srivastava, S.; Saxena, R. Recent advances in pharmacotherapy of glaucoma. Indian J. Pharm. 2008, 40, 197–208. [Google Scholar] [CrossRef] [PubMed]
  42. Jung, H.J.; Abou-Jaoude, M.; Carbia, B.E.; Plummer, C.; Chauhan, A. Glaucoma therapy by extended release of timolol from nanoparticle loaded silicone-hydrogel contact lenses. J. Control. Release 2013, 165, 82–89. [Google Scholar] [CrossRef] [PubMed]
  43. McNamara, N.A.; Polse, K.A.; Brand, R.J.; Graham, A.D.; Chan, J.S.; McKenney, C.D. Tear mixing under a soft contact lens: Effects of lens diameter. Am. J. Ophthalmol. 1999, 127, 659–665. [Google Scholar] [CrossRef]
  44. Creech, J.L.; Chauhan, A.; Radke, C.J. Dispersive Mixing in the Posterior Tear Film Under a Soft Contact Lens. Ind. Eng. Chem. Res. 2001, 40, 3015–3026. [Google Scholar] [CrossRef]
  45. Zhu, H.; Chauhan, A. Effect of viscosity on tear drainage and ocular residence time. Optom. Vis. Sci. Off. Publ. Am. Acad. Optom. 2008, 85, 715–725. [Google Scholar] [CrossRef]
  46. Li, C.-C.; Chauhan, A. Modeling Ophthalmic Drug Delivery by Soaked Contact Lenses. Ind. Eng. Chem. Res. 2006, 45, 3718–3734. [Google Scholar] [CrossRef]
  47. Kwon, S.; Kim, S.H.; Khang, D.; Lee, J.Y. Potential Therapeutic Usage of Nanomedicine for Glaucoma Treatment. Int. J. Nanomed. 2020, 15, 5745–5765. [Google Scholar] [CrossRef]
  48. Peppas, N.A.; Bures, P.; Leobandung, W.; Ichikawa, H. Hydrogels in pharmaceutical formulations. Eur. J. Pharm. Biopharm. 2000, 50, 27–46. [Google Scholar] [CrossRef]
  49. Jin, R.; Dijkstra, P.J. Hydrogels for Tissue Engineering Applications. In Biomedical Applications of Hydrogels Handbook; Ottenbrite, R.M., Okano, T., Eds.; Springer: Berlin/Heidelberg, Germany, 2010. [Google Scholar] [CrossRef]
  50. Efron, N.; Brennan, N.A.; Chalmers, R.L.; Jones, L.; Lau, C.; Morgan, P.B.; Nichols, J.J.; Szczotka-Flynn, L.B.; Willcox, M.D. Thirty years of ‘quiet eye’ with etafilcon A contact lenses. Contact Lens Anterior Eye 2020, 43, 285–297. [Google Scholar] [CrossRef]
  51. Peng, H.T.; Shek, P.N. Novel wound sealants: Biomaterials and applications. Expert Rev. Med. Devices 2010, 7, 639–659. [Google Scholar] [CrossRef]
  52. Taniguchi, E.V.; Kalout, P.; Pasquale, L.R.; Kohane, D.S.; Ciolino, J.B. Clinicians’ perspectives on the use of drug-eluting contact lenses for the treatment of glaucoma. Ther. Deliv. 2014, 5, 1077–1083. [Google Scholar] [CrossRef] [PubMed]
  53. Xinming, L.; Yingde, C.; Lloyd, A.W.; Mikhalovsky, S.V.; Sandeman, S.R.; Howel, C.A.; Liewen, L. Polymeric hydrogels for novel contact lens-based ophthalmic drug delivery systems: A review. Contact Lens Anterior Eye 2008, 31, 57–64. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, J.; Li, X.; Sun, F. In vitro and in vivo evaluation of ketotifen fumarate-loaded silicone hydrogel contact lenses for ocular drug delivery. Drug Deliv. 2011, 18, 150–158. [Google Scholar] [CrossRef] [PubMed]
  55. Ciolino, J.B.; Ross, A.E.; Tulsan, R.; Watts, A.C.; Wang, R.F.; Zurakowski, D.; Serle, J.B.; Kohane, D.S. Latanoprost-Eluting Contact Lenses in Glaucomatous Monkeys. Ophthalmology 2016, 123, 2085–2092. [Google Scholar] [CrossRef]
  56. Guzman-Aranguez, A.; Colligris, B.; Pintor, J. Contact lenses: Promising devices for ocular drug delivery. J. Ocul. Pharmacol. Ther. 2013, 29, 189–199. [Google Scholar] [CrossRef] [PubMed]
  57. Hsu, K.-H.; Gause, S.; Chauhan, A. Review of ophthalmic drug delivery by contact lenses. J. Drug Deliv. Sci. Technol. 2014, 24, 123–135. [Google Scholar] [CrossRef]
  58. White, C.J.; Byrne, M.E. Molecularly imprinted therapeutic contact lenses. Expert Opin. Drug Deliv. 2010, 7, 765–780. [Google Scholar] [CrossRef]
  59. Yan, F.; Liu, Y.; Han, S.; Zhao, Q.; Liu, N. Bimatoprost Imprinted Silicone Contact Lens to Treat Glaucoma. AAPS PharmSciTech 2020, 21, 63. [Google Scholar] [CrossRef]
  60. Cegielska, O.; Sajkiewicz, P. Targeted Drug Delivery Systems for the Treatment of Glaucoma: Most Advanced Systems Review. Polymers 2019, 11, 1742. [Google Scholar] [CrossRef]
  61. Peng, C.-C.; Burke, M.T.; Carbia, B.E.; Plummer, C.; Chauhan, A. Extended drug delivery by contact lenses for glaucoma therapy. J. Control. Release 2012, 162, 152–158. [Google Scholar] [CrossRef]
  62. Al-Kinani, A.A.; Zidan, G.; Elsaid, N.; Seyfoddin, A.; Alani, A.W.G.; Alany, R.G. Ophthalmic gels: Past, present and future. Adv. Drug Deliv. Rev. 2018, 126, 113–126. [Google Scholar] [CrossRef] [PubMed]
  63. Sahoo, S.K.; Dilnawaz, F.; Krishnakumar, S. Nanotechnology in ocular drug delivery. Drug Discov. Today 2008, 13, 144–151. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, J.; Li, X.; Sun, F. Preparation and evaluation of a contact lens vehicle for puerarin delivery. J. Biomater. Sci. Polym. Ed. 2010, 21, 271–288. [Google Scholar] [CrossRef] [PubMed]
  65. Choi, S.W.; Kim, J. Therapeutic contact lenses with polymeric vehicles for ocular drug delivery: A review. Materials 2018, 11, 1125. [Google Scholar] [CrossRef]
  66. Gulsen, D.; Chauhan, A. Ophthalmic drug delivery through contact lenses. Investig. Ophthalmol. Vis. Sci. 2004, 45, 2342–2347. [Google Scholar] [CrossRef]
  67. Gulsen, D.; Li, C.-C.; Chauhan, A. Dispersion of DMPC liposomes in contact lenses for ophthalmic drug delivery. Curr. Eye Res. 2005, 30, 1071–1080. [Google Scholar] [CrossRef]
  68. Gulsen, D.; Chauhan, A. Dispersion of microemulsion drops in HEMA hydrogel: A potential ophthalmic drug delivery vehicle. Int. J. Pharm. 2005, 292, 95–117. [Google Scholar] [CrossRef]
  69. Kapoor, Y.; Chauhan, A. Ophthalmic delivery of Cyclosporine A from Brij-97 microemulsion and surfactant-laden p-HEMA hydrogels. Int. J. Pharm. 2008, 361, 222–229. [Google Scholar] [CrossRef]
  70. Kapoor, Y.; Chauhan, A. Drug and surfactant transport in Cyclosporine A and Brij 98 laden p-HEMA hydrogels. J. Colloid Interface Sci. 2008, 322, 624–633. [Google Scholar] [CrossRef]
  71. Goyal, G.; Garg, T.; Rath, G.; Goyal, A.K. Current nanotechnological strategies for treating glaucoma. Crit. Rev. Ther. Drug Carr. Syst. 2014, 31, 365–405. [Google Scholar] [CrossRef]
  72. Pita-Thomas, D.W.; Goldberg, J.L. Nanotechnology and glaucoma: Little particles for a big disease. Curr. Opin. Ophthalmol. 2013, 24, 130–135. [Google Scholar] [CrossRef]
  73. Lloyd, A.W.; Faragher, R.G.; Denyer, S.P. Ocular biomaterials and implants. Biomaterials 2001, 22, 769–785. [Google Scholar] [CrossRef]
  74. Hiratani, H.; Fujiwara, A.; Tamiya, Y.; Mizutani, Y.; Alvarez-Lorenzo, C. Ocular release of timolol from molecularly imprinted soft contact lenses. Biomaterials 2005, 26, 1293–1298. [Google Scholar] [CrossRef] [PubMed]
  75. Hiratani, H.; Mizutani, Y.; Alvarez-Lorenzo, C. Controlling drug release from imprinted hydrogels by modifying the characteristics of the imprinted cavities. Macromol. Biosci. 2005, 5, 728–733. [Google Scholar] [CrossRef] [PubMed]
  76. Alvarez-Lorenzo, C.; Hiratani, H.; Gómez-Amoza, J.L.; Martínez-Pacheco, R.; Souto, C.; Concheiro, A. Soft contact lenses capable of sustained delivery of timolol. J. Pharm. Sci. 2002, 91, 2182–2192. [Google Scholar] [CrossRef] [PubMed]
  77. Hiratani, H.; Alvarez-Lorenzo, C. Timolol uptake and release by imprinted soft contact lenses made of N,N-diethylacrylamide and methacrylic acid. J. Control Release 2002, 83, 223–230. [Google Scholar] [CrossRef]
  78. Venkatesh, S.; Sizemore, S.P.; Byrne, M.E. Biomimetic hydrogels for enhanced loading and extended release of ocular therapeutics. Biomaterials 2007, 28, 717–724. [Google Scholar] [CrossRef]
  79. Hiratani, H.; Alvarez-Lorenzo, C. The nature of backbone monomers determines the performance of imprinted soft contact lenses as timolol drug delivery systems. Biomaterials 2004, 25, 1105–1113. [Google Scholar] [CrossRef]
  80. Jung, H.J.; Chauhan, A. Temperature sensitive contact lenses for triggered ophthalmic drug delivery. Biomaterials 2012, 33, 2289–2300. [Google Scholar] [CrossRef]
  81. Maulvi, F.A.; Lakdawala, D.H.; Shaikh, A.A.; Desai, A.R.; Choksi, H.H.; Vaidya, R.J.; Ranch, K.M.; Koli, A.R.; Vyas, B.A.; Shah, D.O. In vitro and in vivo evaluation of novel implantation technology in hydrogel contact lenses for controlled drug delivery. J. Control. Release 2016, 226, 47–56. [Google Scholar] [CrossRef]
  82. Maulvi, F.A.; Soni, P.D.; Patel, P.J.; Desai, A.R.; Desai, D.T.; Shukla, M.R.; Shah, S.A.; Shah, D.O.; Willcox, M.D. Controlled bimatoprost release from graphene oxide laden contact lenses: In vitro and in vivo studies. Colloids Surf. B Biointerfaces 2021, 208, 112096. [Google Scholar] [CrossRef] [PubMed]
  83. Ciolino, J.B.; Stefanescu, C.F.; Ross, A.E.; Salvador-Culla, B.; Cortez, P.; Ford, E.M.; Wymbs, K.A.; Sprague, S.L.; Mascoop, D.R.; Rudina, S.S.; et al. In vivo performance of a drug-eluting contact lens to treat glaucoma for a month. Biomaterials 2014, 35, 432–439. [Google Scholar] [CrossRef] [PubMed]
  84. Sekar, P.; Chauhan, A. Effect of vitamin-E integration on delivery of prostaglandin analogs from therapeutic lenses. J. Colloid Interface Sci. 2019, 539, 457–467. [Google Scholar] [CrossRef] [PubMed]
  85. Peng, C.-C.; Kim, J.; Chauhan, A. Extended delivery of hydrophilic drugs from silicone-hydrogel contact lenses containing vitamin E diffusion barriers. Biomaterials 2010, 31, 4032–4047. [Google Scholar] [CrossRef] [PubMed]
  86. Hsu, K.-H.; Carbia, B.E.; Plummer, C.; Chauhan, A. Dual drug delivery from vitamin E loaded contact lenses for glaucoma therapy. Eur. J. Pharm. Biopharm. 2015, 94, 312–321. [Google Scholar] [CrossRef]
  87. Nicolson, P.C. Continuous Wear Contact Lens Surface Chemistry and Wearability. Eye Contact Lens 2003, 29, S30–S32. Available online: https://journals.lww.com/claojournal/Fulltext/2003/01001/Continuous_Wear_Contact_Lens_Surface_Chemistry_and.9.aspx (accessed on 1 April 2022). [CrossRef]
  88. Soluri, A.; Hui, A.; Jones, L. Delivery of ketotifen fumarate by commercial contact lens materials. Optom. Vis. Sci. 2012, 89, 1140–1149. [Google Scholar] [CrossRef]
  89. Tran, N.P.D.; Yang, M.C.; Tran-Nguyen, P.L. Evaluation of silicone hydrogel contact lenses based on poly(dimethylsiloxane) dialkanol and hydrophilic polymers. Colloids Surf. B Biointerfaces 2021, 206, 111957. [Google Scholar] [CrossRef]
  90. Chen, J.; Wang, D.; Wang, F.; Shi, S.; Chen, Y.; Yang, B.; Tang, Y.; Huang, C. Exendin-4 inhibits structural remodeling and improves Ca2+ homeostasis in rats with heart failure via the GLP-1 receptor through the eNOS/cGMP/PKG pathway. Peptides 2017, 90, 69–77. [Google Scholar] [CrossRef]
  91. Pitt, W.G.; Jack, D.R.; Zhao, Y.; Nelson, J.L.; Pruitt, J.D. Loading and release of a phospholipid from contact lenses. Optom. Vis. Sci. 2011, 88, 502–506. [Google Scholar] [CrossRef]
  92. Rykowska, I.; Nowak, I.; Nowak, R. Soft Contact Lenses as Drug Delivery Systems: A Review. Molecules 2021, 26, 5577. [Google Scholar] [CrossRef] [PubMed]
  93. García-Fernández, M.J.; Tabary, N.; Martel, B.; Cazaux, F.; Oliva, A.; Taboada, P.; Concheiro, A.; Alvarez-Lorenzo, C. Poly-(Cyclo)Dextrins as Ethoxzolamide Carriers in Ophthalmic Solutions and in Contact Lenses. Carbohydr Polym 2013, 98, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  94. Mohammadi, S.; Jones, L.; Gorbet, M. Extended Latanoprost Release from Commercial Contact Lenses: In Vitro Studies Using Corneal Models. PLoS ONE 2014, 9. [Google Scholar] [CrossRef] [PubMed]
  95. Peng, C.C.; Ben-Shlomo, A.; MacKay, E.O.; Plummer, C.E.; Chauhan, A. Drug Delivery by Contact Lens in Spontaneously Glaucomatous Dogs. Curr Eye Res 2012, 37, 204–211. [Google Scholar] [CrossRef] [PubMed]
  96. Das, S.; Saha, D.; Majumdar, S.; Giri, L. Imaging Methods for the Assessment of a Complex Hydrogel as an Ocular Drug Delivery System for Glaucoma Treatment: Opportunities and Challenges in Preclinical Evaluation. Mol. Pharm. 2022, 19, 733–748. [Google Scholar] [CrossRef] [PubMed]
  97. Cheng, Y.-H.; Ko, Y.-C.; Chang, Y.-F.; Huang, S.-H.; Liu, C.J.-L. Thermosensitive chitosan-gelatin-based hydrogel containing curcumin-loaded nanoparticles and latanoprost as a dual-drug delivery system for glaucoma treatment. Exp. Eye Res. 2019, 179, 179–187. [Google Scholar] [CrossRef] [PubMed]
  98. Cheng, Y.-H.; Hung, K.H.; Tsai, T.H.; Lee, C.J.; Ku, R.Y.; Chiu, A.W.H.; Chiou, S.H.; Liu, C.J.L. Sustained delivery of latanoprost by thermosensitive chitosan-gelatin-based hydrogel for controlling ocular hypertension. Acta Biomater. 2014, 10, 4360–4366. [Google Scholar] [CrossRef]
  99. Yang, H.; Leffler, C.T. Hybrid dendrimer hydrogel/poly(lactic-co-glycolic acid) nanoparticle platform: An advanced vehicle for topical delivery of antiglaucoma drugs and a likely solution to improving compliance and adherence in glaucoma management. J. Ocul. Pharmacol. Ther. 2013, 29, 166–172. [Google Scholar] [CrossRef]
  100. Yadav, K.S.; Rajpurohit, R.; Sharma, S. Glaucoma: Current treatment and impact of advanced drug delivery systems. Life Sci. 2019, 221, 362–376. [Google Scholar] [CrossRef]
  101. Kabiri, M.; Kamal, S.H.; Pawar, S.V.; Roy, P.R.; Derakhshandeh, M.; Kumar, U.; Hatzikiriakos, S.G.; Hossain, S.; Yadav, V.G. A stimulus-responsive, in situ-forming, nanoparticle-laden hydrogel for ocular drug delivery. Drug Deliv. Transl. Res. 2018, 8, 484–495. [Google Scholar] [CrossRef]
  102. Gupta, S.; Samanta, M.K.; Raichur, A.M. Dual-drug delivery system based on in situ gel-forming nanosuspension of forskolin to enhance antiglaucoma efficacy. AAPS PharmSciTech 2010, 11, 322–335. [Google Scholar] [CrossRef]
  103. Almeida, J.F.; Fonseca, A.; Baptista, C.M.S.G.; Leite, E.; Gil, M.H. Immobilization of drugs for glaucoma treatment. J. Mater. Sci. Mater. Med. 2007, 18, 2309–2317. [Google Scholar] [CrossRef]
  104. Patel, P.; Patel, G. Formulation, ex-vivo and preclinical in-vivo studies of combined ph and ion-sensitive ocular sustained in situ hydrogel of timolol maleate for the treatment of glaucoma. Biointerface Res. Appl. Chem. 2021, 11, 8242–8265. [Google Scholar] [CrossRef]
  105. Taka, E.; Karavasili, C.; Bouropoulos, N.; Moschakis, T.; Andreadis, D.D.; Zacharis, C.K.; Fatouros, D.G. Ocular co-Delivery of Timolol and Brimonidine from a Self-Assembling Peptide Hydrogel for the Treatment of Glaucoma: In Vitro and Ex Vivo Evaluation. Pharmaceuticals 2020, 13, 126. [Google Scholar] [CrossRef] [PubMed]
  106. Hsiao, M.-H.; Chiou, S.H.; Larsson, M.; Hung, K.H.; Wang, Y.L.; Liu, C.J.L.; Liu, D.M. A temperature-induced and shear-reversible assembly of latanoprost-loaded amphiphilic chitosan colloids: Characterization and in vivo glaucoma treatment. Acta Biomater. 2014, 10, 3188–3196. [Google Scholar] [CrossRef]
  107. Chou, S.-F.; Luo, L.-J.; Lai, J.-Y. In vivo Pharmacological Evaluations of Pilocarpine-Loaded Antioxidant-Functionalized Biodegradable Thermogels in Glaucomatous Rabbits. Sci. Rep. 2017, 7, 42344. [Google Scholar] [CrossRef] [PubMed]
  108. El-Feky, G.S.; Zayed, G.M.; Elshaier, Y.A.M.M.; Alsharif, F.M. Chitosan-Gelatin Hydrogel Crosslinked With Oxidized Sucrose for the Ocular Delivery of Timolol Maleate. J. Pharm. Sci. 2018, 107, 3098–3104. [Google Scholar] [CrossRef]
  109. Review, C. Glaucoma and its treatment. Hosp. Pharm. 1979, 14, 90–101. [Google Scholar]
  110. Natu, M.V.; Sardinha, J.P.; Correia, I.J.; Gil, M.H. Controlled release gelatin hydrogels and lyophilisates with potential application as ocular inserts. Biomed. Mater. 2007, 2, 241–249. [Google Scholar] [CrossRef] [PubMed]
  111. Lai, J.-Y. Biodegradable in situ gelling delivery systems containing pilocarpine as new antiglaucoma formulations: Effect of a mercaptoacetic acid/N-isopropylacrylamide molar ratio. Drug Des. Dev. Ther. 2013, 7, 1273–1285. [Google Scholar] [CrossRef]
  112. Nguyen, D.D.; Luo, L.-J.; Lai, J.-Y. Dendritic Effects of Injectable Biodegradable Thermogels on Pharmacotherapy of Inflammatory Glaucoma-Associated Degradation of Extracellular Matrix. Adv. Healthc. Mater. 2019, 8, e1900702. [Google Scholar] [CrossRef] [PubMed]
  113. Luo, L.-J.; Nguyen, D.D.; Lai, J.-Y. Benzoic acid derivative-modified chitosan-g-poly(N-isopropylacrylamide): Methoxylation effects and pharmacological treatments of Glaucoma-related neurodegeneration. J. Control. Release 2020, 317, 246–258. [Google Scholar] [CrossRef]
  114. Fernández-Colino, A.; Quinteros, D.A.; Allemandi, D.A.; Girotti, A.; Palma, S.D.; Arias, F.J. Self-Assembling Elastin-Like Hydrogels for Timolol Delivery: Development of an Ophthalmic Formulation Against Glaucoma. Mol. Pharm. 2017, 14, 4498–4508. [Google Scholar] [CrossRef]
  115. Marquis, R.E.; Whitson, J.T. Management of glaucoma: Focus on pharmacological therapy. Drugs Aging 2005, 22, 1–21. [Google Scholar] [CrossRef]
  116. Schmidl, D.; Schmetterer, L.; Garhöfer, G.; Popa-Cherecheanu, A. Pharmacotherapy of glaucoma. J. Ocul. Pharmacol. Ther. 2015, 31, 63–77. [Google Scholar] [CrossRef]
  117. Esteban-Pérez, S.; Andrés-Guerrero, V.; López-Cano, J.J.; Molina-Martínez, I.; Herrero-Vanrell, R.; Bravo-Osuna, I. Gelatin Nanoparticles-HPMC Hybrid System for Effective Ocular Topical Administration of Antihypertensive Agents. Pharmaceutics 2020, 12, 306. [Google Scholar] [CrossRef]
  118. Zhang, H.; Luo, Q.; Yang, Z.; Pan, W.; Nie, S. Novel ophthalmic timolol meleate liposomal-hydrogel and its improved local glaucomatous therapeutic effect in vivo. Drug Deliv. 2011, 18, 502–510. [Google Scholar] [CrossRef]
  119. Holden, C.A.; Tyagi, P.; Thakur, A.; Kadam, R.; Jadhav, G.; Kompella, U.B.; Yang, H. Polyamidoamine dendrimer hydrogel for enhanced delivery of antiglaucoma drugs. Nanomedicine 2012, 8, 776–783. [Google Scholar] [CrossRef]
  120. Kulkarni, G.T.; Sethi, N.; Awasthi, R.; Pawar, V.K.; Pahuja, V. Development of Ocular Delivery System for Glaucoma Therapy Using Natural Hydrogel as Film Forming Agent and Release Modifier. Polim. Med. 2016, 46, 25–33. [Google Scholar] [CrossRef]
  121. Karavasili, C.; Komnenou, A.; Katsamenis, O.L.; Charalampidou, G.; Kofidou, E.; Andreadis, D.; Koutsopoulos, S.; Fatouros, D.G. Self-Assembling Peptide Nanofiber Hydrogels for Controlled Ocular Delivery of Timolol Maleate. ACS Biomater. Sci. Eng. 2017, 3, 3386–3394. [Google Scholar] [CrossRef]
  122. Dubey, A.; Prabhu, P. Formulation and evaluation of stimuli-sensitive hydrogels of timolol maleate and brimonidine tartrate for the treatment of glaucoma. Int. J. Pharm. Investig. 2014, 4, 112–118. [Google Scholar] [CrossRef]
  123. Singh, V.; Bushetti, S.S.; Appala, R.; Shareef, A.; Imam, S.S.; Singh, M. Original Article Stimuli-sensitive hydrogels: A novel ophthalmic drug delivery system. Indian J. Ophthalmol. 2010, 58, 477–481. [Google Scholar] [CrossRef]
  124. Pakzad, Y.; Fathi, M.; Omidi, Y.; Mozafari, M.; Zamanian, A. Synthesis and characterization of timolol maleate-loaded quaternized chitosan-based thermosensitive hydrogel: A transparent topical ocular delivery system for the treatment of glaucoma. Int. J. Biol. Macromol. 2020, 159, 117–128. [Google Scholar] [CrossRef]
  125. Wang, J.; Li, B.; Huang, D.; Norat, P.; Grannonico, M.; Cooper, R.C.; Gui, Q.; Chow, W.N.; Liu, X.; Yang, H. Nano-in-Nano Dendrimer Gel Particles for Efficient Topical Delivery of Antiglaucoma Drugs into the Eye. Chem. Eng. J. 2021, 425, 130498. [Google Scholar] [CrossRef]
  126. Wang, F.; Song, Y.; Huang, J.; Wu, B.; Wang, Y.; Pang, Y.; Zhang, W.; Zhu, Z.; Ma, F.; Wang, X. Lollipop-Inspired Multilayered Drug Delivery Hydrogel for Dual Effective, Long-Term, and NIR-Defined Glaucoma Treatment. Macromol. Biosci. 2021, 21, e2100202. [Google Scholar] [CrossRef]
  127. Rahman, M.Q.; Ramaesh, K.; Montgomery, D.M. Brimonidine for glaucoma. Expert Opin. Drug Saf. 2010, 9, 483–491. [Google Scholar] [CrossRef]
  128. Fedorchak, M.V.; Conner, I.P.; Schuman, J.S.; Cugini, A.; Little, S.R. Long Term Glaucoma Drug Delivery Using a Topically Retained Gel/Microsphere Eye Drop. Sci. Rep. 2017, 7, 8639. [Google Scholar] [CrossRef]
  129. Wang, J.; Williamson, G.S.; Lancina Iii, M.G.; Yang, H. Mildly Cross-Linked Dendrimer Hydrogel Prepared via Aza-Michael Addition Reaction for Topical Brimonidine Delivery. J. Biomed. Nanotechnol. 2017, 13, 1089–1096. [Google Scholar] [CrossRef]
  130. Wang, J.; Williamson, G.S.; Yang, H. Branched polyrotaxane hydrogels consisting of alpha-cyclodextrin and low-molecular-weight four-arm polyethylene glycol and the utility of their thixotropic property for controlled drug release. Colloids Surf. B Biointerfaces 2018, 165, 144–149. [Google Scholar] [CrossRef]
  131. Bellotti, E.; Fedorchak, M.V.; Velankar, S.; Little, S.R. Tuning of thermoresponsive pNIPAAm hydrogels for the topical retention of controlled release ocular therapeutics. J. Mater. Chem. B 2019, 7, 1276–1283. [Google Scholar] [CrossRef]
  132. Digiuni, M.; Fogagnolo, P.; Rossetti, L. A review of the use of latanoprost for glaucoma since its launch. Expert Opin. Pharmacother. 2012, 13, 723–745. [Google Scholar] [CrossRef]
  133. Cheng, Y.-H.; Tsai, T.H.; Jhan, Y.Y.; Chiu, A.W.H.; Tsai, K.L.; Chien, C.S.; Chiou, S.H.; Liu, C.J.L. Thermosensitive chitosan-based hydrogel as a topical ocular drug delivery system of latanoprost for glaucoma treatment. Carbohydr. Polym. 2016, 144, 390–399. [Google Scholar] [CrossRef]
  134. Podos, S.M. 2. Epinephrine. Ophthalmology 1980, 87, 721–723. [Google Scholar] [CrossRef]
  135. Hsiue, G.-H.; Hsu, S.; Yang, C.-C.; Lee, S.-H.; Yang, I.-K. Preparation of controlled release ophthalmic drops, for glaucoma therapy using thermosensitive poly-N-isopropylacrylamide. Biomaterials 2002, 23, 457–462. [Google Scholar] [CrossRef]
  136. Prasannan, A.; Tsai, H.-C.; Chen, Y.-S.; Hsiue, G.-H. A thermally triggered in situ hydrogel from poly(acrylic acid-co-N-isopropylacrylamide) for controlled release of antiglaucoma drugs. J. Mater. Chem. B 2014, 2, 1988–1997. [Google Scholar] [CrossRef]
  137. Abu Hashim, I.I.; El-Dahan, M.S.; Yusif, R.M.; Abd-Elgawad, A.-E.H.; Arima, H. Potential use of niosomal hydrogel as an ocular delivery system for atenolol. Biol. Pharm. Bull. 2014, 37, 541–551. [Google Scholar] [CrossRef]
  138. Chang, M.R.; Cheng, Q.; Lee, D.A. Basic science and clinical aspects of wound healing in glaucoma filtering surgery. J. Ocul. Pharm. Ther. 1998, 14, 75–95. [Google Scholar] [CrossRef]
  139. Li, Z.; Van Bergen, T.; Van de Veire, S.; Van de Vel, I.; Moreau, H.; Dewerchin, M.; Maudgal, P.C.; Zeyen, T.; Spileers, W.; Moons, L.; et al. Inhibition of vascular endothelial growth factor reduces scar formation after glaucoma filtration surgery. Investig. Ophthalmol. Vis. Sci. 2009, 50, 5217–5225. [Google Scholar] [CrossRef]
  140. Schlunck, G.; Meyer-ter-Vehn, T.; Klink, T.; Grehn, F. Conjunctival fibrosis following filtering glaucoma surgery. Exp. Eye Res. 2016, 142, 76–82. [Google Scholar] [CrossRef]
  141. Greenfidd, D.S.; Liebmann, J.M.; Jee, J.; Ritch, R. Late-onset bleb leaks after glaucoma filtering surgery. Arch. Ophthalmol. 1998, 116, 443–447. [Google Scholar] [CrossRef]
  142. Kitazawa, Y.; Taniguchi, T.; Nakano, Y.; Shirato, S.; Yamamoto, T. 5-Fluorouracil for trabeculectomy in glaucoma. Graefe’s Arch. Clin. Exp. Ophthalmol. 1987, 225, 403–405. [Google Scholar] [CrossRef]
  143. Mochizuki, K.; Jikihara, S.; Ando, Y.; Hori, N.; Yamamoto, T.; Kitazawa, Y. Incidence of delayed onset infection after trabeculectomy with adjunctive mitomycin C or 5-fluorouracil treatment. Br. J. Ophthalmol. 1997, 81, 877–883. [Google Scholar] [CrossRef]
  144. Yamamoto, T.; Sawada, A.; Mayama, C.; Araie, M.; Ohkubo, S.; Sugiyama, K.; Kuwayama, Y.; Treatment Study Group. The 5-year incidence of bleb-related infection and its risk factors after filtering surgeries with adjunctive mitomycin C: Collaborative bleb-related infection incidence and treatment study 2. Ophthalmology 2014, 121, 1001–1006. [Google Scholar] [CrossRef]
  145. Blake, D.A.; Sahiner, N.; John, V.T.; Clinton, A.D.; Galler, K.E.; Walsh, M.; Arosemena, A.; Johnson, P.Y.; Ayyala, R.S. Inhibition of cell proliferation by mitomycin C incorporated into P(HEMA) hydrogels. J. Glaucoma 2006, 15, 291–298. [Google Scholar] [CrossRef]
  146. Miyata, T.; Asami, N.; Uragani, T. A reversibly antigen-responsive hydrogel. Nature 1999, 399, 766–768. [Google Scholar] [CrossRef]
  147. Guo, T.C.; Qian, Z.Y.; Huang, M.J.; Kan, B.; Gu, Y.C.; Gong, C.Y.; Yang, J.L.; Wang, K.; Dai, M.; Li, X.Y.; et al. Synthesis, characterization, and hydrolytic degradation behavior of a novel biodegradable pH-sensitive hydrogel based on polycaprolactone, methacrylic acid, and poly(ethylene glycol). J. Biomed. Mater. Res. A 2008, 85, 36–46. [Google Scholar] [CrossRef]
  148. Branco, M.C.; Pochan, D.J.; Wagner, N.J.; Schneider, J.P. Macromolecular diffusion and release from self-assembled β-hairpin peptide hydrogels. Biomaterials 2009, 30, 1339–1347. [Google Scholar] [CrossRef]
  149. Liu, Y.; Lu, W.L.; Wang, J.C.; Zhang, X.; Zhang, H.; Wang, X.Q.; Zhou, T.Y.; Zhang, Q. Controlled delivery of recombinant hirudin based on thermo-sensitive Pluronic® F127 hydrogel for subcutaneous administration: In vitro and in vivo characterization. J. Control. Release 2007, 117, 387–395. [Google Scholar] [CrossRef]
  150. Han, Q.; Wang, Y.; Li, X.; Peng, R.; Li, A.; Qian, Z.; Yu, L. Effects of bevacizumab loaded PEG-PCL-PEG hydrogel intracameral application on intraocular pressure after glaucoma filtration surgery. J. Mater. Sci. Mater. Med. 2015, 26, 225. [Google Scholar] [CrossRef]
  151. Liang, L.; Xu, X.-D.; Zhang, X.-Z.; Feng, M.; Peng, C.; Jiang, F.-G. Prevention of filtering surgery failure by subconjunctival injection of a novel peptide hydrogel into rabbit eyes. Biomed. Mater. 2010, 5, 45008. [Google Scholar] [CrossRef]
  152. Martin, G.; Lübke, J.; Schefold, S.; Jordan, J.F.; Schlunck, G.; Reinhard, T.; Kanokwijitsilp, T.; Prucker, O.; Rühe, J.; Anton, A. Prevention of Ocular Tenon Adhesion to Sclera by a PDMAA Polymer to Improve Results after Glaucoma Surgery. Macromol. Rapid Commun. 2020, 41, 1900352. [Google Scholar] [CrossRef]
  153. Frazier, K.; Williams, S.; Kothapalli, D.; Klapper, H.; Grotendorst, G.R. Stimulation of Fibroblast Cell Growth, Matrix Production, and Granulation Tissue Formation by Connective Tissue Growth Factor. J. Investig. Dermatol. 1996, 107, 404–411. [Google Scholar] [CrossRef]
  154. Chen, B.; Wu, P.; Liang, L.; Zhao, C.; Wang, Z.; He, L.; Zhang, R.; Xu, N. Inhibited effect of an RGD peptide hydrogel on the expression of β1-integrin, FAK, and Akt in Tenon’s capsule fibroblasts. J. Biomed. Mater. Res.—Part B Appl. Biomater. 2021, 109, 1857–1865. [Google Scholar] [CrossRef]
  155. Nagata, T.; Harada, Y.; Arai, M.; Hirose, T.; Kondo, H. Polyethylene Glycol-Based Synthetic Hydrogel Sealant for Filtration Bleb Leaks: An In Vivo and Histologic Study. Transl. Vis. Sci. Technol. 2020, 9, 24. [Google Scholar] [CrossRef]
  156. Xi, L.; Wang, T.; Zhao, F.; Zheng, Q.; Li, X.; Luo, J.; Liu, J.; Quan, D.; Ge, J. Evaluation of an injectable thermosensitive hydrogel as drug delivery implant for ocular glaucoma surgery. PLoS ONE 2014, 9, e100632. [Google Scholar] [CrossRef] [PubMed]
  157. Kojima, S.; Sugiyama, T.; Takai, S.; Jin, D.; Ueki, M.; Oku, H.; Tabata, Y.; Ikeda, T. Effects of gelatin hydrogel loading mitomycin C on conjunctival scarring in a canine filtration surgery model. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2601–2605. [Google Scholar] [CrossRef] [PubMed]
  158. Yang, L.Q.; Lan, Y.Q.; Guo, H.; Cheng, L.Z.; Fan, J.Z.; Cai, X.; Zhang, L.M.; Chen, R.F.; Zhou, H.S. Ophthalmic drug-loaded N,O-carboxymethyl chitosan hydrogels: Synthesis, in vitro and in vivo evaluation. Acta Pharmacol. Sin. 2010, 31, 1625–1634. [Google Scholar] [CrossRef] [PubMed]
  159. Peng, R.; Qin, G.; Li, X.; Lv, H.; Qian, Z.; Yu, L. The PEG-PCL-PEG hydrogel as an implanted ophthalmic delivery system after glaucoma filtration surgery; a pilot study. Med. Hypothesis Discov. Innov. Ophthalmol. 2014, 3, 3–8. [Google Scholar]
  160. Wilgus, T.A.; Ferreira, A.M.; Oberyszyn, T.M.; Bergdall, V.K.; DiPietro, L.A. Regulation of scar formation by vascular endothelial growth factor. Lab. Investig. 2008, 88, 579–590. [Google Scholar] [CrossRef]
  161. Kojima, S.; Sugiyama, T.; Takai, S.; Jin, D.; Shibata, M.; Oku, H.; Tabata, Y.; Ikeda, T. Effects of Gelatin Hydrogel Containing Chymase Inhibitor on Scarring in a Canine Filtration Surgery Model. Invest Ophthalmol Vis Sci 2011, 52, 7672–7680. [Google Scholar] [CrossRef]
  162. He, S.; Walls, A.F. Human mast cell chymase induces the accumulation of neutrophils, eosinophils and other in¯ammatory cells in vivo. Br. J. Pharm. 1998, 125, 1491–1500. [Google Scholar] [CrossRef]
  163. Maruichi, M.; Takai, S.; Sugiyama, T.; Ueki, M.; Oku, H.; Sakaguchi, M.; Okamoto, Y.; Muramatsu, M.; Ikeda, T.; Miyazaki, M. Role of chymase on growth of cultured canine Tenon’s capsule fibroblasts and scarring in a canine conjunctival flap model. Exp. Eye Res. 2004, 79, 111–118. [Google Scholar] [CrossRef]
  164. Maeda, M.; Kojima, S.; Sugiyama, T.; Jin, D.; Takai, S.; Oku, H.; Kohmoto, R.; Ueki, M.; Ikeda, T. Effects of gelatin hydrogel containing anti-transforming growth factor-β antibody in a canine filtration surgery model. Int. J. Mol. Sci. 2017, 18, 985. [Google Scholar] [CrossRef]
  165. Jampel, H.D.; Roche, N.; Stark, W.J.; Roberts, A.B. Transforming growth factor-β in human aqueous humor. Curr. Eye Res. 1990, 9, 963–969. [Google Scholar] [CrossRef]
  166. Sun, J.; Liu, X.; Lei, Y.; Tang, M.; Dai, Z.; Yang, X.; Yu, X.; Yu, L.; Sun, X.; Ding, J. Sustained subconjunctival delivery of cyclosporine A using thermogelling polymers for glaucoma filtration surgery. J. Mater. Chem. B 2017, 5, 6400–6411. [Google Scholar] [CrossRef]
  167. Qiao, X.; Peng, X.; Qiao, J.; Jiang, Z.; Han, B.; Yang, C.; Liu, W. Evaluation of a photocrosslinkable hydroxyethyl chitosan hydrogel as a potential drug release system for glaucoma surgery. J. Mater. Sci. Mater. Med. 2017, 28, 149. [Google Scholar] [CrossRef] [PubMed]
  168. Fan, S.Q.; Qin, L.Y.; Cai, J.L.; Zhu, G.Y.; Bin, X.; Yan, H.S. Effect of heparin on production of basic fibroblast growth factor and transforming growth factor-beta1 by human normal skin and hyperplastic scar fibroblasts. J. Burn Care Res. 2007, 28, 734–741. [Google Scholar] [CrossRef] [PubMed]
  169. Chun, Y.Y.; Yap, Z.L.; Seet, L.F.; Chan, H.H.; Toh, L.Z.; Chu, S.W.; Lee, Y.S.; Wong, T.T.; Tan, T.T. Positive-charge tuned gelatin hydrogel-siSPARC injectable for siRNA anti-scarring therapy in post glaucoma filtration surgery. Sci. Rep. 2021, 11, 1470. [Google Scholar] [CrossRef] [PubMed]
  170. Qiao, Y.; Qin, G.; Yu, L. The triblock copolymers hydrogel through intracameral injection may be a new potential ophthalmic drug delivery with antiscaring drugs after glaucoma filtration surgery. Med. Hypotheses 2013, 80, 23–25. [Google Scholar] [CrossRef]
  171. Lin, A.P.; Chung, J.E.; Zhang, K.S.; Chang, M.M.; Orengo-Nania, S.; Gross, R.L.; Chang, P.T. Outcomes of surgical bleb revision for late-onset bleb leaks after trabeculectomy. J. Glaucoma 2013, 22, 21–25. [Google Scholar] [CrossRef]
  172. Radhakrishnan, S.; Quigley, H.A.; Jampel, H.D.; Friedman, D.S.; Ahmad, S.I.; Congdon, N.G.; McKinnon, S. Outcomes of surgical bleb revision for complications of trabeculectomy. Ophthalmology 2009, 116, 1713–1718. [Google Scholar] [CrossRef]
  173. Song, A.; Scott, I.U.; Flynn, H.W.J.; Budenz, D.L. Delayed-onset bleb-associated endophthalmitis: Clinical features and visual acuity outcomes. Ophthalmology 2002, 109, 985–991. [Google Scholar] [CrossRef]
  174. Georgoulas, S.; Dahlmann-Noor, A.; Brocchini, S.; Khaw, P.T. Modulation of wound healing during and after glaucoma surgery. Prog. Brain Res. 2008, 173, 237–254. [Google Scholar] [CrossRef]
  175. Tannenbaum, D.P.; Hoffman, D.; Greaney, M.J.; Caprioli, J. Outcomes of bleb excision and conjunctival advancement for leaking or hypotonous eyes after glaucoma filtering surgery. Br. J. Ophthalmol. 2004, 88, 99–103. [Google Scholar] [CrossRef]
  176. Calladine, D.; Ratnarajan, G.; McAllister, J. New polyethylene glycol ‘hydrogel bandage’ technique to cover the conjunctival-limbal wound in fornix-based trabeculectomy surgery. Clin. Exp. Ophthalmol. 2011, 39, 921–923. [Google Scholar] [CrossRef]
Figure 1. Causes of therapy failure.
Figure 1. Causes of therapy failure.
Gels 08 00510 g001
Figure 2. A nanoparticle-loaded ring implant placed between partially polymerized hydrogel CL.
Figure 2. A nanoparticle-loaded ring implant placed between partially polymerized hydrogel CL.
Gels 08 00510 g002
Table 1. Studies on contact lens hydrogel.
Table 1. Studies on contact lens hydrogel.
AuthorYearDrugGroupPolymer NameManufacturingIOP DecreaseDuration
Maulvi F.A. [82]2021BimatoprostProstaglandinGraphene oxideBimatoprost before polymerization/
Garcia Fernandez M.J. [93]2013EthoxzolamideCAIPoly-HEMA and poly-HEMA-co-APMADry disk immersed into/10 days
Jung H. J. [80]2012TimololB-blockersPropoxylated glyceryl triacylateAdding particles to the polymerization mixture/<=5 days
Maulvi F.A. [81]2016TimololB-blockersEthyl cellulose nanoparticle-laden ringTM loaded ring in hydrogel contact lensDecrease by 6.3 ± 1.92 mmHg after three hours8 days (in vivo)
Mohammadi S. [94]2014LatanoprostProstaglandinBalaficon A/Senofilcon AIncubation in drug solution >24 h
Peng C. C. [95]2012TimololB-blockersNIGHT&DAY silicone hydrogel contact lenses With/without vit. ESoakedDecreased by 5/
Ciolino J.B. [55]2016LatanoprostProstaglandinMethafilicon+ methacrylic acid (Hydrogel)PhotopolymerizationLow dose: decreased > 6. High dose decreased> 10/
Sekar P. [84]2019Bimatoprost and LatanoprostProstaglandinVit E added to ACUVUE OASIS and ACUVUE TRUE EYESoaked/>10 days
Yan F. [59]2020BimatoprostProstaglandinHEMA (hydroxyl ethylmethacrylate)Imprinting vs. soaked/Imprinted 36–60 h
Xu J. [64]2010PuerarinChinese medicine ability to block b-receptorspHEMA-NVP-MASoaked/350 min
Table 3. Studies on anti-scarring hydrogels in glaucoma filtration surgery.
Table 3. Studies on anti-scarring hydrogels in glaucoma filtration surgery.
AuthorIn vitro/VivoHydrogelFunctionDrug DeliveredActivation ModeAdministration Site
Blake et al.
J Glaucoma, 2006 [145]
in vitroP(HEMA)Drug delivery systemMitomycin C//
Liang et al.
Biomed Mater., 2010 [151]
in vivoPeptide hydrogel with RGD sequenceKeeping tissues apart,
inflammatory inhibition
//Filtering bleb
Yang et al.
Acta Pharmacol Sin., 2010 [158]
in vitro and vivoCMCSDrug delivery system5-fluorouracil,
bevacizumab
/Filtering bleb
Kojima et al.
Invest Ophthalmol Vis Sci., 2011 [161]
in vivoGelatin-hydrogelDrug delivery systemChymase inhibitor/Filtering bleb
Xi et al.
PLoS One., 2014 [156]
in vitro and vivoPTMC15-F127-PTMC15Drug delivery systemMitomycin CBody temperatureFiltering bleb
Peng et al.
Med Hypothesis Discov Innov Ophthalmol., 2014 [159]
in vivoPECEDrug delivery systemBevacizumabBody temperatureAnterior chamber
Han et al.
J Mater Sci Mater Med., 2015 [150]
in vitro and vivoPECEDrug delivery systemBevacizumabBody temperatureAnterior chamber
Kojima et al.
Invest Ophthalmol Vis Sci., 2015 [157]
in vivoGelatin-hydrogelDrug delivery systemMitomycin C/Filtering bleb
Sun et al.
J Mater Chem B., 2017 [166]
in vitro and vivoPLGA-PEG-PLGADrug delivery systemCyclosporine ABody temperatureFiltering bleb
Qiao et al.
J Mater Sci Mater Med., 2017 [167]
in vivoHECTSDrug delivery systemHeparinUV irradiationUnder scleral flap
Maeda et al.
Int J Mol Sci., 2017 [164]
in vivoGelatin-hydrogelDrug delivery systemTGF-β antibody/Filtering bleb
Martin et al.
Macromol Rapid Commun., 2020 [152]
in vitroDMAA + AOAQFibroblast cells repellent/UV irradiation/
Chen et al.
J Biomed Mater Res B Appl Biomater., 2021 [154]
in vitroPeptide hydrogel with RGD sequencePeptide competition on protein binding site///
Chun et al.
Sci Rep., 2021 [169]
in vitro and vivoGelatin-tyramineDrug delivery systemsiRNACharge tunabilityFiltering bleb
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fea, A.M.; Novarese, C.; Caselgrandi, P.; Boscia, G. Glaucoma Treatment and Hydrogel: Current Insights and State of the Art. Gels 2022, 8, 510. https://doi.org/10.3390/gels8080510

AMA Style

Fea AM, Novarese C, Caselgrandi P, Boscia G. Glaucoma Treatment and Hydrogel: Current Insights and State of the Art. Gels. 2022; 8(8):510. https://doi.org/10.3390/gels8080510

Chicago/Turabian Style

Fea, Antonio Maria, Cristina Novarese, Paolo Caselgrandi, and Giacomo Boscia. 2022. "Glaucoma Treatment and Hydrogel: Current Insights and State of the Art" Gels 8, no. 8: 510. https://doi.org/10.3390/gels8080510

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

Article Metrics

Back to TopTop