3,299
Views
7
CrossRef citations to date
0
Altmetric
Research Article

Therapeutic applications of contact lens-based drug delivery systems in ophthalmic diseases

, , , , ORCID Icon &
Article: 2219419 | Received 14 Feb 2023, Accepted 15 May 2023, Published online: 02 Jun 2023

Abstract

Traditional ophthalmic drugs, such as eye drops, gels and ointments, are accompanied by many problems, including low bioavailability and potential drug side effects. Innovative ophthalmic drug delivery systems have been proposed to overcome the limitations associated with traditional formulations. Recently, contact lens-based drug delivery systems have gained popularity owing to their advantages of sustained drug delivery, prolonged drug retention, improved bioavailability, and few drug side effects. Various methods have been successfully applied to drug-loaded contact lenses and prolonged the drug release time, such as chemical crosslinking, material embedding, molecular imprinting, colloidal nanoparticles, vitamin E modification, drug polymer film/coating, ion ligand polymerization systems, and supercritical fluid technology. Contact lens-based drug delivery systems play an important role in the treatment of multifarious ophthalmic diseases. This review discusses the latest developments in drug-loaded contact lenses for the treatment of ophthalmic diseases, including preparation methods, application in ophthalmic diseases and future prospects.

1. Introduction

Due to special anatomical and physiological factors, eyes are prone to various diseases such as keratitis, cataract, glaucoma and retinopathy, which endanger the health of patients and affect their quality of life. Currently, vision impairment due to ophthalmic diseases is the primary cause of disability worldwide. By 2015, 34.3 million blind people, 24.3 million people with severe visual impairment, and 214 million with moderate visual impairment were reported worldwide (Stevens et al., Citation2013). Therefore, the active prevention and treatment of ophthalmic diseases is crucial.

Topical drug delivery is the most widely preferred route of drug administration to treat ophthalmic diseases such as keratitis, conjunctivitis, dry eye disease, glaucoma, uveitis and so on. To date, there are multifarious forms of traditional ophthalmic drugs, and approximately 90% of drugs are administered in the form of eye drops (Patel et al., Citation2013). Owing to the complicated structure of eye, lipophilicity of corneal epithelium, defense mechanisms, bonding of drug with proteins contained in tears, enzymolysis, and metabolism, traditional ophthalmic drugs have disadvantages such as low bioavailability and potential drug side effects. In addition, many patients (especially elderly patients) have difficulties in correctly instilling the eye drops, which can reduce the efficiency of the drug and even lead to potential contamination of a chronically used bottle. All these factors limit their development () (Stone et al., Citation2009; Hennessy et al., Citation2010; Toda et al., Citation2011; Ali et al., Citation2016; Awwad et al., Citation2017; Mutlu et al., Citation2019; Akhter et al., Citation2022).

Table 1. Characteristics of different forms of traditional ophthalmic drugs.

To overcome the limitations of traditional ophthalmic drugs, researchers are committed to developing innovative drug formulations with long residence times and high bioavailability. New formulations and improved methods have been continuously developed such as viscosity or permeation enhancers, cyclodextrins, nanoparticles, nanosuspensions, liposomes, dendrimers, polymeric gels, punctal plugs, iontophoresis and so on (Yellepeddi et al., Citation2015; Gote et al., Citation2019; Jumelle et al., Citation2020; Perez et al., Citation2020; Singh et al., Citation2020; Xu et al., Citation2021). Although these measures have achieved initial results eventually, they cannot completely avoid all defects of traditional ophthalmic drugs and also face their own challenges in preparation, quality control and application (Baranowski et al., Citation2014). Therefore, there is an urgent need to develop new drug delivery forms that could maintain pharmacokinetics and pharmacodynamics at least comparable to eye drops, and possess the characteristic of better bioavailability, high safety, good biocompatibility and fewer side effects. Also, this method should preferably provide extended drug release at a therapeutic rate. These factors make drug-loaded contact lenses an obvious choice.

Drug-loaded contact lenses, as new drug delivery tools that use different techniques to load ophthalmic drugs into contact lenses, have attracted considerable attention. Particularly, drug-loaded contact lenses can overcome the problem of incorrectly instilling eyedrops, and also can continuously release drugs from lens to tear film, prolong drug retention time on the ocular surface and improve bioavailability. In the past few years, the development of drug-loaded contact lenses has progressed rapidly, and the contact lens-based drug delivery systems have been proved to play an important role in preventing of keratitis, glaucoma, dry eye, color blindness, corneal injury, keratoconus, ocular scar, retinopathy, and other ophthalmic diseases () (White et al., Citation2011; Ciolino et al., Citation2014; Huang et al., Citation2016; Salih et al., Citation2021; Zhao et al., Citation2021a; Mun et al., Citation2022). To the best of our knowledge, there are no comprehensive and systematic review articles on the role of drug-loaded contact lenses in different ophthalmic diseases. Hence, we summarized the new progress of drug-loaded contact lenses with novel designs in different ophthalmic diseases.

Figure 1. Application of novel contact lenses in ophthalmic diseases.

Figure 1. Application of novel contact lenses in ophthalmic diseases.

2. Novel methods for preparing drug-loaded contact lenses

Until now, immersion method is the simplest way to prepare drug-loaded contact lenses. Nonetheless, owing to the influence of water content of the contact lens and molecular solubility of the drug, this method has disadvantages of low drug loading and fast release speed (Soluri et al., Citation2012). To improve the drug loading capacity and prolong the drug release, many innovations have been made in the lens production process, such as molecular imprinting, polymer nanoparticles, vitamin E barriers, drug polymer film/coating, ionic interactions, supercritical fluid technology, and so on () (Bengani & Chauhan, Citation2013; Nasr et al., Citation2016; Maulvi et al., Citation2016a; Torres-Luna et al., Citation2019a; Varela-Garcia et al., Citation2020; Franco & De Marco, Citation2021; Li et al., Citation2021a).

Table 2. Summary of novel methods for preparing drug-loaded contact lenses and the application in ophthalmic diseases.

2.1. Molecular imprinting

Molecular imprinting is a polymer synthesis technology that uses a template-mediated polymerization mechanism to synthesize macromolecular networks with tailored affinities, capacities, and selectivity for template molecules (White & Byrne, Citation2010). The drug is first polymerized with the functional monomer and then extracted after polymerization, leaving a high-affinity drug-recognition cavity in the polymer network. While reloading, drugs can bind with high-affinity cavities to increase the partition coefficient and interact with functional groups in the polymer network to reduce the diffusivity and prolong the release time (Lanier et al.,Citation2020; Zhang et al., Citation2020). Currently, this technology is widely used to produce drug-loaded contact lenses with sustained release time. Tieppo et al. demonstrated that the residence time of ketotifen for imprinted lenses was 4 and 50 fold greater than non-imprinted lenses and eye drops, respectively (Tieppo et al., Citation2012). Omranipour et al. studied the binding and release characteristics of brimonidine imprinted soft contact lenses and found that all imprinted polymers had higher affinity for brimonidine than non-imprinted polymers, demonstrating the positive effect of the molecular imprinting technique on improving the capacity for drug loading and sustained release (Omranipour et al., Citation2015).

2.2. Polymer nanoparticles

In this technique, drugs are encapsulated with colloidal nanoparticles (polymer nanoparticles, nanoparticles, etc.) and then dispersed into the polymerization medium of unreacted monomers to prepare contact lenses (Maulvi et al., Citation2016a). In many cases, drug molecules need to diffuse out of the nanoparticles and travel through the lens matrix to reach tear film, which generate low diffusion and long release time (Jung & Chauhan, Citation2012). Besides, drug-loaded nanoparticles can enhance drug efficacy by preventing drug interactions with polymer mixtures and avoiding drug metabolism caused by enzymes like lysosomes in tears (Maulvi et al., Citation2016a). Many studies have shown that nanoparticle-loaded hydrogels could be used for extended-delivery drugs. Nasr et al. studied the effects of nanoparticles loading on drug release and found that the hydrogel embedded with nanoparticles can release the drug for a period of 12 days (Nasr et al., Citation2016). In addition, studies about the effects of gold nanoparticles on the loading and delivery of drugs in contact lenses have confirmed that gold nanoparticles can increase drug uptake from soaking solution and improved the release kinetics without affecting the critical properties of contact lenses for therapeutic application (Maulvi et al., Citation2019; Li et al., Citation2021b). Drug-loaded nanocontact lenses have successfully achieved controlled and sustained drug delivery, which is a promising approach to treat ophthalmic diseases (Rodrigues et al., Citation2020; Khan et al., Citation2021; Kusrini et al., Citation2021; Liu et al., Citation2021; Maulvi et al., Citation2021; Li et al., Citation2021a).

2.3. Vitamin E barrier

Vitamin E barrier is a measure to reduce drug delivery rate and extend the release time by setting barriers on the drug transport path. Vitamin E can extend drug-release time through different mechanisms. Hydrophilic drug molecules can be forced to diffuse out of the lens through tortuous pathways, whereas hydrophobic drug molecules can be dissolved at high concentrations (Paradiso et al., Citation2016; Sekar & Chauhan, Citation2019; Liu et al., Citation2022). As reported, adding vitamin E to the lenses preserves visible light transmission and other properties. Vitamin E integration does not affect latanoprost transport and can make the bioavailability of bimatoprost greater than 50% (Sekar & Chauhan, Citation2019). Rad et al. found that when contact lenses were soaked in vitamin E solutions, the water content was significantly reduced, the betamethasone loading capacity enhanced, and the release rate decreased. What’s more, they also found applying vitamin E loading solutions, with 0.1 and 0.2 g/mL concentrations, could effectively enhance cipro release time from 2 hours (in a non-vitamin E–loaded lens) to 14 to 17 and 30 to 33 days, respectively (Rad et al., Citation2016; Rad & Mohajeri, Citation2017). Moreover, the strong antioxidant properties of vitamin E can improve the stability of combined drugs, protect the cornea from UV radiation, and inhibit the process underlying conditions such as cataracts and age-related macular degeneration (Palazzo et al., Citation2020; Tanito, Citation2021; Edwards et al., Citation2022). The advantages of vitamin E in prolonging drug release time without affecting important properties of lens and the strong antioxidant properties are beneficial for the development of drug delivery contact lenses.

2.4. Liposomes

Liposomes are composed of lipid bilayers with stable hydrophobic, hydrophilic or amphiphilic functions and good biodegradability, which are often used as drug delivery carriers. Owing to their special structure, hydrophilic drugs can be encapsulated in the aqueous center surrounded by hydrophilic head, while hydrophobic drugs can be anchored in the hydrophobic tail (Lanier et al., Citation2020). Liposome particles can break down and allow the drug to spread freely through the contact lens matrix before being released into tear film; they can also penetrate the lens and lipid layers of target cells before rupturing and releasing the encapsulated drug, which can prolong the drug release from contact lenses (Sercombe et al., Citation2015; Jain & Shastri, Citation2011). Danion et al. demonstrated that contact lenses bearing surface-immobilized layers of intact liposomes loaded with levofloxacin can provide a sustained release over 6 days, which showed antibacterial activity against Staphylococcus aureus (S. aureus) (Danion et al., Citation2007). In addition, ciprofloxacin released from the liposomes coated on the contact lenses can inhibit both Pseudomonas aeruginosa (P. aeruginosa) and S. aureus, and approximately 40% of ciprofloxacin was retained up to a period of 3 months at 4 °C (Jain & Shastri, Citation2011). The liposome system is highly specific for the sustained application of the drug.

2.5. Ionic interactions

Because ophthalmic drugs are mostly charged under physiological conditions, ionic interactions can be used to increase drug binding to the substrate. The hydrophobic interactions between ionic surfactants and gel polymers and the electrostatic interactions with drugs can lead to stronger adsorption. Ionic drugs can be adsorbed on surfactant-covered polymers to reduce the transport rate and prolong the release time. Torres-Luna et al. incorporated the microemulsion approach with a cationic surfactant in the fabrication of contact lenses and found that cationic surfactant and microemulsions at low cetalkonium chloride weight percentage could prolong the release time (Torres-Luna et al., Citation2019b). The partition coefficient of the drug exponentially increased with surfactant loading in the gel, and drug release duration was increased from about 2 hours to 50 hours in 1-day ACUVUE contact lens (with 10% surfactant). Furthermore, ionic surfactants can increase wettability and reduce the adsorption of proteins without affecting the lens transparency (Bengani & Chauhan, Citation2013; Bengani et al., Citation2013). These advantages make ionic interactions effective for preparing drug-loaded contact lenses.

2.6. Supercritical fluid technology

Supercritical fluid technology is a way that uses supercritical solvents such as CO2 to load/impregnate hydrophilic and hydrophobic drugs into contact lenses. Braga et al. investigated the loading of hydrophobic drug into soft contact lenses using a supercritical fluid-assisted molecular imprinting method. They found both the loading amount and sustained release time of drug were improved after the lens processed by supercritical fluid, and can be controlled by adjusting operating parameters including pressure, temperature and decompression rate (Braga et al., Citation2010, Citation2011; Yañez et al., Citation2011).

In summary, in addition to the above methods, free radical polymerization, drug polymer film/coating, β-cyclodextrin, and artificial intelligence sensors are also effective in improving drug loading and prolonging drug release time, and have been applied in drug-loaded contact lenses (García-Fernández et al., Citation2013; Phan et al., Citation2014a; Aouak et al., Citation2019; Hewitt et al., Citation2020; Li et al., Citation2020; Liu et al., Citation2020; Pillai et al., Citation2020; Zhao et al., Citation2021a, Citation2021b; Wong et al., Citation2022).

3. Novel contact lenses in ophthalmic diseases

3.1. Keratitis

Keratitis is a corneal inflammatory reaction caused by external pathogens or internal diseases that attack corneal tissue, and is often accompanied by symptoms such as eye pain, photophobia, and vision loss. In recent years, its incidence has been increasing yearly with the increase in vegetative ocular trauma, misuse of broad-spectrum antibiotics or hormones, improper use of contact lenses and extensive development of eye surgery (Kam et al., Citation2017; Das et al., Citation2020; Brown et al., Citation2021; Durand et al., Citation2021). Besides, new methods for the treatment of keratitis have been explored, among which drug-loaded contact lenses have been proved to have great potential for clinical application (Shi et al., Citation2013; Hui et al., Citation2014; Phan et al., Citation2014b; Qin et al., Citation2017; Dixon et al., Citation2018). New therapeutic contact lenses for different types of keratitis are described below.

3.1.1. Bacterial keratitis

Bacterial keratitis is a type of purulent keratitis caused by different species of bacteria, P. aeruginosa being among the most common and destructive. Keratitis caused by P. aeruginosa develops rapidly and can evolve into an acute suppurative ulcer within 24–48 hours, lead to vision loss and even eyeball removal if not treated promptly. Recently, the incidence has increased with the widespread use of contact lenses, but treatment options remain scarce and antibiotics lose their specificity due to microbial resistance. Therefore, new therapeutic targets are urgently needed to fill these treatment gaps (Sy et al., Citation2012; Vazirani et al., Citation2015; Fernandes et al., Citation2016; Ung & Chodosh, Citation2021; Gurnani & Kaur, Citation2022; Tuft et al., Citation2022).

New strategies for preparing antibacterial contact lenses, such as antimicrobial peptides, natural antimicrobial products, and antibacterial coatings/films, have been reported (Xiao et al., Citation2018; Nahum et al., Citation2019). Malakooti et al. demonstrated that the imprinted contact lenses loaded with polymyxin B and related antimicrobial peptides can effectively inhibit P. aeruginosa (Malakooti et al., Citation2015). Dutta and coworkers found that melimine-coated lenses can significantly reduce the incidence of P. aeruginosa keratitis in rabbit models (Dutta et al., Citation2013, Citation2014, Citation2016). Moreover, they also demonstrated that some silicone hydrogel contact lenses, which were plasma-coated with acrylic acid followed by Mel4 antimicrobial peptide immobilization by covalent coupling, had excellent bactericidal functions (Dutta et al., Citation2018). Besides, Gallagher et al. reported that contact lenses prepared with an antibacterial peptide hydrogel and penicillin G or poly-ε-lysine can reduce the growth of S. aureus and Escherichia coli (Gallagher et al., Citation2016). Therapeutic contact lenses containing antibacterial peptides offer a potential intervention strategy for the prevention of microbial infections.

Antibacterial coatings/films or antimicrobial drugs also show excellent application value in inhibiting bacterial keratitis. Contact lenses with poly(dimethyl siloxane)-gentamicin sulfate or poly(dimethyl siloxane)-triclosan blend films exhibit good bactericidal and sufficient biofilm inhibition against gram-positive bacteria (Wang et al., Citation2016). Thiosemicarbazone-loaded pHEMA-co-β-cyclodextrin contact lens can effectively inhibit the growth of P. aeruginosa and S. aureus (Glisoni et al., Citation2013). Polyvinyl alcohol (PVA)/anionic collagen membranes as carriers of ciprofloxacin hydrochloride can inhibit bacterial activity and reduce inflammation in canine corneal ulcers (Daza et al., Citation2020). Furthermore, a multifunctional coating containing ZnO nanoparticles, chitosan, and gallic acid can be engineered on contact lens in a one-step sonochemical process, exhibiting high antibacterial efficiency and effectively combating S. aureus (Hoyo et al., Citation2019). Pillai et al. recently found that the contact lenses with antibacterial coating prepared by ozone activation or thermal polymerization showed antibacterial activity against methicillin-resistant S. aureus bacteria with a killing efficacy >99.99% (Pillai et al., Citation2020). Zhao et al. prepared epigallocatechin gallate-loaded starch hydrogel contact lens by free radical polymerization and demonstrated that they can sustainably release drugs for 14 days and significantly reduce the adhesion of P. aeruginosa (Zhao et al., Citation2021a). Moreover, Silva et al. designed a layer-by-layer coating using chitosan, sodium alginate, sodium hyaluronate, and genipin, which can endow contact lenses with ability to regulate the release of the anti-inflammatory diclofenac sodium salt, decreasing the initial burst and inhibiting bacterial growth (Silva et al., Citation2020).

In general, reducing the deposition of proteins and lipids is also important to prevent microbial adhesion and inhibit biofilm formation. Yeh and coworkers developed a stable superhydrophilic zwitterionic interface on polydimethylsiloxane elastomer by covalent silanization of sulfobetaine silane. The zwitterionic silane endows contact lenses with the ability to resist the nonspecific adsorption of proteins and lipids, potentially enhancing the antibacterial performance (Yeh et al., Citation2014). Additionally, Liu et al. confirmed that the contact lenses modified by zwitterionic and antimicrobial metal-phenolic networks can effectively prevent the adhesion of proteins, inhibit biofilm formation, and reduce many eye problems caused by bacterial colonization (Liu et al., Citation2020).

3.1.2. Fungal keratitis

Fungal keratitis is an infectious corneal disease caused by pathogenic fungi. It has a slow onset and long course, and corneal ulcer can appear within a few days of onset. Unfortunately, even if the diagnosis is clear and medication is administered timely, the disease cannot be completely controlled and often requires surgical treatment (Niu et al., Citation2020; Brown et al., Citation2021). In this context, contact lenses, as ideal drug delivery systems, have attracted the interest of researchers in developing antifungal products. In the early exploration, the contact lens could maintain a 100% fungicidal rate within 21 days by adding econazole-impregnated poly(lactic-co-glycolic) acid films (Ciolino et al., Citation2011). Moreover, chlorhexidine-loaded contact lenses can extend the drug-release time to 170 hours after modification with vitamin E (Paradiso et al., Citation2016). Although initial explorations have shown promising results, new preparation strategies compatible with a wide range of drugs should be used to prepare antifungal contact lenses for practical applications (Phan et al., 2014).

Huang et al. used quaternized chitosan, graphene oxide, silver nanoparticles and voriconazole to manufacture a hybrid hydrogel contact lens with antibacterial and antifungal properties. This drug delivery system significantly improved fungal keratitis in 7 days in a fungus-infected mouse model, exhibiting potential for the rapid and effective treatment of fungal keratitis (Huang et al., Citation2016). Additionally, Khan and coworkers developed a surface-modified multifunctional contact lens using a simple and efficient sonochemical approach. The surfaces were modified by applying multifunctional nanocoating comprising tobramycin, gallic acid, and phytomolecules-coated zinc oxide nanoparticles, which allowed the lenses to exhibit remarkable anti-pollution properties. Interestingly, the antifouling features improved the patient comfort level and enhanced the antimicrobial potential of the lenses by inhibiting the adhesion of proteins, lipids and platelets. This design provides a solution for simultaneously addressing the wettability, antifouling and antimicrobial requirements of contact lenses (Khan et al., Citation2021).

3.1.3. Viral keratitis

Viral keratitis is a common ophthalmic disease caused by various viruses, among which herpesviruses are the predominant etiologic agent (Koganti et al., Citation2021). So far, the treatment mainly relies on antiviral eye drops, and novel drug delivery systems are being developed due to eye barriers prevent drugs penetration (Polat et al., Citation2022). Varela-Garcia et al. recently fabricated contact lenses with an affinity for acyclovir and valaciclovir using molecular imprinting techniques and used computational models to screen hydrogel functional monomers suitable for interacting with these drugs. Results clearly suggest that valaciclovir had a stronger electrostatic interaction with methacrylic acid than acyclovir, which increasing the drug loading amount. Moreover, this kind of contact lens could release valaciclovir for 10 hours and shows great potential for the treatment of viral keratitis (Varela-Garcia et al., Citation2020).

3.1.4. Acanthamoeba keratitis

Acanthamoeba keratitis (AK) is a serious inflammation caused by Acanthamoeba sp., which is often contracted following corneal trauma, improper use of contact lenses, corneal transplantation, or contact with contaminated water. In general, AK can cause severe eye pain, inflammation, epithelial or stromal defects, and even vision loss if not diagnosed early and promptly treated (Fanselow et al., Citation2021). Until now, avoiding the risk factors and diagnosing the disease early are the most effective ways to combat AK, and there is an urgent need to develop novel methods to preventing AK (Morgan et al., Citation2020). A recent study detailed the development and testing of dysprosium-based nanoparticles for treating AK. Dysprosium-based nanoparticles, particularly Fe3O4-PEG-Dy2O3 nanocomposites, showed considerable antiamoebic cytotoxicity against Acanthamoeba sp. even after seven days of incubation. This kind of dysprosium-based nanocontact lens is a promising method for preventing AK (Kusrini et al., Citation2021).

3.1.5. Noninfectious keratitis

Recently, with extensive progress in eye surgery and corneal transplantation, the incidence of noninfectious keratitis has increased. In this context, development of drug-loaded contact lenses for the prevention and treatment of postoperative inflammation has important clinical significance (Kaczmarek et al., Citation2014; Behl et al., Citation2016). Carreira et al. prepared a vancomycin-loaded film using chitosan, PVA, and glyoxal to prevent inflammation after keratoprosthesis. The excellent transparency, biosafety, and sustained release effect of the film enable the production of vancomycin-eluted contact lenses (Carreira et al., Citation2014). What’s more, Jeencham et al. successfully developed chitosan and regenerated silk fibroin-blended films, which can be loaded with the anti-inflammatory agent diclofenac sodium to prepare daily disposable therapeutic contact lenses. The results demonstrated that the incorporation of regenerated silk fibroins increased the amorphous portion of the films and prolonged the drug release. This type of therapeutic contact lens can reduce the side effects of drugs and prevent postoperative inflammation (Jeencham et al., Citation2020).

3.2. Corneal wound healing

Dry eye, corneal dystrophy, ocular trauma, limbal stem cell deficiency, and other factors can cause imperfect corneal wound healing, which results in serious complications such as infection, ulcer, and even perforation if not treated promptly. At present, autologous serum, drug-eluting bandages, and amniotic membrane transplantation remain the main therapeutic methods (Sandri et al., Citation2016; Dhillon et al., Citation2020). To develop a safe and effective method, Zhao and coworkers fabricated a rutin-encapsulated gelatin hydrogel contact lens that could release rutin for 14 days and could significantly promote corneal wound healing in rabbits (Zhao et al., Citation2021b). Wei et al. successfully reduced ocular inflammation and facilitated epithelial healing by adding hyaluronic acid (HA) and Pluronic®F127 into contact lenses (Wei et al., Citation2022). Besides, Yin et al. covalently bonded HA with bovine serum albumin/silver (BSA/Ag) porous films via chemical crosslinking and evaluated the therapeutic potential of this complex in an alkali burn-induced corneal injury mouse model. Results proved that the BSA/Ag/HA films had high potential in fabricating contact lenses, exhibiting the capacity to relieve inflammation and improve the rate of corneal healing (Yin et al., Citation2021). Since electric stimulation for corneal wound healing could imitate the natural wound-healing mechanism of the endogenous electric field to facilitate epithelial repair. Wu and colleagues developed a wireless-powered electrical bandage contact lens, which powered by wireless power transfer and can create an electric field on the corneal surface to stimulate corneal epithelial cells and accelerate wound healing. Furthermore, the wireless electrical stimulation circuit employed a flower-shaped layout design that could be compactly integrated on a bandage contact lens without blocking vision. Hence, the wireless and wearable device is promising for the treatment of corneal injuries (Wu et al., Citation2022).

3.3. Proliferative ocular diseases

The eye is an important information collector, whose function depends on the integrity of refractive media. However, proliferative ocular diseases, such as scarring after glaucoma filtration surgery, proliferative vitreoretinopathy, and alkali burns, can affect the function of refractive media and cause certain visual impairments. Pirfenidone (PFD), as a safe and effective innovative broad-spectrum antifibrotic drug, can inhibit proliferative ocular diseases by regulating matrix metalloproteinases, transforming growth factors, and collagen (Zhong et al., Citation2011; Yang et al., Citation2016; Khanum et al., Citation2017; Diaz-Palomera et al., Citation2022). A study conducted by Wu et al. investigated the efficiency of drug delivery in PFD-loaded contact lenses that prepared by embedding an insert of PFD and PVA into two layers of silicone elastomer. Results demonstrated that the contact lens could prolong the residence time of PFD in tears and aqueous humor by five times, while the drug loaded was only one-tenth of that used in eye drops. Furthermore, the addition of PVA makes the lens equipped with higher oxygen permeability and lower protein adsorption characteristics. PFD-PVA-loaded contact lens is a promising drug delivery system for inhibiting proliferative ocular diseases (Wu et al., Citation2021a, Citation2021b).

3.4. Dry eye disease

Dry eye disease (DED) is a chronic illness characterized by symptoms of ocular discomfort and visual dysfunction, which result from abnormal tear quantity, quality, or fluid dynamics. At present, reducing tear evaporation, improving the function of related glands, applying eye drops, and removing inducements play an important role in the treatment of DED. For removing inducements, a study found that contact lenses embedded with ceria nanoparticles can effectively remove reactive oxygen species and avoid DED in high H2O2 environment (Choi et al., Citation2020). Moreover, scleral lenses can retain drugs during prolonged periods due to their tear film reservoir (Lim et al., Citation2009; Ciralsky et al., Citation2015; Polania-Baron et al., Citation2021), and have become a viable option to relieve the symptoms of dry eye syndrome (Bavinger et al., Citation2015; La Porta Weber et al., Citation2016; Marty et al., Citation2022). To alleviate ocular symptoms, White et al. fabricated a hydroxypropyl methylcellulose-loaded contact lens using molecular imprinting technology, which can continuously release hydroxypropyl methylcellulose for up to 60 days and effectively combat DED caused by contact lens (White et al., Citation2011). Besides, Akbari and coworkers prepared HA-loaded chitosan nanoparticles via ionic gelation and then embedded them into a PVA hydrogel implant ring to obtain therapeutic contact lenses. The results demonstrated that the lens could release HA for up to 14 days and effectively treat DED (Akbari et al., Citation2021). To reduce tear evaporation, graphene has been used to prepare contact lenses due to the mass impermeability of pristine graphene lattice (Choi & Park, Citation2017). Lee et al. first demonstrated the dehydration protection of graphene-coated contact lenses by monitoring the change of water evaporation rate from the vial capped with contact lens (Lee et al., Citation2017). In general, drug-loaded contact lenses are the most convenient platform for relieving DED symptoms.

3.5. Keratoconus and myopia

The corneal collagen layer weakens in certain diseases, such as myopia and keratoconus, and its mechanical properties also decline. Until now, corneal collagen cross-linking is often used to solve this problem, but it sometimes requires de-epithelialization, which can damage the eye (Saad et al., Citation2020; Shetty et al., Citation2020; Santodomingo-Rubido et al., Citation2022). A recent study showed that collagen cross-linking can be achieved by embedding flexible reservoirs of hyaluronate-rose bengal (HA-RB) conjugate in a contact lens. More specifically, the contact lens can receive signals using an ASIC chip and deliver the HA-RB conjugate to cornea on-demand via electrical signals. The combination of HA and RB enhances the permeability of RB and allows it to penetrate deep corneal layer without de-epithelialization. What’s more, loading HA-RB conjugate into contact lens can improve the delivery efficiency and prolong residence time of the conjugate on corneal surface, which is beneficial for achieving effective corneal collagen cross-linking (Mun et al., Citation2022). The smart contact lens is expected to be used for noninvasive biophotonic myopia vision correction or keratoconus treatment.

3.6. Ocular cystinosis

Cystinosis is a hereditary disease characterized by the accumulation of cystine crystals in various tissues and organs, eventually leading to progressive multiorgan damage and dysfunction. The disease always affects the kidneys and eyes first, and the accumulation of crystals in the eye often causes photophobia, blepharospasm, corneal scarring, retinopathy, cataract, and even blindness (Biswas et al., Citation2018; Castro-Balado et al., Citation2020). Cysteamine is currently the only effective drug for treating cystinosis, and new drug delivery systems are urgently needed. Dixon et al. designed a cysteamine-loaded contact lens containing 0.3% carbon black, which effectively alleviated photophobia symptoms by reducing transmittance by approximately 50% without changing the lens transparency. Additionally, vitamin E loading can reduce the accumulation of cystine crystals by prolonging the release time of cysteamine and providing additional protection by blocking UV radiation (Dixon & Chauhan, 2019; Hsu et al., Citation2013). Moreover, Liu et al. designed gold nanoparticle-loaded contact lenses by reverting gold precursor in the lenses. This method effectively avoids the loss and the instability or separation of gold nanoparticles in the traditional polymerization process. Owing to the high affinity between cystine and gold, this contact lens can bind cystine to cure ocular cystinosis (Liu et al., Citation2021).

3.7. Glaucoma

Glaucoma is a chronic eye disease caused by the progressive degeneration of retinal ganglion cells, which can lead to secondary optic atrophy, visual field loss, and even permanent blindness. Currently, the limited availability of glaucoma drugs has increased the need for novel anti-glaucoma drug delivery mechanisms with fewer toxic side effects (Aref, Citation2017; Carvalho et al., Citation2015). As reported, implanting timolol maleate-loaded ethyl cellulose nanoparticle-laden ring in hydrogel contact lenses continuously reduce the intra ocular pressure for eight days in a rabbit glaucoma model (Maulvi et al., Citation2016b). Besides, encapsulating a thin latanoprost-polymer film within the periphery of a hydrogel contact lens sustainably released drugs and effectively reduced intraocular pressure in glaucoma monkeys (Ciolino et al., Citation2016). Furthermore, contact lens loaded with latanoprost-poly(lactic-co-glycolic acid) film sustainably release drugs for one month (Ciolino et al., Citation2014). Jung & Chauhan investigated that timolol-loaded nanocontact lenses using diluents as polymeric modifiers extended drug-release time from 1–2 hour to approximately 2–4 weeks (Jung & Chauhan, Citation2012). Additionally, copolymerization and molecular imprinting technologies have also shown good results in glaucoma treatment (Anirudhan et al., Citation2016; Hu et al., Citation2016; Yan et al., Citation2020).

With continuous innovation in preparation technology, innovative drug-loaded contact lenses for treatment of glaucoma have been developed. Kim et al. wrapped diamond particles in polyethyleneimine and then cross-linked them with chitosan and timolol maleate to prepare lysozyme-dependent polysaccharide degradation-mediated diamond nanogel contact lenses, which provides a new platform for lysozyme-triggered delivery system (Kim et al., Citation2014). A study on the preparation of inner layer-embedded contact lenses by sandwich method combined with photopolymerization found that this PH-triggered controlled release system had good stability and could continuously release drugs for more than 10 days in rabbit models (Zhu et al., Citation2018). Mehta et al. used electrohydrodynamic atomization to engineer on-demand coatings for contact lenses. In this technique, poly(N-isopropylacrylamide) and polyvinylpyrrolidone polymers were utilized to encapsulate maleate and were electrically atomized to produce contact lens coatings. Borneol and chitosan were used to modulate the release of timolol maleate. This method provides an alternative formulation for glaucoma treatment (Mehta et al., Citation2019). Prakash designed a highly amorphous hydrophilic composite with β-cyclodextrin and acetazolamide using co-evaporation technology, and subsequently prepared a polymer film on the contact lens surface by combining it with the wet agent PVA. Results demonstrated that the contact lens simultaneously treated glaucoma and dry eye, and the polymer film extended drug-release time from 5 minutes to 3 hours (Prakash & Dhesingh, Citation2017). Hsu et al. added vitamin E to timolol and dorzolamide co-loaded contact lenses to explore the duration of drug release and the efficacy of the combination. The results suggested that vitamin E increased the release durations of both drugs to about 2-days and the lens showed superior IOP reduction with 3–4-fold lower drug payload compared to that of eye drops. In particular, a more important benefit of the lens was that the IOP reduction was maintained for about one week after the removal of contact lenses (Hsu et al., Citation2015). These results are encouraging and advance the development of drug delivery systems for glaucoma treatment.

3.8. Uveitis

Uveitis is a vision-threatening disease that can be caused by noninfectious or infectious etiologies. Owing to therapeutic drugs often need to penetrate the ocular surface tissue into the inner eye, and the required concentration and dose are very high, resulting in greater drug side effects and poor compliance (Sève et al., Citation2017). With the development of new materials, several approaches have been proposed for developing novel sustainable delivery systems. Bengani et al. encapsulated a ring-shaped dexamethasone-polymer film into the periphery of a methafilcon contact lens and evaluated its safety and efficacy by rabbit models of experimental anterior uveitis. Results showed that the lens had good biological safety and could sustained release dexamethasone for at least seven days, which effectively inhibited lipopolysaccharide-induced anterior uveitis for five days (Bengani et al.,Citation2020). Similarly, DiPasquale and coworkers used a new macromolecular memory strategy to successfully load bromfenac into silico-hydrogel contact lenses that could sustainably be released for eight days in rabbits (DiPasquale et al., Citation2022). These studies highlight the great potential of drug-release lenses as a platform strategy and provide a new drop-free clinical strategy for uveitis treatment.

3.9. Color vision deficiency

Color vision deficiency (CVD) is an incurable congenital eye disorder that limits patient’s abilities to distinguish specific colors. Patients with CVD often need to use color vision filters like chromogen filters, tinted glass/lens to enhance their color perception, which is considered unesthetic (Badawy et al., Citation2018; Sekar et al., Citation2019; Male et al., Citation2022). Recently, with the development of metasurfaces and nanoparticle technology, innovative contact lenses for CVD have emerged. Nanocomposite contact lenses targeting red-green CVD have been developed to improve color perception ability. In this design, the orientation of nanocomposites can be shifted by uniaxial drawing to filter out the optical wavelengths of specific colors that are difficult to distinguish for CVD patients (Salih et al., Citation2021). Besides, Karepov et al. embedded metasurfaces into contact lenses and simulated their effects on color perception by using conventional models of human color-sensitive photoreceptors. As results suggested, the metasurface-based contact lenses could shift incorrectly perceived pigments back better approximate the original pigments (Karepov & Ellenbogen, Citation2020). Further, Roostaei team prepared a 2D biocompatible and flexible plasmonic contact lens using polydimethylsiloxane based on the soft nano-lithography method, which offers a good color filter for CVD correction (Roostaei & Hamidi, Citation2022). Consequently, these studies demonstrate that novel contact lenses provide new ideas for CVD correction and other color filtration.

3.10. Diabetic-related eye complications

Diabetes is a chronic metabolic disease caused by genetic, environmental, living habits, and other factors, which can easily to induce various tissue complications. Among these, ocular complications, such as diabetic retinopathy and cataract, can affect people’s health and quality of life (Seewoodhary, Citation2021). Hence, continuous blood glucose monitoring is essential to eliminate the health risks and long-term complications associated with diabetes. However, traditional detection methods are mostly invasive and only provide point but not continuous glucose monitoring information.

Owing to the discovery of glucose in tears and the development of ophthalmic sensors, wearable contact lens biosensors for glucose monitoring have attracted the attention of researchers (Farandos et al., Citation2015; Zha et al., Citation2020; Dennyson Savariraj et al., Citation2021; Kar et al., Citation2022). Elsherif et al. developed a glucose-sensitive photonic microstructure sensor integrated with a contact lens for continuous glucose monitoring by using smartphone camera readouts (Elsherif et al., Citation2018). They also created a bifocal contact lens by attaching a hydrogel glucose sensor to a soft contact lens to monitor the glucose concentration and correct myopia and presbyopia. When tear glucose level is increased, the contact lens can respond rapidly to this change in the form of changes in the refractive index and groove depth of the Fresnel lens. Quantitative readouts can then be obtained from smartphones and photodetectors to reflect the blood glucose concentration by measuring the changes in optical power reflected from the bifocal contact lens (Elsherif et al., Citation2021).

In addition to blood glucose monitoring, the treatment of diabetes-related eye complications is also important for improving quality of life. Alvarez-Rivera et al. designed a contact lens for local prevention/treatment of diabetes-related ocular diseases by adding bioinspired functional groups and an aldose reductase inhibitor (epalrestat) to the hydrogel material. Under hyperglycemic conditions, the contact lenses continuously release epalrestat to prevent lens opacification (Alvarez-Rivera et al., Citation2018). Ross et al. prepared an innovative delivery system by encapsulating dexamethasone polymer films inside contact lenses to continuously deliver drugs to retina. In a rabbit model of diabetic retinopathy induced by intravitreal injection of vascular endothelial growth factor, this type of contact lens successfully inhibited retinal vascular leakage (Ross et al., Citation2019). In summary, these studies confirmed that the contact lens, as a minimally invasive diagnostic and drug delivery platform, is expected to play an important role in blood glucose monitoring and the treatment of diabetes-related ocular complications.

Although various technological innovations have extended drug-release time from contact lenses, several challenges in application limit the commercialization. For example, particle instability in nanoparticle-loaded contact lenses can occur during polymerization, sterilization, or storage. Highly cross-linked structure of molecularly imprinted hydrogels can affect optical and physical properties of contact lenses, and low water content will reduce the permeability of ions and oxygen. Besides, vitamin E barrier can affect the physical properties of contact lenses, reduce oxygen permeability and increase protein adsorption. Multilayer liposomes can reduce the permeability of oxygen and carbon dioxide. In a word, contact lens-based drug delivery systems in ophthalmic diseases needs to be further optimized and developed.

4. Conclusions

Contact lens-based drug delivery systems has been widely studied because of their advantages including sustained drug delivery, prolonged drug retention, improved bioavailability and few drug side effects. They are expected to be potential treatments for ophthalmic diseases. Despite series of technological innovations have been achieved in contact lens-based drug delivery systems, most of them are still limited to the laboratory level. Technological challenges such as optical and physical properties, manufacturing, sterilization, and storage, together with non-technological challenges including complications associated with long-term wear, public acceptance of the technology, market regulation and so on, should be overcome for scaling up contact lens-based drug delivery systems. In the future, we need to balance optimization for optical and physical properties with adequate drug loading and release, and solve the processing and storage issues to increase chances of successful commercialization. Besides, it is also necessary to incorporate drugs to reduce complications such as microbial keratitis, allergic conjunctivitis, and dry eye syndrome. These measurements will not only provide new strategies for clinically treating ophthalmic diseases and but also to some extent benefit commercialization of drug-loaded contact lenses.

Authors’ contributions

Lianghui Zhao: sorting out the references and writing original draft; Jike Song: drawing the figure and tables; Yongle Du and Cong Ren: consulting and sorting out the references; Bin Guo and Hongsheng Bi: conceiving and revising the manuscript. All authors have read and agreed to the published version of the manuscript, and that all authors agree to be accountable for all aspects of the work.

Disclosure statement

The authors declare no conflict of interest.

Additional information

Funding

This work was supported by the National Key Research and Development Project (2019YFC1710200), the Focus on Research and Development Plan in Shandong Province (2021LCZX09), and the Natural Science Foundation of Shandong Province (ZR2021LZY045 and ZR2020MH393). The funders had no role in study design, data collection and analysis, the decision to publish, or the preparation of the manuscript.

References

  • Akbari E, Imani R, Shokrollahi P, et al. (2021). Preparation of nanoparticle-containing ring-implanted poly(vinyl alcohol) contact lens for sustained release of hyaluronic acid. Macromol Biosci 21:1.
  • Akhter MH, Ahmad I, Alshahrani MY, et al. (2022). Drug delivery challenges and current progress in nanocarrier-based ocular therapeutic system. Gels 8:82.
  • Ali J, Fazil M, Qumbar M, et al. (2016). Colloidal drug delivery system: amplify the ocular delivery. Drug Deliv 23:710–14.
  • Alvarez-Rivera F, Concheiro A, Alvarez-Lorenzo C. (2018). Epalrestat-loaded silicone hydrogels as contact lenses to address diabetic-eye complications. Eur J Pharm Biopharm 122:126–36.
  • Anirudhan TS, Nair AS, Parvathy J. (2016). Extended wear therapeutic contact lens fabricated from timolol imprinted carboxymethyl chitosan-g-hydroxy ethyl methacrylate-g-poly acrylamide as a onetime medication for glaucoma. Eur J Pharm Biopharm 109:61–71.
  • Aouak T, Saeed WS, Al-Hafi NM, et al. (2019). Poly (2-hydroxyethylmethacrylate -co-methylmethacrylate)/lignocaine contact lens preparation, characterization, and in vitro release dynamic. Polymers 11:917.
  • Aref AA. (2017). Sustained drug delivery for glaucoma: current data and future trends. Curr Opin Ophthalmol 28:169–74.
  • Awwad S, Mohamed Ahmed AHA, Sharma G, et al. (2017). Principles of pharmacology in the eye. Br J Pharmacol 174:4205–23.
  • Badawy AR, Hassan MU, Elsherif M, et al. (2018). Contact lenses for color blindness. Adv Healthc Mater 7:e1800152.
  • Baranowski P, Karolewicz B, Gajda M, et al. (2014). Ophthalmic drug dosage forms: characterisation and research methods. ScientificWorldJournal 2014:861904.
  • Bavinger JC, DeLoss K, Mian SI. (2015). Scleral lens use in dry eye syndrome. Curr Opin Ophthalmol 26:319–24.
  • Behl G, Iqbal J, O’Reilly NJ, et al. (2016). Synthesis and characterization of poly(2-hydroxyethylmethacrylate) contact lenses containing chitosan nanoparticles as an ocular delivery system for dexamethasone sodium phosphate. Pharm Res 33:1638–48.
  • Bengani LC, Chauhan A. (2013). Extended delivery of an anionic drug by contact lens loaded with a cationic surfactant. Biomaterials 34:2814–21.
  • Bengani LC, Hsu KH, Gause S, et al. (2013). Contact lenses as a platform for ocular drug delivery. Expert Opin Drug Deliv 10:1483–96.
  • Bengani LC, Kobashi H, Ross AE, et al. (2020). Steroid-eluting contact lenses for corneal and intraocular inflammation. Acta Biomater 116:149–61.
  • Biswas S, Gaviria M, Malheiro L, et al. (2018). Latest clinical approaches in the ocular management of cystinosis: a review of current practice and opinion from the ophthalmology cystinosis forum. Ophthalmol Ther 7:307–22.
  • Braga ME, Yañez F, Alvarez-Lorenzo C, et al. (2010). Improved drug loading/release capacities of commercial contact lenses obtained by supercritical fluid assisted molecular imprinting methods. J Control Release 148:e102–e104.
  • Braga ME, Costa VP, Pereira MJ, et al. (2011). Effects of operational conditions on the supercritical solvent impregnation of acetazolamide in Balafilcon A commercial contact lenses. Int J Pharm 420:231–43.
  • Brown L, Leck AK, Gichangi M, et al. (2021). The global incidence and diagnosis of fungal keratitis. Lancet Infect Dis 21:e49–e57.
  • Carreira AS, Ferreira P, Ribeiro MP, et al. (2014). New drug-eluting lenses to be applied as bandages after keratoprosthesis implantation. Int J Pharm 477:218–26.
  • Carvalho IM, Marques CS, Oliveira RS, et al. (2015). Sustained drug release by contact lenses for glaucoma treatment-a review. J Control Release 202:76–82.
  • Castro-Balado A, Mondelo-García C, Varela-Rey I, et al. (2020). Recent research in ocular cystinosis: drug delivery systems, cysteamine detection methods and future perspectives. Pharmaceutics 12:1177.
  • Choi K, Park HG. (2017). Smart reinvention of the contact lens with graphene. ACS Nano 11:5223–6.
  • Choi SW, Cha BG, Kim J. (2020). Therapeutic contact lens for scavenging excessive reactive oxygen species on the ocular surface. ACS Nano 14:2483–96.
  • Ciolino JB, Hudson SP, Mobbs AN, et al. (2011). A prototype antifungal contact lens. Invest Ophthalmol Vis Sci 52:6286–91.
  • Ciolino JB, Stefanescu CF, Ross AE, et al. (2014). In vivo performance of a drug-eluting contact lens to treat glaucoma for a month. Biomaterials 35:432–9.
  • Ciolino JB, Ross AE, Tulsan R, et al. (2016). Latanoprost-eluting contact lenses in glaucomatous monkeys. Ophthalmology 123:2085–92.
  • Ciralsky JB, Chapman KO, Rosenblatt MI, et al. (2015). Treatment of refractory persistent corneal epithelial defects: a standardized approach using continuous wear PROSE therapy. Ocul Immunol Inflamm 23:219–24.
  • Danion A, Arsenault I, Vermette P. (2007). Antibacterial activity of contact lenses bearing surface-immobilized layers of intact liposomes loaded with levofloxacin. J Pharm Sci 96:2350–63.
  • Das S, Garg P, Mullick R, et al. (2020). Keratitis following laser refractive surgery: clinical spectrum, prevention and management. Indian J Ophthalmol 68:2813–8.
  • Daza JHU, Righetto GM, Chaud MV, et al. (2020). PVA/anionic collagen membranes as drug carriers of ciprofloxacin hydrochloride with sustained antibacterial activity and potential use in the treatment of ulcerative keratitis. J Biomater Appl 35:301–12.
  • Dennyson Savariraj A, Salih A, Alam F, et al. (2021). Ophthalmic sensors and drug delivery. ACS Sens 6:2046–76.
  • Dhillon HK, Bahadur H, Raj A. (2020). A comparative study of tarsorrhaphy and amniotic membrane transplantation in the healing of persistent corneal epithelial defects. Indian J Ophthalmol 68:29–33.
  • Diaz-Palomera CD, Vidal-Paredes IA, Navarro-Partida J, et al. (2022). Topical pirfenidone-loaded liposomes ophthalmic formulation reduces haze development after corneal alkali burn in mice. Pharmaceutics 14:316.
  • DiPasquale SA, Wuchte LD, Mosley RJ, et al. (2022). One week sustained in vivo therapeutic release and safety of novel extended-wear silicone hydrogel contact lenses. Adv Healthc Mater 11:e2101263.
  • Dixon P, Ghosh T, Mondal K, et al. (2018). Controlled delivery of pirfenidone through vitamin E-loaded contact lens ameliorates corneal inflammation. Drug Deliv Transl Res 8:1114–26.
  • Dixon P, Chauhan A. (2019). Carbon black tinted contact lenses for reduction of photophobia in cystinosis patients. Curr Eye Res 44:497–504.
  • Durand ML, Barshak MB, Chodosh J. (2021). Infectious keratitis in 2021. JAMA 326:1319–20.
  • Dutta D, Cole N, Kumar N, et al. (2013). Broad spectrum antimicrobial activity of melimine covalently bound to contact lenses. Invest Ophthalmol Vis Sci 54:175–82.
  • Dutta D, Ozkan J, Willcox MD. (2014). Biocompatibility of antimicrobial melimine lenses: rabbit and human studies. Optom Vis Sci 91:570–81.
  • Dutta D, Vijay AK, Kumar N, et al. (2016). Melimine-coated antimicrobial contact lenses reduce microbial keratitis in an animal model. Invest Ophthalmol Vis Sci 57:5616–24.
  • Dutta D, Kamphuis B, Ozcelik B, et al. (2018). Development of silicone hydrogel antimicrobial contact lenses with Mel4 peptide coating. Optom Vis Sci 95:937–46.
  • Edwards G, Olson CG, Euritt CP, et al. (2022). Molecular mechanisms underlying the therapeutic role of vitamin E in age-related macular degeneration. Front Neurosci 16:890021.
  • Elsherif M, Hassan MU, Yetisen AK, et al. (2018). Wearable contact lens biosensors for continuous glucose monitoring using smartphones. ACS Nano 12:5452–62.
  • Elsherif M, Alam F, Salih AE, et al. (2021). Wearable bifocal contact lens for continual glucose monitoring integrated with smartphone readers. Small 17:e2102876.
  • Fanselow N, Sirajuddin N, Yin XT, et al. (2021). Acanthamoeba keratitis, pathology, diagnosis and treatment. Pathogens 10:323.
  • Farandos NM, Yetisen AK, Monteiro MJ, et al. (2015). Contact lens sensors in ocular diagnostics. Adv Healthc Mater 4:792–810.
  • Fernandes M, Vira D, Medikonda R, et al. (2016). Extensively and pan-drug resistant pseudomonas aeruginosa keratitis: clinical features, risk factors, and outcome. Graefes Arch Clin Exp Ophthalmol 254:315–22.
  • Franco P, De Marco I. (2021). Contact lenses as ophthalmic drug delivery systems: a review. Polymers 13:1102.
  • Gallagher AG, Alorabi JA, Wellings DA, et al. (2016). A novel peptide hydrogel for an antimicrobial bandage contact lens. Adv Healthc Mater 5:2013–8.
  • García-Fernández MJ, Tabary N, Martel B, et al. (2013). Poly-(cyclo)dextrins as ethoxzolamide carriers in ophthalmic solutions and in contact lenses. Carbohydr Polym 98:1343–52.
  • Glisoni RJ, García-Fernández MJ, Pino M, et al. (2013). β-Cyclodextrin hydrogels for the ocular release of antibacterial thiosemicarbazones. Carbohydr Polym 93:449–57.
  • Gote V, Sikder S, Sicotte J, et al. (2019). Ocular drug delivery: present innovations and future challenges. J Pharmacol Exp Ther 370:602–24.
  • Gurnani B, Kaur K. (2022). Bacterial keratitis. In: StatPearls. Treasure Island (FL): StatPearls Publishing.
  • Hennessy AL, Katz J, Covert D, et al. (2010). Videotaped evaluation of eyedrop instillation in glaucoma patients with visual impairment or moderate to severe visual field loss. Ophthalmology 117:2345–52.
  • Hewitt MG, Morrison PWJ, Boostrom HM, et al. (2020). In vitro topical delivery of chlorhexidine to the cornea: enhancement using drug-loaded contact lenses and β-cyclodextrin complexation, and the importance of simulating tear irrigation. Mol Pharm 17:1428–41.
  • Hoyo J, Ivanova K, Guaus E, et al. (2019). Multifunctional ZnO NPs-chitosan-gallic acid hybrid nanocoating to overcome contact lenses associated conditions and discomfort. J Colloid Interface Sci 543:114–21.
  • Hsu KH, Fentzke RC, Chauhan A. (2013). Feasibility of corneal drug delivery of cysteamine using vitamin E modified silicone hydrogel contact lenses. Eur J Pharm Biopharm 85:531–40.
  • Hsu KH, Carbia BE, Plummer C, et al. (2015). Dual drug delivery from vitamin E loaded contact lenses for glaucoma therapy. Eur J Pharm Biopharm 94:312–21.
  • Hu X, Tan H, Hao L. (2016). Functional hydrogel contact lens for drug delivery in the application of oculopathy therapy. J Mech Behav Biomed Mater 64:43–52.
  • Huang JF, Zhong J, Chen GP, et al. (2016). A hydrogel-based hybrid theranostic contact lens for fungal keratitis. ACS Nano 10:6464–73.
  • Hui A, Willcox M, Jones L. (2014). In vitro and in vivo evaluation of novel ciprofloxacin-releasing silicone hydrogel contact lenses. Invest Ophthalmol Vis Sci 55:4896–904.
  • Jain RL, Shastri JP. (2011). Study of ocular drug delivery system using drug-loaded liposomes. Int J Pharm Investig 1:35–41.
  • Jeencham R, Sutheerawattananonda M, Rungchang S, et al. (2020). Novel daily disposable therapeutic contact lenses based on chitosan and regenerated silk fibroin for the ophthalmic delivery of diclofenac sodium. Drug Deliv 27:782–90.
  • Jumelle C, Gholizadeh S, Annabi N, et al. (2020). Advances and limitations of drug delivery systems formulated as eye drops. J Control Release 321:1–22.
  • Jung HJ, Chauhan A. (2012). Temperature sensitive contact lenses for triggered ophthalmic drug delivery. Biomaterials 33:2289–300.
  • Kaczmarek JC, Tieppo A, White CJ, et al. (2014). Adjusting biomaterial composition to achieve controlled multiple-day release of dexamethasone from an extended-wear silicone hydrogel contact lens. J Biomater Sci Polym Ed 25:88–100.
  • Kam KW, Yung W, Li GKH, et al. (2017). Infectious keratitis and orthokeratology lens use: a systematic review. Infection 45:727–35.
  • Kar A, Ahamad N, Dewani M, et al. (2022). Wearable and implantable devices for drug delivery: applications and challenges. Biomaterials 283:121435.
  • Karepov S, Ellenbogen T. (2020). Metasurface-based contact lenses for color vision deficiency. Opt Lett 45:1379–82.
  • Khan SA, Shahid S, Mahmood T, et al. (2021). Contact lenses coated with hybrid multifunctional ternary nanocoatings (phytomolecule-coated ZnO nanoparticles: gallic acid: tobramycin) for the treatment of bacterial and fungal keratitis. Acta Biomater 128:262–76.
  • Khanum BNMK, Guha R, Sur VP, et al. (2017). Pirfenidone inhibits post-traumatic proliferative vitreoretinopathy. Eye (Lond) 31:1317–28.
  • Kim HJ, Zhang K, Moore L, et al. (2014). Diamond nanogel-embedded contact lenses mediate lysozyme-dependent therapeutic release. ACS Nano 8:2998–3005.
  • Koganti R, Yadavalli T, Naqvi RA, et al. (2021). Pathobiology and treatment of viral keratitis. Exp Eye Res 205:108483.
  • Kusrini E, Sabira K, Hashim F, et al. (2021). Design, synthesis and antiamoebic activity of dysprosium-based nanoparticles using contact lenses as carriers against Acanthamoeba sp. Acta Ophthalmol 99:e178–e188.
  • La Porta Weber S, Becco de Souza R, Gomes JÁP, et al. (2016). The use of the Esclera scleral contact lens in the treatment of moderate to severe dry eye disease. Am J Ophthalmol 163:167–73.e1.
  • Lanier OL, Christopher KG, Macoon RM, et al. (2020). Commercialization challenges for drug eluting contact lenses. Expert Opin Drug Deliv 17:1133–49.
  • Lee S, Jo I, Kang S, et al. (2017). Smart contact lenses with graphene coating for electromagnetic interference shielding and dehydration protection. ACS Nano 11:5318–24.
  • Li Z, Liu M, Ke L, et al. (2021a). Flexible polymeric nanosized micelles for ophthalmic drug delivery: research progress in the last three years. Nanoscale Adv 3:5240–54.
  • Li Q, Ma C, Ma Y, et al. (2021b). Sustained bimatoprost release using gold nanoparticles laden contact lenses. J Biomater Sci Polym Ed 32:1618–34.
  • Li R, Guan X, Lin X, et al. (2020). Poly(2-hydroxyethyl methacrylate)/β-cyclodextrin-hyaluronan contact lens with tear protein adsorption resistance and sustained drug delivery for ophthalmic diseases. Acta Biomater 110:105–18.
  • Lim M, Jacobs DS, Rosenthal P, et al. (2009). The Boston Ocular Surface Prosthesis as a novel drug delivery system for bevacizumab. Semin Ophthalmol 24:149–55.
  • Liu G, Li K, Wang H, et al. (2020). Stable fabrication of zwitterionic coating based on copper-phenolic networks on contact lens with improved surface wettability and broad-spectrum antimicrobial activity. ACS Appl Mater Interfaces 12:16125–36.
  • Liu Z, Kompella UB, Chauhan A. (2021). Gold nanoparticle synthesis in contact lenses for drug-less ocular cystinosis treatment. Eur J Pharm Biopharm 165:271–8.
  • Liu Z, Overton M, Chauhan A. (2022). Transport of vitamin E from ethanol/water solution into contact lenses and impact on drug transport. J Ocul Pharmacol Ther 38:396–403.
  • Malakooti N, Alexander C, Alvarez-Lorenzo C. (2015). Imprinted contact lenses for sustained release of polymyxin B and related antimicrobial peptides. J Pharm Sci 104:3386–94.
  • Male SR, Shamanna BR, Bhardwaj R, et al. (2022). Color vision devices for color vision deficiency patients: a systematic review and meta-analysis. Health Sci Rep 5:e842.
  • Marty AS, Jurkiewicz T, Mouchel R, et al. (2022). Benefits of scleral lens in the management of irregular corneas and dry eye syndrome after refractive surgery. Eye Contact Lens 48:318–21.
  • Maulvi FA, Soni TG, Shah DO. (2016a). A review on therapeutic contact lenses for ocular drug delivery. Drug Deliv 23:3017–26.
  • Maulvi FA, Lakdawala DH, Shaikh AA, et al. (2016b). In vitro and in vivo evaluation of novel implantation technology in hydrogel contact lenses for controlled drug delivery. J Control Release 226:47–56.
  • Maulvi FA, Patil RJ, Desai AR, et al. (2019). Effect of gold nanoparticles on timolol uptake and its release kinetics from contact lenses: in vitro and in vivo evaluation. Acta Biomater 86:350–62.
  • Maulvi FA, Desai DT, Shetty KH, et al. (2021). Advances and challenges in the nanoparticles-laden contact lenses for ocular drug delivery. Int J Pharm 608:121090.
  • Mehta P, Al-Kinani AA, Arshad MS, et al. (2019). Engineering and development of chitosan-based nanocoatings for ocular contact lenses. J Pharm Sci 108:1540–51.
  • Morgan SR, Pilia N, Hewitt M, et al. (2020). Controlled in vitro delivery of voriconazole and diclofenac to the cornea using contact lenses for the treatment of Acanthamoeba keratitis. Int J Pharm 579:119102.
  • Mun J, Kim TY, Myung D, et al. (2022). Smart contact lens containing hyaluronate-rose bengal conjugate for biophotonic myopia vision correction. Biomater Sci 10:4997–5005.
  • Mutlu Z, Shams Es-Haghi S, Cakmak M. (2019). Recent trends in advanced contact lenses. Adv Healthc Mater 8:e1801390.
  • Nahum Y, Israeli R, Mircus G, et al. (2019). Antibacterial and physical properties of a novel sonochemical-assisted Zn-CuO contact lens nanocoating. Graefes Arch Clin Exp Ophthalmol 257:95–100.
  • Nasr FH, Khoee S, Dehghan MM, et al. (2016). Preparation and evaluation of contact lenses embedded with polycaprolactone-based nanoparticles for ocular drug delivery. Biomacromolecules 17:485–95.
  • Niu L, Liu X, Ma Z, et al. (2020). Fungal keratitis: pathogenesis, diagnosis and prevention. Microb Pathog 138:103802.
  • Omranipour HM, Sajadi Tabassi SA, Kowsari R, et al. (2015). Brimonidine imprinted hydrogels and evaluation of their binding and releasing properties as new ocular drug delivery systems. Curr Drug Deliv 12:717–25.
  • Palazzo M, Vizzarri F, Ondruška L, et al. (2020). Corneal UV protective effects of a topical antioxidant formulation: a pilot study on in vivo rabbits. IJMS 21:5426.
  • Paradiso P, Serro AP, Saramago B, et al. (2016). Controlled release of antibiotics from vitamin E-loaded silicone-hydrogel contact lenses. J Pharm Sci 105:1164–72.
  • Patel A, Cholkar K, Agrahari V, et al. (2013). Ocular drug delivery systems: an overview. World J Pharmacol 2:47–64.
  • Perez VL, Wirostko B, Korenfeld M, et al. (2020). Ophthalmic drug delivery using iontophoresis: recent clinical applications. J Ocul Pharmacol Ther 36:75–87.
  • Phan CM, Subbaraman LN, Jones L. (2014a). In vitro drug release of natamycin from β-cyclodextrin and 2-hydroxypropyl-β-cyclodextrin-functionalized contact lens materials. J Biomater Sci Polym Ed 25:1907–19.
  • Phan CM, Subbaraman L, Jones L. (2014b). Contact lenses for antifungal ocular drug delivery: a review. Expert Opin Drug Deliv 11:537–46.
  • Pillai SKR, Reghu S, Vikhe Y, et al. (2020). Novel antimicrobial coating on silicone contact lens using glycidyl methacrylate and polyethyleneimine based polymers. Macromol Rapid Commun 41:e2000175.
  • Polat HK, Kurt N, Aytekin E, et al. (2022). Novel drug delivery systems to improve the treatment of keratitis. J Ocul Pharmacol Ther 38:376–95.
  • Polania-Baron EJ, Santana-Cruz O, Lichtinger A, et al. (2021). Treatment of severe infectious keratitis with scleral contact lenses as a reservoir of moxifloxacin 0.5. Cornea 40:831–6.
  • Prakash M, Dhesingh RS. (2017). Nanoparticle modified drug loaded biodegradable polymeric contact lenses for sustainable ocular drug delivery. Curr Drug Deliv 14:555–65.
  • Qin G, Zhu Z, Li S, et al. (2017). Development of ciprofloxacin-loaded contact lenses using fluorous chemistry. Biomaterials 124:55–64.
  • Rad MS, Sajadi Tabassi SA, Moghadam MH, et al. (2016). Controlled release of betamethasone from vitamin E-loaded silicone-based soft contact lenses. Pharm Dev Technol 21:894–9.
  • Rad MS, Mohajeri SA. (2017). Extended ciprofloxacin release using Vitamin E diffusion barrier from commercial silicone-based soft contact lenses. Eye Contact Lens 43:103–9.
  • Rodrigues FSC, Campos A, Martins J, et al. (2020). Emerging trends in nanomedicine for improving ocular drug delivery: light-responsive nanoparticles, mesoporous silica nanoparticles, and contact lenses. ACS Biomater Sci Eng 6:6587–97.
  • Roostaei N, Hamidi SM. (2022). Two-dimensional biocompatible plasmonic contact lenses for color blindness correction. Sci Rep 12:2037.
  • Ross AE, Bengani LC, Tulsan R, et al. (2019). Topical sustained drug delivery to the retina with a drug-eluting contact lens. Biomaterials 217:119285.
  • Saad S, Saad R, Jouve L, et al. (2020). Corneal crosslinking in keratoconus management. J Fr Ophtalmol 43:1078–95.
  • Salih AE, Elsherif M, Alam F, et al. (2021). Gold nanocomposite contact lenses for color blindness management. ACS Nano 15:4870–80.
  • Sandri G, Bonferoni MC, Rossi S, et al. (2016). Platelet lysate and chondroitin sulfate loaded contact lenses to heal corneal lesions. Int J Pharm 509:188–96.
  • Santodomingo-Rubido J, Carracedo G, Suzaki A, et al. (2022). Keratoconus: an updated review. Cont Lens Anterior Eye 45:101559.
  • Seewoodhary M. (2021). An overview of diabetic retinopathy and other ocular complications of diabetes mellitus. Nurs Stand 36:71–6.
  • Sekar P, Chauhan A. (2019). Effect of vitamin-E integration on delivery of prostaglandin analogs from therapeutic lenses. J Colloid Interface Sci 539:457–67.
  • Sekar P, Dixon PJ, Chauhan A. (2019). Pigmented contact lenses for managing ocular disorders. Int J Pharm 555:184–97.
  • Sercombe L, Veerati T, Moheimani F, et al. (2015). Advances and challenges of liposome assisted drug delivery. Front Pharmacol 6:286.
  • Sève P, Cacoub P, Bodaghi B, et al. (2017). Uveitis: diagnostic work-up. A literature review and recommendations from an expert committee. Autoimmun Rev 16:1254–64.
  • Shetty R, D’Souza S, Khamar P, et al. (2020). Biochemical markers and alterations in keratoconus. Asia Pac J Ophthalmol (Phila) 9:533–40.
  • Shi Y, Lv H, Fu Y, et al. (2013). Preparation and characterization of a hydrogel carrier to deliver gatifloxacin and its application as a therapeutic contact lens for bacterial keratitis therapy. Biomed Mater 8:055007.
  • Silva D, de Sousa HC, Gil MH, et al. (2020). Diclofenac sustained release from sterilised soft contact lens materials using an optimised layer-by-layer coating. Int J Pharm 585:119506.
  • Singh M, Bharadwaj S, Lee KE, et al. (2020). Therapeutic nanoemulsions in ophthalmic drug administration: concept in formulations and characterization techniques for ocular drug delivery. J Control Release 328:895–916.
  • Soluri A, Hui A, Jones L. (2012). Delivery of ketotifen fumarate by commercial contact lens materials. Optom Vis Sci 89:1140–9.
  • Stevens GA, White RA, Flaxman SR, et al. (2013). Global prevalence of vision impairment and blindness: magnitude and temporal trends, 1990–2010. Ophthalmology 120:2377–84.
  • Stone JL, Robin AL, Novack GD, et al. (2009). An objective evaluation of eyedrop instillation in patients with glaucoma. Arch Ophthalmol 127:732–6.
  • Sy A, Srinivasan M, Mascarenhas J, et al. (2012). Pseudomonas aeruginosa keratitis: outcomes and response to corticosteroid treatment. Invest Ophthalmol Vis Sci 53:267–72.
  • Tanito M. (2021). Reported evidence of vitamin E protection against cataract and glaucoma. Free Radic Biol Med 177:100–19.
  • Toda R, Kawazu K, Oyabu M, et al. (2011). Comparison of drug permeabilities across the blood-retinal barrier, blood-aqueous humor barrier, and blood-brain barrier. J Pharm Sci 100:3904–11.
  • Torres-Luna C, Hu N, Tammareddy T, et al. (2019a). Extended delivery of non-steroidal anti-inflammatory drugs through contact lenses loaded with vitamin E and cationic surfactants. Cont Lens Anterior Eye 42:546–52.
  • Torres-Luna C, Hu N, Koolivand A, et al. (2019b). Effect of a cationic surfactant on microemulsion globules and drug release from hydrogel contact lenses. Pharmaceutics 11:262.
  • Tieppo A, White CJ, Paine AC, et al. (2012). Sustained in vivo release from imprinted therapeutic contact lenses. J Control Release 157:391–7.
  • Tuft S, Somerville TF, Li JO, et al. (2022). Bacterial keratitis: identifying the areas of clinical uncertainty. Prog Retin Eye Res 89:101031.
  • Ung L, Chodosh J. (2021). Foundational concepts in the biology of bacterial keratitis. Exp Eye Res 209:108647.
  • Varela-Garcia A, Gomez-Amoza JL, Concheiro A, et al. (2020). Imprinted contact lenses for ocular administration of antiviral drugs. Polymers 12:2026.
  • Vazirani J, Wurity S, Ali MH. (2015). Multidrug-resistant pseudomonas aeruginosa keratitis: risk factors, clinical characteristics, and outcomes. Ophthalmology 122:2110–4.
  • Wang B, Liu H, Zhang B, et al. (2016). Development of antibacterial and high light transmittance bulk materials: incorporation and sustained release of hydrophobic or hydrophilic antibiotics. Colloids Surf B Biointerfaces 141:483–90.
  • Wei N, Xu X, Huang C, et al. (2022). Retraction note: hyaluronic acid-pluronic®F127-laden soft contact lenses for corneal epithelial healing: in vitro and in vivo studies. AAPS Pharm Sci Tech 23:242.
  • White CJ, Byrne ME. (2010). Molecularly imprinted therapeutic contact lenses. Expert Opin Drug Deliv 7:765–80.
  • White CJ, McBride MK, Pate KM, et al. (2011). Extended release of high molecular weight hydroxypropyl methylcellulose from molecularly imprinted, extended wear silicone hydrogel contact lenses. Biomaterials 32:5698–705.
  • Wong A, Fallon M, Celiksoy V, et al. (2022). A Composite system based upon hydroxypropyl cyclodextrins and soft hydrogel contact lenses for the delivery of therapeutic doses of econazole to the cornea, in vitro. Pharmaceutics 14:1631.
  • Wu C, Or PW, Chong JIT, et al. (2021a). Extended delivery of pirfenidone with novel, soft contact lenses in vitro and in vivo. J Ocul Pharmacol Ther 37:75–83.
  • Wu C, Or PW, Chong JIT, et al. (2021b). Controllable release of pirfenidone by polyvinyl alcohol film embedded soft contact lenses in vitro and in vivo. Drug Deliv 28:634–41.
  • Wu Q, Yang C, Chen W, et al. (2022). Wireless-powered electrical bandage contact lens for facilitating corneal wound healing. Adv Sci (Weinh) 9:e2202506.
  • Xiao A, Dhand C, Leung CM, et al. (2018). Strategies to design antimicrobial contact lenses and contact lens cases. J Mater Chem B 6:2171–86.
  • Xu X, Awwad S, Diaz-Gomez L, et al. (2021). 3D printed punctal plugs for controlled ocular drug delivery. Pharmaceutics 13:1421.
  • Yañez F, Martikainen L, Braga ME, et al. (2011). Supercritical fluid-assisted preparation of imprinted contact lenses for drug delivery. Acta Biomater 7:1019–30.
  • Yang M, Yang Y, Lei M, et al. (2016). Experimental studies on soft contact lenses for controlled ocular delivery of pirfinedone: in vitro and in vivo. Drug Deliv 23:3538–43.
  • Yan F, Liu Y, Han S, et al. (2020). Bimatoprost imprinted silicone contact lens to treat glaucoma. AAPS Pharm Sci Tech 21:63.
  • Yeh SB, Chen CS, Chen WY, et al. (2014). Modification of silicone elastomer with zwitterionic silane for durable antifouling properties. Langmuir 30:11386–93.
  • Yellepeddi VK, Sheshala R, McMillan H, et al. (2015). Punctal plug: a medical device to treat dry eye syndrome and for sustained drug delivery to the eye. Drug Discov Today 20:884–9.
  • Yin C, Qi X, Wu J, et al. (2021). Therapeutic contact lenses fabricated by hyaluronic acid and silver incorporated bovine serum albumin porous films for the treatment of alkali-burned corneal wound. Int J Biol Macromol 184:713–20.
  • Zha XJ, Zhang ST, Pu JH, et al. (2020). Nanofibrillar poly(vinyl alcohol) ionic organohydrogels for smart contact lens and human-interactive sensing. ACS Appl Mater Interfaces 12:23514–22.
  • Zhang X, Cao X, Qi P. (2020). Therapeutic contact lenses for ophthalmic drug delivery: major challenges. J Biomater Sci Polym Ed 31:549–60.
  • Zhao L, Wang H, Feng C, et al. (2021a). Preparation and evaluation of starch hydrogel/contact Lens composites as epigallocatechin gallate delivery systems for inhibition of bacterial adhesion. Front Bioeng Biotechnol 9:759303.
  • Zhao L, Qi X, Cai T, et al. (2021b). Gelatin hydrogel/contact lens composites as rutin delivery systems for promoting corneal wound healing. Drug Deliv 28:1951–61.
  • Zhong H, Sun G, Lin X, et al. (2011). Evaluation of pirfenidone as a new postoperative antiscarring agent in experimental glaucoma surgery. Invest Ophthalmol Vis Sci 52:3136–42.
  • Zhu Q, Cheng H, Huo Y, et al. (2018). Sustained ophthalmic delivery of highly soluble drug using pH-triggered inner layer-embedded contact lens. Int J Pharm 544:100–11.