Open access peer-reviewed chapter

Ocular Drug Delivery System: Barrier for Drug Permeation, Method to Overcome Barrier

Written By

Prakriti Diwan, Rajendra Jangde, Sulekha Khunte, Harish Bhardwaj and Preeti K. Suresh

Submitted: 15 February 2022 Reviewed: 13 May 2022 Published: 07 October 2022

DOI: 10.5772/intechopen.105401

From the Edited Volume

Drug Development Life Cycle

Edited by Juber Akhtar, Badruddeen, Mohammad Ahmad and Mohammad Irfan Khan

Chapter metrics overview

192 Chapter Downloads

View Full Metrics

Abstract

The physiological and anatomical barriers are major obstacles in the field of ocular drug delivery systems. The barriers involve nasolacrimal drainage, blinking, cornea, sclera, and blood-aqueous barriers whereas dynamic barriers involve conjunctival blood flow, lymphatic clearance and tear drainage. These barriers are showing natural protective functions, as well as limiting drug entry into the eye. Nanocarriers have been found to be effective at overcoming the problems and associated with conventional ophthalmic dosage forms. In this chapter emphasizes overcome to barriers and discusses advanced novel techniques used in the field of ocular drug delivery systems including nano dispersion systems, nanomicelles, lipidic nanocarriers, polymeric nanoparticles, liposomes, noisome, and dendrimer, have been investigated for improved permeation and effective targeted drug delivery to various ophthalmic site.

Keywords

  • ocular drug delivery
  • inflammation
  • conjunctiva
  • microphages
  • aqueous humor

1. Introduction

The eyes are one of the most important and complex sensory organs; they serve as a gateway for collecting and transmitting extraneous images to the brain as signals via the optic nerve. They maintain a relationship between the body and our surroundings through this action. Various disorders, such as inflammations or bacterial and viral infections, affect the eye’s behavior. Such kind of disorders affecting the anterior eye tissues can be smoothly treated with high doses of drugs. However, diseases affecting the posterior tissue of the eye are tough to teach and treat. Age-related disorders including macular decay, glaucoma, diabetic macular edema, and proliferative vitreoretinopathies are some of familiar posterior eye diseases that can lead to vision loss if left untreated.

The complex structures of the eyes provide a high level of drug/treatment resistance. A thorough understanding of ocular anatomy, physiology, and barriers is essential for providing effective treatment for diseases affecting both anterior and posterior ocular tissues. This assists with clarifying the difficulties related with drug conveyance to eye. In this part, we give a point by point depiction of visual life systems and physiology as well as the blockade those stances difficulties to sedate conveyance.

Advertisement

2. Structure of the eye

The eye can be partitioned into two divisions the front fragment and the back section in structure of eye (Figure 1). The front fragment stays alive of the cornea, conjunctiva, watery humor, iris, ciliary body, and translucent focal point. These elaborate roughly 33% of the front of the eye. The leftover part for example back section made out of the sclera, choroid, Bruch’s layer, retinal shade epithelium (RPE), nonpartisan retina, and glassy humor. An exhaustive depiction of the life systems and physiology of the eye is introduced beneath.

Figure 1.

Structure of human eye.

2.1 Cornea

The cornea is delicate, straight, smooth, vascularized, generally deciduous, and the most meticulous tissue in the body. The curved and rounded design allows it to be inserted directly into the external environment. The cornea develops in the white part of the eye called the sclera and the translucent tissue called the conjunctiva. The limitation of the cornea from which it connects to the sclera is known as the annulus. Limbus is strangely angiogenic and is thought to be a reservoir of pluri potent juvenile microorganisms. The surface of the cornea, which is not covered by the external environment, is invaded by the tear film, and its inner surface is mainly in contact with liquid called liquid humor. The thickness of the cornea often increases from the center to the periphery. These are seen in the twist of the cornea, which is huge in the center and has the smallest ring.

Corneal shape, surface perfection, straightforwardness, and refractive list manage the optical properties of the cornea. The corneal stroma is fixed with an almost homogeneous and methodical course of action of collagen fitters (distance across 25, 35 nm). Such a course of action is wanted to be answerable for forestalling and canceling dissipated light obstruction from the occurrence beam of light on collagen. The unequivocal plan and capacity of strands permit the light beams to ignore the cornea with next to no impedance. Corneal perfection is constrained by the corneal epithelium and tear film. Any redirection from the typical engineering of corneal collagen strands and nonappearance of tear movie inclusion causes dry eye and dissipating of episode light beams and prompts loss of corneal forms, straightforwardness, and perfection.

As mentioned above, the cornea is deeply attached to soft spots that are 300,400 times thicker than the skin [1]. Tactile nerves, long ciliary nerves, and well-thought-out involuntary nerve cords persuade the panniculus tissue. The ciliary nerve of the trigeminal nerve of the eye provides the tactile nerve to the membrane. The marginal nerve ring is formed by the longciliary nerve, which sheds its myelin sheath (at certain short distances) after entering the cornea, infiltrates the Bowman layer and then annihilate it with vane cells. These nerve cords further radiate anteriorly cross into the stroma, building the sub epithelial plexus [2]. Damage or loss of the corneal epithelium reveals external climate-sensitive areas. Causes extreme visual distress [3]. The typical cornea has no venous supply. Therefore, this tissue is treated as one of the internal tissues in the body along with ligaments and focal points. Internally, corneal epithelial cells and endothelial cells are metabolically dynamic and are effectively involved in injury healing. Both cell layers receive blood parts and various necessities from the veins of the internal and external carotid corridors that make up the conduit around the marginal cornea [4, 5]. Aqueous humor provides the cornea with the glucose it needs and a limited amount of oxygen. Most of the oxygenation of the cornea is evident in the opening to the air where the oxygen taken up by the lacrimal layer diffuses into the corneal epithelial cells. This openness of the saturated tear layer on the surface of the cornea is essential for oxygenation and maintenance of integrity and virtue. The histological section shows that the cornea is composed of six different layers, specifically the corneal epithelium, Bowman’s layer, stromal, DNA layer, Descemet’s membrane, and endothelium. The corneal epithelium is composed of 5–6 layers of discrete squamous epithelial cells that are not cornealized. The various epithelial layers of the cornea are composed of several layers of flat cells and wing cells and a single layer of basal cells. Various corneal epithelial cells are formed by rectangular parallelepiped basal cells with a tight cross-sectional structure that prevents tears from entering the intercellular space. After separation, these cells become delicately smooth as they migrate to the surface of the cornea. Electron microscopy shows that the outer layer of flat epithelial cells is sporadic and that rim-like disruption of the cell membrane is shown as microplica (Figure 2) [6]. This collapse expands the contact area. Microprica is covered with a very fine, tightly packed, charged sugar-coated layer that aids in scattering.

Figure 2.

Histology of corneal layer of the eye.

Figure 3.

Diagrammatic portrayal of front office of natural eye with shut eye covers showing three particular conjunctivas (striking layers). (1) Forniceal conjunctiva, (2) bulbar conjunctiva, (3) palpebral conjunctiva, (4) upper eye top, (5) watery humor, (6) lower eye lid, (7) cornea, (8) sclera and (9) lacrimal gland,(10) translucent focal point and (11) iris ciliary body.

Figure 4.

Flow direction of aqueous humor from iris to canal of Schlemm or towards cornea.

The fluid spreads evenly over the entire corneal surface at each strabismus. Flatepithelial cells are revived every 710 days by pluri potent microorganisms in the cornea llimbus [7]. More established cells are shed after their tight interface is destroyed by the tear film. The new cell layer controls tight junctions with the help of vane cells, which form lateral junctions between cells [8, 9]. Epithelial cells are connected to a 0.05 atm thick basement membrane consisting of columnar epithelium and are approximately 20 cm high. The presence of tight cells overlaps between the cell and the framework structure, and they are deeply impervious. Basal cells are the major corneal epithelial cells suitable for mitosis [10]. The Bowman’s layer is a band of undefined fibrous material with a thickness of 81 Skm that simply resides beneath the base of corneal epithelial cells. This layer is non-regenerative, forming a boundary between the stromaof the cornea and epithelial cells and maintaining the shape of the cornea. It has been hypothesized that it is associated with re-epithelialization [11]. The Bowman layer is composed of short type I collagen fibrils embedded in the proteoglycan lattice. The stroma of the cornea intervenes between the Bowman layer and the Descemet’s membrane and occupies 95% of the thickness of the cornea. The stroma contains corneal keratocytes, fibroblasts, brain tissue, and Schwann cells. Most of the interstitium is composed of collagen fibrils, mainly type I collagen [12]. These fibrils extend from one end of the annulus to the next and are calculated symmetrically at 90° in the anterior interstitium and symmetrically in the posterior interstitium. Collagen fibrils maintain the cornea with mechanical strength. The posterior interstitial fibrils are more tightly coordinated than the anterior, which clearly contributes to the mechanical and bipodal strength of the cornea [13]. The Dua’s layer is a highly detailed, cell-free solid layer that resides on the Descemet’s membrane [14]. Its physiological function in the cornea has not yet been investigated. The Descemet’s membrane layer is 10 pm thick, the surface is undefined and huge. Upon invasion, this layer is alternative, homogeneous, cell-free and gradually transforms into Descemet’s membrane [15, 16]. This layer is thrown into many creases that appear as stripes, because of awry enlarging of the back stroma, and underlying limitations name by the limbus.

The corneal endothelium is the most profound monolayers of cells, which is non-mitotic and in direct association with fluid humor, while endothelium control itself, stroma leftover in a deliquescent state to give corneal clearness [17]. This monolayer has characterized penetrability to particle transition, which is important to construct an osmotic angle. Endothelial cell numbers by and large downfall with age. During sickness, cells help in size (polymegathism) and show shape variety (polymorphism) to make up for the spaces made by declining cells.

2.2 Conjunctiva

Histologically, conjunctiva is primarily comprised shallow multifaceted epithelium and a fundamental stroma [18]. Conjunctival epithelial cells holds less close intercellular intersections with transepitlielial electric obstruction in the scope of 0.7S to −1.5 Kf2cm’ [19, 20]. Conjunctiva moreover contains pores of 5.5 nm sweep [21]. Implanted inside the conjunctival epithelium are challis cells, mucous organs, organs of Manz and the sepulchers of Henle [22]. Challis cells are matched in electrolyte, liquid and bodily fluid discharge [23] to plan the tear film. Transportation of challis cells in various areas of conjunctiva persue a particular request: mediocre > predominant > nasal > fleeting conjunctiva [24]. The apical surface of conjunctiva shows out foldings called microvilli and microplicae [25]. During tear film part emission the conjunctival apical layer cells are breakdown alongside secretory granules and different parts into the tear film. Conjunctival apical out foldings might advance in expanding surface region, offer help or balance out and attach with tear film [25].

Figure 5.

Histology of lens.

The conjunctiva is a thin, highly vascularized, opaque, mucous-draining tissue that covers the inner layers of the upper and lower eyelids [26]. It reflects off the eye as a faint, transparent tissue on the sclera and extends toward the annulus of the cornea. This tissue is very fast with efferent, afferent, tangible nerves, and also lymphoid tissue. The total surface area of the conjunctiva is many times the total surface area of the cornea [27, 28]. Due to its versatile nature, the conjunctiva promotes eye and eyelid movement. The lamina propria of tears combined with a small amount of aqueous humor protects the internal visual tissue from the external climate. Depending on the area, thickness, and angiogenesis, this tissue can be broadly divided into three types: eyelid, eyelid, and hamburger conjunctiva. The upper and lower eyelids are lined inside by a medium palpebral conjunctiva, with a small portion of the conjunctiva called the bulbous conjunctiva adjacent to the sclera. The bulbar conjunctiva is very light, standard on the cornea, and has open eyes with an air interface and is exposed to the outside climate (Figure 3). The eyelids and parts of the eyeball are connected by a small piece of tissue called the vestibular conjunctiva. The vestibular and volar conjunctiva are rich in veins and composed of heterogeneous tissue [26].

Figure 6.

Layers and histology of normal adult human retina.

Figure 7.

Diagrammatic representations of rod and cone cells of neural retina.

Conjunctival stroma is parleyed between the foremost epithelium and back sclera. This layer is equiped with blood and lymph vessels and innervated with sensitive spots. This is interlaced with mature lymphocytes, overwhelmingly with T cells comparative with B cells. Mucosa-connected lymphoid tissue assumes a urgent part in the resistant reaction [29]. It is made out of lymphatic cells present underneath the epithelium, which achieve a safe reaction against antigens by emitting dissolvable antibodies. Lymphocytes are circled over particular vascular framework alluded high endothelial venules [30, 31, 32]. Electron micrograph of the tear film surface of the conjunctival epithelium. It shows (a) the mucous layer (m) and (b) the sugar coating (studs) present in the micropreca. (A) In this movie, we used various mucins, MUC1 (red), MUC4 (blue) and MUC1 (green). These protrude as solid bars to form the sugar coating from the microvilli of the epithelial cells. (B) The higher emphasis in the area (a) indicates fine fibers extending into the sugar coating (sharp stones), probably film-related mucin atoms. In the cytoplasm of microplica, actin fibers extend to the surface film, from which layered mucin emerges (giant bolts). (C) Small immunoelectron confinement of the immune agent H185 that senses carbohydrate epitopes on the layer-related mucin MUC16 [25].

2.3 Fluid humor

Watery humor is an optically clear, gently basic visual liquid that is developing from plasma by epithelial cells of ciliary body [33]. Three distinct cycles dissemination, ultra filtration and dynamic emission, commit to the compound organization and arrangement of fluid humor. It is anticipated that the whole watery humor is supplanted in roughly 100 min [34]. This liquid comprises somewhat less protein, egg whites and y-globulins, than plasma. Moreover, glucose, lactic corrosive, ascorbic corrosive and immunoglobulin G are similarly present [35, 36]. Fluid humor supplies supplements and an oxygen to the visual a vascular tissue, alluded as cornea and focal point. It clean up squanders items, macrophages, blood and other trash from the back of the cornea and front of the focal point. Additionally, it assumes a vital part in keeping up with the shape and interior afflictions of the eyeball over creation of intraocular pressure. The liquid humor that is assembled and drained into the posterior eye ignores the pupil in the anterior chamber. It is drained into the venous bloodstream through the trabecular meshwork and Schlemm’s canal (Figure 4). Approximately 510% of the aqueous humor is used to secure the uveal scleral pathway [37, 38].

2.4 Iris—ciliary body

The iris and the ciliary body are two distinct tissues with distinct physical and physiological properties. Their life systems and physiology are explained combined for simple mindfulness and close physical limits. The iris is placed at the back of the cornea and appears as a ciliary body spirit. The iris is made up of three layers: endothelium, stroma, and epithelium, according to histology. The iris creates a small roundabout opening or gap in the bleeding edge of the focus point, known as the student, which helps to control the amount of light that reaches the retina. Each ciliary body has a ciliary cycle, which conveys a fibrovascular core that is in continual contact with the ciliary body’s stroma. Blood flow from the choroidal veins in the front to the back. The blood from the ciliary body of the eye is drained from the border of the vortex vein. The ciliary body is physically adjacent to the iris and is in charge of adjusting three vital capacities in the eye: it secretes watery humour, which runs before the focal point and is depleted out of the eye through tubules known as the trabecular meshwork and schlemm channel close to the intersection of cornea and iris (ii) this tissue also contains smooth muscles, which demonstrate through zonular filaments on the glasslike focal point to acclimate centigrade (iii) it can coordinate in emptying fluid humor out of the eye into the bordering trabecular meshwork by broadening smooth muscle strands and ligaments.

2.5 Focal point

The focal point is clear, internal, non-innervated and biconvex. It is situated behind the student and iris with the help of the ciliary body’s zonular strands [39]. The foremost focal point is covered with fluid humor and the back with glassy humor. The focal point film (otherwise called the case) keeps up with latent trade of metabolic substrates and waste through straightforward dissemination [40] checked by their size and charge [41, 42, 43, 44]. The focal point comprises of four explicit parts: the case, epithelium, cortex (fiber cell mass) and core. Additionally, it controls light passage into the eye and its refraction.

The case is an undisturbed, uncomplicated, versatile foundation layer that embodies the entire focus and provides the framework underlying the focus of the eye. Container thickness varies between species and within similar assemblies. For example, in mice, rodents, and rabbits, the container thickness is approximately 10 μm, 13 μm, and 14 μm, respectively. The thickness of the human front shell ranges from 25 to 30 μm compared to 14:00 on the back [45, 46]. A layer of casing protects the focus from direct contact with adjacent visual tissue and aqueous fluids. In addition, it acts as a sea of developmental factors, sequestering them [47, 48, 49, 50] and forming a defensive barrier against microbial attack [51, 52, 53]. The arrival of developing factors from the container supports the improvement and isolation of focal cells. The instrument used to give the focal point its shape and surface curvature is fluffy. A tall, columnar monolayer of epithelium lies beneath the foremost focal vessel. The epithelium is not present behind the focal point in contact with the vitreous liquid. The cortex lies below the vessel and later in the epithelium of focus. It contains 68.6 of water [54]. The focal cortex is composed of newly assembled filaments that make up most of the focal point. The strands are tightly packed, the newly assembled filaments make up the nucleated organelles [2, 55, 56], and with age the strands shift towards the focal point (Figure 5) [46]. During this development period, filaments lose organelles and nuclei, providing focus directness. The preparation of these filaments in adult focus involves four pointed stellar programs [57]. Entanglement between adjacent fiber cells can cause interlocks in deeper focal strands [57, 58]. Further migration to the focal cortex near the nucleus, the cells appear in the form of edgy hexagons. The locking system works together to even out the pressure during operation, perhaps preventing the fiber cells from slipping against each other.

The focal point core is exceptionally gotten by its area. It contains 63.4% water [54] and is started by testimony of old fiber cells that movement to the middle from the fringe. As the consequence of gathering of old cells in this zone, it turns out to be exceptionally thick and denser.

2.6 Sclera

The sclera, commonly referred to as the white of the eye, is a highly vascularized and flexible tissue beneath the conjunctiva and dilated cornea. The optic nerve exits the eye posteriorly through a scleral opening surrounded by a complex, grid-like, filamentous tissue network called the cribriform plate. The sclera (the top layer of the sclera) provides the expected nutrition to the sclera. The sclera occupies about 80% of the skin of the eye and the rest is composed of the inner cornea. Its physical area, the anterior sclera (near the annulus), thickens as it moves towards the equator, near and around the optic nerve, towards the back of the eye. It will decrease [59]. The sclera is composed of a sloppy tissue of filaments emanating from the dura mater of the focal sensory system. In this type of plan, all visible light frequencies are scattered and appear dazzlingly white in the shadows. At the interface between the sclera and the cornea, changes to true standard changes occur sporadically from nowhere, leading directly to such dark white scleral changes.

Similar collagen strands are available in the cornea yet are coordinated in a standard example that provisions straightforwardness to the tissue, hydrated sclera filaments stay obscure while corneal strands do not require water and become straightforward. The corneal endothelium serves to its straightforwardness by the droping out of water. Sclera, being the layer of the eyeball, is presenting to visit changes of outer alongside of intraocular pressure. Width of the sclera at tropical zone goes from 25 to 230 nm and at the fluid measurement of the sclera goes from 20 to 80 nm [60, 61].

2.7 Choroid

Choroid is put between fringe sclera and internal retinal epithelium. It is famously vascularized and innervated tissue fixing melanocytes alongside bodily fluid like extracellular liquid. It comprises of four particular parts: from external to internal suprachoroid, haller’s layer, sattler’s layer and Bruch’s film. Suprachoroid is made by 6–10 layers, around 30 qm in which lay out the connection point between external sclera and internal [62]. The suprachoroid supported anteriorly with supraciliary and last posteriorly up to the optic nerve. This locale is exceptionally aroused with nerve strands and ganglion yet no vasculature. Meager lamellar filaments, in touch to one another, related the choroid and sclera. This procedure develops a small space between these tissues called the space on or around the choroid. This space is scarce as it moves backwards towards the macula.

The vascular layer below the supra choroid lamina is composed of three specific vascular layers, which are narrow and the distance within the lumen is continuously reduced. These blood vessels are surrounded by pigmented melanocytes and non-pigmented fibroblasts, reaching the maximum amount of choroid. Blood vessels, fibroblasts, and melanocytes appear in discreet amounts of choroidal stroma. The thickness of melanocytes increases from concentration to the periphery. Choroidal vessels are suggested by the luminal distance across the site as follows: Haller layer with larger outer blood vessels. Currently being tracked on a medium-sized vessel at Sattler’s Layer. Well-traced choroid capillaries with narrowed blood vessels. Choroidal blood flow are also high in other visual tissues and the brain [63, 64]. The enlarged choroidal blood circulation provides an additional supply and diffusion of oxygen within the retina of the brain. Metabolic waste from the retina is carried away by changes in intraocular temperature caused by the visual cycle. This accelerated circulatory system also seems to predict sections when monitoring intraocular pressure [65, 66, 67]. Bruch’s film is the last and deepest choroidal layer that covers the RPE [68]. Also known as the vitreous membrane. It is a thin, pentahedral, multifaceted, cell-free, layered structure initiated by the interaction of villous capillaries with RPE. Bruch’s film grows from behind the eye, such as the optic nerve, to the oraserrata of the iris, with a continuous decrease in thickness from behind the eye to the edge. This film separates RPE and villous capillaries. Retinal cell associations include seven unique types of cells, including RPE cells. These are photoreceptor cells (poles and cones), squamous cells, amacrine cells, interstitial cells, bipolar cells, ganglion cells and glial cells (Figure 6).

2.8 Retinal pigment epithelium

Each individual eye contains approximately 3.5 million RPE cells [69], which attach to each other to form a narrow intersection (closed zone). The retina epithelium (RPE) contains non-dividing cells that form the monolayer that covers the retina of the cerebrum, but these cells can proliferate under masochistic conditions and are therefore isolated. I have not. It provides confirmation to the internal visual tissue and secretes large amounts of growth factors (vascular endothelial enhancer factor, ciliaryneurotrophin factor, and platelet engineering enhancer factor). This monolayer monitors visual resistance and protects against oxidative damage by releasing immune modulatory cytokines [70, 71]. RPE cells mediate several compounds such as superoxide dismutase, catalase, glutathione, and melanin clay. RPE supports the function of villous capillaries and photoreceptors and plays a fundamental role in patience. Therefore, its presence is important for the adjustment of visual boundaries [72]. Its capabilities include regulation of priotoceptors to external parts, retinoid processing, management of the visual cycle, and maintenance of sub retinal complex ecological variables [73, 74, 75].

2.9 Neural retina

Geologically, the retina is fashioned into macula, optic plate, fovea and fringe retina. The macula or are acentral is round 5 mm in thickness and positioned kind of three mm farfar from the optic circle. Macula accepts its call from the yellow carotenoid color ationxanthophylls, because the macula lutea. The focal factor of the macula serves a huge locale of visible sharpness [76] and is known as the fovea. The fovea has the maximum noteworthy thickness of confined and extended cone receptors to misrepresent mild location [77]. The focal factor of the fovea is vascular as much as 500 atm and the bloods deliver to this locale offers from chorio capillaries. Retinal fleeting veins encase the fovea. The outside layers of the fovea is thick and contain cores of photoreceptor cells. The relaxation fringe retina, bodily one layer of ganglionic cells, are to be had outside the fleeting retinal courses. Most outside retina receives its blood conveyances from the choroidal course, whilst standard retinal go with the drift satisfies inward retinal blood deliver [78, 79]. The inner arranging of the eyeball is made from mild-sensitive mind cells, alluded the mind retina, which talk tactile records to the cerebrum and interplay with the out of doors climate. These tactile nerves emerge from the focal sensory system [80, 81]. Brain retina is framed of round 7.7 million poles and five million cones (Figure 7) [82, 83]. The photoreceptor cells made from bars and cones. These cones basically ability to capture and extra de over the photons right into a nerve signal [84]. Retinal pole cells are accountable for keeping apart colorings in exquisite mild alien though cone cells has a tendency in keeping apart exceptionally contrasting shading in stupid mild. Biggest portions of cones are discovered with inside the fovea; whilst the bars are appropriated during the retina bar the focal fovea. The cones and bars are interconnected with among neurons named bipolar cells. Visual records is passing directly to ganglion cells through the a macrine cells, which cross approximately as an extension. Ganglionic cells talk records symptoms and symptoms to the focal sensory system, as an example mind. During power transmission Muller’s telephonesassist in directing the community microenvironment for visible working.

2.10 Vitreous humor

The glassy humor is a straightforward, dismal, coagulated mass that consume the space in the eye between the focal point and the retina. It is encased by a layer of collagen named the glassy film (or hyaloid film or glassy cortex) isolating it from the remainder of the eye. It makes up four-fifths of the volume of the eyeball. The glassy humor is liquid like close by the middle, and gel-like close by the edges.

The glassy humor is in touch with the glassy film overlying the retina. Collagen fibrils associate with the glassy at the optic nerve circle and the oraserrata, (where the retina closes anteriorly), at the Wieger-band (the dorsal side of the focal point). The glassy likewise unequivocally connects to the focal point case, retinal vessels, and the macula, the region of the retina which gives better detail and focal vision [85].

Thickness of the liquid shrinkage with developing age. As this occurs, foremost watery humor might penetrate into the back glassy, bringing about a pulling impact at the association point of retina and glassy liquid. Moreover, this might source the arrival of cells into the liquid, which show up as floaters, and if a huge pulling impact is created it might pull off or withdraw the retina [86].

2.11 Hindrances to visual medication conveyance

Ophthalmic medications are accessible as arrangements, suspensions and balms, which are fundamentally directed topically. Elective courses of medication conveyance to the eye are oral, intravenous, intra vitreal, sub conjunctival and peri ocular infusions or inserts [87]. A challenge task in drug conveyance is to defeated visual boundaries and convey sedates definitively to the designated visual tissue. Drugs are meddling from arriving at designated visual tissues by static and dynamic natural visual boundaries. These obstructions can be ordered by their physical area and their practical properties. Generally, these obstructions can be named foremost and back section boundaries.

Advertisement

3. Front portion hindrances

There are two assortments of boundaries, for example static or dynamic boundaries. Static boundaries cover cornea, conjunctiva, iris-ciliary body, focal point, and blood watery boundary and efflux siphons propose on cell surface, for example, multidrug-safe proteins: penetrability glycoprotein. Conjunctival lymph, blood stream, inverse directional progression of fluid humor and tear creation together cover under the powerful boundaries. These obstructions are shown exhaustively underneath.

3.1 Cornea

The life systems and physiology of the cornea have been delineated before in this section. Cornea go about as a multifaceted mechanical obstruction to keep away from exogenous substances, including topically applied drugs, from infiltrating further into visual tissues. Expanding size influences the conjunctival penetrability. Bulbar and palpebral do not show a huge contrast [88]. Penetrability correlation that the conjunctiva is two times as porous as the sclera, while is roughly multiple times more penetrable than cornea. The epithelial pore size and thickness are two and multiple times more noteworthy in cornea and sclera individually. An anticipated complete paracellular space in conjunctiva is multiple times higher than that of cornea. External corneal layer, with 90% epithelium cells, convey this profoundly lipophilic. Mature corneal epithelial cells go about as a district for drug ingestion since they are firmly limited by cell bond, for example, Occludin, ZO1 and ZO2 [89]. These proteins tenaciously seal cells and establish a tight convergence for paracellular drug transport across the cornea.

Drug transport across films by a variety of devices, specifically (I) paracells, (II) transcellular, (iii) dynamic, (iv) carrier-mediated, and (v) receptor-mediated transport. Paracellular scattering of ionic (polar) particles is hampered by the narrow convergence of the cornea, but lipophilic preparations can be beneficially diffused through the transcellular portion across the lipophilic cornea. Bowman’s shift does not have a significant impact on countering drugs with a wider distribution. Hydrophilic stroma is a rate-determining disorder of fat-soluble molecules, preventing more important visual media. In the case of a superbly lipophilic and almost hydrophilic combination, the epithelium and stroma act as a reservoir. Descemet’smembrane film is not involved in blocking drug intake on a regular basis. The corneal monolayer endothelium forms a cellular barrier between the stroma and the aqueous humor. This boundary mediates a flawed narrow intersection that promotes the free development of macromolecules between stroma and body fluids [90]. The distribution of drug intake through the cornea usually occurs in the aqueous humor. Subatomic size does not play an important role in transport through the corneal epithelium, but ionization of the mixture reduces transcellular dispersal. Therefore, particle physicochemical properties, ionization resistance, and visual medicine pH play important roles in maintaining and determining visual medicine porosity in the corneal epithelium 4. Vernal kerato conjunctivitis is a severely helpless conjunctivitis that impairs the dignity of the corneal epithelium and limits its prosperity. During this condition, eosinophils transport cytotoxic proteins such as myc-sponsored restricted protein (MBP1), eosinophil peroxidase, eosinophil-specific neurotoxin, and eosinophil-cationic proteins into the tears and the corneal epithelium. Reverse rotation of the epithelium [91, 92, 93, 94, 95].

Corneal ulcers are caused by the sensation of corneal fibroblasts by neutrophils. These fibroblasts are involved in the deterioration of nearby corneal collagen through various components such as disease putrefactive factors, interleukin 4, and interleukin 13 [96, 97, 98]. Therefore, the cornea loses an undeniable collagen strategy that can affect the corneal barrier properties against locally controlled drugs. Corneal fibroblasts are involved in the reverse rotation of collagen and can change parts of the basal layer to metastasize corneal ulcers. Tears from patients with vernal kerato conjunctivitis destroy type IV collagen and laminin, dominate part of the tornado protective layer, and accordingly lead to the development of corneal ulcers [99, 100, 101, 102]. Conjunctiva Conjunctival tissue continues as a permeability limit to topically applied drugs. One of the limit properties can be connected with its transepithelial electric protection [103]. Various courses of prescription osmosis, for instance, paracellular, transcellular, dynamic and endocytic courses accept a key part in visual medicine transport through conjunctiva for topically applied drug things. The paracellular course of medicine immersion is noticeably bound in view of the presence of tight crossing points at the epithelial surface, which is the rate limiting development for drug maintenance [104]. The presence of secretory organs presented inside the conjunctiva could have a shock on drug osmosis [105, 106]. Tear creation is a careful response of the eye considering topically applied xenobiotics. A limit reduces the prescription concentration and bioavailability at the conjunctival apical surface. Physicochemical properties of the meds, for instance, hydrophilicity and subnuclear weight were found to accept a fundamental part in drug infiltration across conjunctiva. Test results explained that hydrophilic prescriptions with under 20 kDa nuclear burdens are vulnerable comparing higher subnuclear weight drug particles. By and large, conjunctiva has more conspicuous paracellular permeability than the cornea for proteins (insulin, subnuclear weight 5.8 kDa) and peptides (pamino clonidine subnuclear weight 245.1 Da) through its greater surface locale and defective vasculature [107108]. On the other hand, lipophilic prescription ingestion through the transcellular course has unequivocally higher responsibility relative with hydrophilic meds. As the conjunctiva has esterase activity [109], it could go probably as another impediment for drug transport. During the wiped out conditions the conjunctiva could expand and cultivate fibrosis as consequence of high proportions of collagen creation.

3.2 Blood liquid limit

Endothelial cells of iris/ciliary veins and the non-pigmented ciliary epithelium together arrangement the blood liquid limit (BAB) in the front piece of the eye. This prevention shapes tight convergences at the cell level and stays aware of the exchanging of solutes between the preeminent and back visual region. Thus, it hinders some vague medication entrance into more profound visual tissues by functioning as a boundary. Cell tight intersections blocks vague medication passage into internal visual tissues and proceed as an obstruction. BAB assists with managing visual straightforwardness and keeps up with synthetic substances structure [110, 111]. An ideal BAB conveys deficient obstruction usefulness; for instance, 40 kDa horseradish peroxidase arrives at watery humor through fenestrated ciliary vessels, which are associated with controlling penetration of plasma proteins, into the fluid humor. The vast majority of the medications arriving at watery humor are clear out into the fundamental course by means of the iris blood vessels. In this way, drugs in fluid humor are consumed by the iris colors and crash into the iris blood dissemination [112, 113]. Then again, little lipophilic medication atoms that withdraw tight intersections and cross BAB can be disposed of more quickly than the bigger hydrophilic particles. Accordingly, drug infiltration to more profound visual tissues is halted.

3.3 Efflux pump

Multidrug obstruction in cells might be to some degree created because of cell layer joined with efflux carrier proteins. These efflux siphons live from the adenosine triphosphate-restricting tape (ABC) superfamily and are effectively taken an interest in effluxing exogenous xenobiotics and cater insurance to the cell. Penetrability glycoprotein (P-gp) and multidrug obstruction protein (MRP) are two efflux proteins that are key members in drug efflux at the cell level, initiated a decreased intracellular medication fixation.

Advertisement

4. Front section dynamic boundaries

4.1 Tear seepage

Ophthalmic medication items are fundamentally managed by skin course into the visual circular drive. A huge segment (around 90%) of regulated drug is lost as the aftereffect of a precorneal obstruction—attack nasolacrimal conduits. Likewise, different variables that required into diminish drug focuses in the parkway incorporate tear weakening, drug restricting to tear proteins and sped up freedom. Ordinarily, the advertised droppers are acclimated to convey a volume of effective drop from 20 to 50 μL. From the applied portion, the precorneal pocket contains just 7–10 μL by supplanting tear, which is as of now present in the precorneal pocket [114]. Such overabundance managed portion might be lost by means of spillage from the precorneal pocket or waste through the nasolacrimal pipe [115]. Precorneal tear waste washes the topically imparted drops/arrangements in the initial 15–30 s, causing a genuine decrease in drug contact time and finely lessening visual bioavailability <5 [116, 117, 118].

4.2 Conjunctival lymph and blood stream

The conjunctiva is immensely vascularized and equipped with lymph vessels. Topically applied drugs into the precorneal pocket may be essentially clear out into the fundamental and lymph dispersal while crossing the conjunctiva [119], achieving lower drug invasion to more significant visual tissue. Conjunctival blood and lymph stunningly limit the vehicle of sodium fluorescein from showing up at the inside retinal tissues [120, 121], exhibiting the effective work in drug end.

4.3 Watery humor

Watery humor produced by the ciliary body overflowing towards the cornea, the alternate way to the medicine entrance applied topically. Topically sedates, what separate across the front corneal epithelial may be also described by liquid humor stream. Meds may be exhausted out through trabecular meshwork into the channel of Schlemm. Greatest hydrophilic meds are seen as discard by watery humor using sub therapeutic levels in the inside visual tissues [122].

BAB activity can be abandoned due to immunological visual deterioration and can prevent reasonable vulnerabilities of experts on various subjects. Intravitreal monitored fluorescein-tested serum protein (FRSA) enters the main chamber after 5 min in the eye, but not in the control/abiotic eye [121]. It is worth noting that the basic fluorescein sodium particles actually pass through the BAB, but not the FRSA. The FRSA scattered in the first room seeks a two-step plan until it appears in the previous room in 23 h, generally with exorbitant obsession (Cmax). After that, the fluorescence continued to decrease. These preliminary results indicate that the student lost the boundaries of the BAB obstacle and lost dosing to the main chamber. In another audit, Kongetal. It has been shown that basic point-edge glaucoma damages the BAB and mediates irritation in the anterior part of the eye. Inconsistent reports between patients with severe terminal glaucoma (PACG) and patients with refractory PACG assume that patients with exceptional PACG have higher BAB damage than patients with consistent PACG [123]. It has been speculated that a significant increase in intra orbital load may be the causative variable for distinguishing BAB in the eye of PACG patients.

Advertisement

5. Posterior segment staticbarriers

5.1 Sclera

The sclera is one of the peripheral layers of the eye that cater security and forestalls the passage of exogenous substances to the back visual tissues. Porousness across sclera predominantly relies upon specific standards of the saturating particle, like atomic range, physicochemical properties and surface charge. Drug porousness across sclera steadily diminishes with expanding lipophilicity and sub-atomic span. Thus, particles with higher lipophilicity and bigger sub-atomic range might be kept from penetration through watery scleral pores. The contrary surface charge of the atom further restricts its pervasion across sclera. Drug atoms become caught in the pores of sclera causing low porousness presumably through connections with contrarily charged pores and proteoglycan network [3]. Scleral thickness varies as per their physical area. It is exceptionally wide posterior segment, close to the optic nerve because of its extensiveness; the back sclera has extremely low porousness for drug particles.

Bruch’s shiftIn a sense, Bruch’s film is concerned with directing the trading of dietary supplements, water, metabolic waste, oxygen, and biomolecules between the choroidal bloodstream and the retina [124]. Bruch’s layer thickness increases with age, resulting in calcification, high cross-linking of collagen fibers, and turnover of glycosaminoglycans. The contrast in the thickness of this layer can also affect the permeability of the film from the sclera to the retinal tissue. Also, most medicines can enter the choroid before reaching Bruch’s layer. In age-related conditions such as macular degeneration, extracellular edema and accumulation of edema result in oxidative pressure and inflammation of the Bruch layer. As extracellular flots and jets grow, the transport of supplements and drug atoms decreases. This may contribute to an increase in the metabolic pressure of the option [6, 123].

5.2 Blood retina obstruction

Blood retinal obstruction (BRB) consists of medial and lateral BRBs. The outer BRB is composed of tight junctions of RPE cells, while the inner BRB contains endothelial cells such as retinal hair. Tight junctions of the external BRB release retinal photoreceptors from the RPE [125, 126]. Two astrocytes and Muller cells provide the practical help expected at these narrow intersections. Together, these two cell types maintain the exchange of substances between the outer choroid and the inner retina. It supports astrocytes, catches up honestly, improves the occlusive properties of retinal endothelial blood vessels [127], and increases the safety of inward retinal cells so that they do not boil the flowing particles during retinal proliferation. The absence of windows on RPE and retinal endothelial cells is indistinguishable from blood brain obstruction and indicates potential drug distribution. RPE gives dissemination of tiny atoms like CO2, O2 and lipophilic particles to inward retinal tissues from the choroid. In this way, the boat of atoms might be intervened by receptor or energy/ATP-subordinate liquid stage pinocytosis. Thus, drug entrance is profoundly controlled because of the presence of tight intersections. Likewise, RPE go about as a siphon by getting dried out the sub retinal space. This includes keeping up with the ordinary neuro retinal pairing with RPE [128]. The siphon working is vital in light of the fact that there is no solid bond between the portions of outer brain retina and RPE. The inter photoreceptor framework, a gel-like thick arrangement happens between retinal cells, cause exceptionally powerless bonds. During the ischemic circumstances, retinal cement powers decline inside the space of minutes [129].

5.3 Efflux siphons

Transmembrane efflux siphons, for example, P-gp or MRP revealed on apical and basal sides of human RPE have been accounted for [130]. Articulation of P-gp on rodent retinal endothelial cells has additionally announced. Additionally, the presence of P-gp, MRP4 and bosom disease opposition.

protein in retinal vasculature of post pregnancy mouse was accounted for. The outflow of P-gp in retinal astrocytes was additionally been accounted for [131]. As of late, unique articulation levels of MRP1, MRP4 and MRP5 efflux proteins in human RPE cells have additionally been accounted for [63]. At cell level, these efflux siphons are energetically associated with diminishing the intracellular medication focuses and play out a significant obstruction to tranquilize conveyance.

5.4 Choroidal blood and lymph circulation

Choroid is an exceptionally vascularized tissue and outfitted with lymph vessels. This tissue has biggest blood vasculature (−80%) of the all out visual blood supply co-connected with iris-ciliary body and retina [7]. Lipophilic medication atoms are effectively depleted into the choroidal and fundamental dissemination, then, at that point, section into the inward visual tissues. This limited medication might prompt sub remedial medication fixation in retinal tissues. This limited medications entry might prompt sub helpful medication focus in retinal tissues. Studies with hydrophilic medications, for example, sodium fluorescein communicated extremely low end through choroid [8].

Advertisement

6. Ocular drug delivery systems

Visual medication conveyance framework there are principle issue of fast and exhaustive end of regular eye drops from the eye [9]. This issue brings about loss of enormous measure of medication. Just hardly any measure of medication enters the corneal layer and enter to inward tissue of eye [10, 11]. The primary explanation of medication misfortune incorporates lachrymal waste and medication weakening by tears [13]. This super smoothness diminishes the visual bioavailability and cause undesirable harmful and incidental effect. Yet, late biomaterials and nanotechnology headways have started a significant development in investigation into biodegradable microparticles and nanoparticles, hydrogel, and visual inserts, all of which can contain visual pharmacologic specialists, considering further developed prescription conveyance. Besides, supported discharge drug treatments might diminish the in general financial effect of visual sicknesses by lessening the aftereffects related with current clinical therapies [14]. Visual medication conveyance is progressing dangerously fast, on account of consistently advancing ways to deal with designated conveyance (Table 1).

Brand nameDosage formUses
Acuvail4.5 mg/ml ketorolac tromethamine arrangement (0.45%) in a solitary use vialCataract medical procedure
Alocril2% is an unmistakable, yellow, sterile solutionAllergic conjunctivitis
Elestat0.05% epinastineHCl ophthalmic solutionAllergic conjunctivitis
Ozurdex0.7 mg dexamethasone intravitreal visual implantRetinal vein impediment
Pred Forteprednisolone acetic acid derivation ophthalmic suspension, USPBulbar conjunctiva
Trivaris80 mg/ml triamicinoloneacetonide injectable suspensionSympathetic ophthalmia
Zymar0.3% gatifloxacin ophthalmic solutionBacterial conjunctivitis
Zymaxid0.5% gatifloxacin ophthalmic solutionBacterial conjunctivitis

Table 1.

Showcased product of ocular drug delivery system.

6.1 Nanoparticles and microparticles

Nanoparticles and microparticles particulate polymeric medication conveyance frameworks comprise of miniature and nanoparticles. The prevalent size of microparticles for the ophthalmic organization in regards to 5–10 mm, over this size, a scratching sensation inside the eye might result after visual application. Microspheres and nanoparticles recommend excited drug transporters for ophthalmic application. The medication restricting depends on the physicochemical properties of the nano-or miniature molecule polymer. There after best medication restricting to those particles, the medication retention inside the eye is worked on widely when contrasted with eye drops. Particulates like nanocapsules, nanoparticles, nanosuspensions and so forth are wont to work on the bioavailability of visually applied drugs [15].

6.2 Fake tear inserts

In 1981 Merck, Sharp and Dohme was fostered an instrument for supported discharge alluded to as counterfeit tear. Industrially Lacrisert is out there inside the market. It contains hydroxyl propyle cellulose without additives used in the treatment of dry eye [132].

6.3 Visual inserts

In conventional arrangement of medication conveyance like suspension, arrangement and balm have various drawbacks. Visual additions limit the matter happen in customary framework. These are strong measurement structure have high ability to hold required convergence of medication in to designated tissue. A few examples of visual supplements are Dexamethasone, Pilocarpine nitrate, Tropicamide and Timolol Maleate [17].

Benefits of visual additions

A few benefits of visual addition which might be summed up as follows:

  • Decrease of complete assimilation (which happens uninhibitedly with eye drops by means of the nasolachrimal channel and nasal mucosa).

  • Likelihood of delivering drugs at a sluggish, consistent rate.

  • Upgrade of visual residence, so that lengthy medication action and a raised bioavailability regarding regular vehicles.

  • Right dosing (talk to eye drops which will be amazingly ingrained by the patient and are somewhat disappeared after organization, each addition are frequently made to contain a precise portion which is completely stayed at the organization site).

  • Further developed time span with worship to fluid arrangements.

  • Better persistent consistence, coming about because of compressed recurrence of organization and a lesser frequency of visual and foundational secondary effects.

  • Plausibility of consolidating different novel substance/innovative methodologies.

6.4 Liposome

Liposomes are spherical phospholipids vesicles made up of one or more concentric bilayers arranged in a ring. They are non-toxic and biodegradable in nature, owing to the fact that they are made up of lipids that are similar to those found in biological membranes. Liposomes have the ability to entrap both hydrophilic and hydrophobic compounds in the aqueous compartments or the lipid layer due to their amphiphilic nature. Liposomes have a positive, negative, or neutral surface charge depending on their composition. Surface charge is one of the most important factors in liposome stability because it reduces the rate of aggregation and fusion during storage. The fact that the corneal epithelium is thinly coated with negatively charged mucin may explain why positively charged liposomes appear to be more important. Negatively charged liposomes are more effective than positively charged liposomes because of lowering of IOP over a longer period of time. The different structures and shape of liposome are classified;1. Multilamella vesicle (MLV): 520 lipid bilayer and very 5000 nm measurements 2. Oligolamera vesicle (OLV): 25 lipid bilayer and 1,001,000 nm measurements 3. Polyvascular vesicle (MVV): Very 1000 nm in diameter. 4. Single-layer vesicle (ULV): Single-layer of lipid Thesecan also be further categorized based on size potential 1. An SUV (more discreet single-layer vesicle) sized at 2040 nm. 2. MUV (medium-sized single-layer vesicle) with a size of 4080 nm. 3. 1,001,000 nm size LUV (larger single layer vesicle) 4. GUV (giant single-layer vesicle) with a size exceeding 1000 nm.

6.5 Collagen shield

Collagens have amazing biocompatibility and security in view of its natural attributes and for the most part used in clinical application since biodegradability and feeble antigenecity. Collagen safeguard was first ready from procine sclera tissues since they bear a collagens equal arrangement thereto of natural eye. The safeguards are hydrated before the inclusion into the eye commonly the medication is stacked into the medication answer for a time of at some point before application.

Collagen safeguard regularly produce a few uneasiness and obscured vision and are not fit every understanding. For improvement of this issue, Kaufman found a substitution idea of medication conveyance by combination of collagen safeguard particles and gets in-tuned with focal points named collasomes. Collasomes could be ingrained under the eyelid and limit the obscured vision issue [26].

6.6 Mucosal adhesive dose structure

In fact, mucosal adherent dose structures for visual delivery present many difficulties [24]. This approach relies on vehicles containing polymers that can bind to conjunctival mucin via non-covalent bonds. Mucoadhesive polymers are conventional polymeric hydrophilic colloids and contain several hydrophilic, practical compounds that can establish electrostatic bonds, similar to carboxyls, hydroxyls, amides, and sulfates. The bioadhesive metric structure showed higher drug bioavailability than the traditional dose structure. The effect of polyacrylicacid as a bioadhesive polymer on the visual bioavailability of timolol was evaluated. It has also been used to improve the visual bioavailability of progesterone [133].

6.7 Microemulsion

Microemulsions have local properties and a clear design. They are finished by automatic emulsification and disinfected directly. These assemblies have high drug dissolving capacity and excellent robustness. Due to these properties, it has excellent bioavailability. The mechanism of action of the drug is assimilation. Microemulsions contain nanospheres inside that hold the drug substance on the outer surface of the cornea and reduce drug waste [134].

6.8 Solution and suspension

These are liquid assemblies containing the active ingredients used in visual drug delivery. The drug substance must remain on the surface of the eye or in the internal area of the eye after passing through the cornea or conjunctiva. These arrangements even have some inconveniences such as unfortunate bioavailability and potency. Issues related to layout planning safety, bioavailability, and visual maintenance seasons are addressed through the work of a wide range of analysts by increasing thickness, using additives, and changing the pH of these definitions [28, 135].

6.9 Sol to gel systems

In 1980s a substitution idea delivering a gel set up was suggested by specialists. This thought of medication conveyance to eye is generally utilized in light of the fact that it expands the consistency and diminished the seepage of medication from cornea. Along these lines the bioavailability of medication naturally expanded. The in-situ gelling frameworks are regularly impacted by temperature, pH or particle enactment. In trial examination of bunny eye examiner found the in-situ gelling framework gives better and delay of medication contrast with the conventional eye drops [18].

Advertisement

7. Conclusion

The broad add visual medication conveyance during the sooner time frame. It’s been plan, to expand the span of topically applied drugs inside the corneal and conjunctiva area. A few new methodologies like nanoparticles, liposome, contact focal points, visual supplements, collagen safeguard, set up initiated gel arrangement, non corneal course of visual medication dissemination, and nanoparticles-based polymeric arrangements and gels are being created by the drug science. An exhaustive depiction of the eye life structures and physiology, alongside its obstruction properties are given in this part. Perusers will be educated with various visual tissues, their physical areas, work and their static and dynamic hindrance nature. The natural eyes are fundamental and biggest delicate and profoundly watched organs in the body. These are extremely confounded organs and which are immediate contact with the outside conditions, this organ is truly helpless against injury and subsequently impermeable to exogenous synthetic compounds. In addition, over the top tear creation in view of reaction to outer upgrades or remedial specialists and furthermore expanded flickering rate due to adding to shield. These security capacities normally created and go about as an obstruction to remotely applied drugs, particularly when these are designated to tissues at the rear of the eye. Articulation of medication efflux siphons at a cell level squares drug section into the sick cell and afterward presents serious difficulties to visual medication conveyance. The physicochemical properties of the medication and conveyance framework can be arched relying upon the designated eye tissue. For the back eye conveyance, different courses of medication organization, for example, periocular infusions/inserts, are successfully utilized to defeat visual static and dynamic boundaries. Accordingly, information on the different visual tissues alongside the dynamic drug and physicochemical boundary properties is vital. The data gave in this part might facilitate the improvement of an ideal definition for designated conveyance.

References

  1. 1. Patton TF, Robinson JR. Quantitative precorneal disposition of topically applied pilocarpine nitrate in rabbit eyes. Journal of Pharmaceutical Sciences. 1976;65(9):1295-1301
  2. 2. Wood RW, Li VH, Kreuter J, Robinson JR. Ocular disposition of poly-hexyl-2-cyano [3-14C] acrylate nanoparticles in the albino rabbit. International Journal of Pharmaceutics. 1985;23(2):175-183
  3. 3. Lee VHL, Robinson JR. Mechanistic and quantitative evaluation of precornealpilocarpine disposition in albino rabbits. Journal of Pharmaceutical Sciences. 1979;68(6):673-684
  4. 4. Joshi A, Ding S, Himmelstein KJ. Reversible gelatin composition and method of use. US Patent No. 5,252,318; October 12 1993
  5. 5. Kumar S, Haglund BO, Himmelstein KJ. In situ-forming gels for ophthalmic drug delivery. Journal of Ocular Pharmacology and Therapeutics. 1994;10(1):47-56
  6. 6. Keister JC, Cooper ER, Missel PJ, Lang JC, Hager DF. Limits on optimizing ocular drug delivery. Journal of Pharmaceutical Sciences. 1991;80(1):50-53
  7. 7. Lee VL. Precorneal, corneal, and postcorneal factors. Drugs and the Pharmaceutical Sciences. 1993;58:59-81
  8. 8. Tang NE, Zuure PL, Pardo RD, de Keizer RJ, Van Best JA. Reflex lacrimation in patients with glaucoma and healthy control subjects by fluorophotometry. Investigative Ophthalmology & Visual Science. 2000;41(3):709-714
  9. 9. White WL, Glover AT, Buckner AB. Effect of blinking on tear elimination as evaluated by dacryoscintigraphy. Ophthalmology. 1991;98(3):367-369
  10. 10. Stewart WC. Review timolol hemihydrate: A new formulation of Timolol for the treatment of glaucoma. Journal of Ocular Pharmacology and Therapeutics. 1996;12(2):225-237
  11. 11. Ahmed I, Patton TF. Importance of the noncorneal absorption route in topical ophthalmic drug delivery. Investigative Ophthalmology & Visual Science. 1985;26(4):584-587
  12. 12. Schoenwald RD, Deshpande GS, Rethwisch DG, Barfknecht CF. Penetration into the anterior chamber via the conjunctival/scleral pathway. Journal of Ocular Pharmacology and Therapeutics. 1997;13(1):41-59
  13. 13. Mishima S, Gasset A, Klyce SD, Baum JL. Determination of tear volume and tear flow. Investigative Ophthalmology & Visual Science. 1966;5(3):264-276
  14. 14. Chrai SS, Patton TF, Mehta A, Robinson JR. Lacrimal and instilled fluid dynamics in rabbit eyes. Journal of Pharmaceutical Sciences. 1973;62(7):1112-1121
  15. 15. allergan.com
  16. 16. Sikandar MK, Sharma PK, Visht S. Ocular drug delivery system: An overview. International Journal of Pharmaceutical Sciences and Research. 2011;2(5):1168
  17. 17. LaMotte J, Grossman E, Hersch J. The efficacy of cellulosic ophthalmic inserts for treatment of dry eye. Journal of the American Optometric Association. 1985;56(4):298-302
  18. 18. Rootman DS, Jantzen JA, Gonzalez JR, Fischer MJ, Beuerman R, Hill JM. Pharmacokinetics and safety of transcornealiontophoresis of tobramycin in the rabbit. Investigative Ophthalmology & Visual Science. 1988;29(9):1397-1401
  19. 19. Nagarsenker MS, Londhe VY, Nadkarni GD. Preparation and evaluation of liposomal formulations of tropicamide for ocular delivery. International Journal of Pharmaceutics. 1999;190(1):63-71
  20. 20. Mitra AK. Ophthalmic Drug Delivery Systems. 2003
  21. 21. Hui HW, Robinson JR. Ocular delivery of progesterone using a bioadhesive polymer. International Journal of Pharmaceutics. 1985;26(3):203-213
  22. 22. Vandamme TF. Microemulsions as ocular drug delivery systems: Recent developments and future challenges. Progress in Retinal and Eye Research. 2002;21(1):15-34
  23. 23. Mueller WH, Deardorff DL. Ophthalmic vehicles: The effect of methylcellulose on the penetration of homatropinehydrobromide through the cornea. Journal of the American Pharmaceutical Association. 1956;45(5):334-341
  24. 24. Krishna N, Brow F. Polyvinyl alcohol as an ophthalmic vehicle: Effect on regeneration of corneal epithelium. American Journal of Ophthalmology. 1964;57(1):99-106
  25. 25. Middleton DL, Robinson JR. Design and evaluation of an ocular bioadhesive delivery system. STP Pharma Sciences. 1991;1(3):200-206
  26. 26. Lerman S, Davis P, Jackson WB. Prolonged release hydrocortisone therapy. Canadian Journal of Ophthalmology. Journal CanadienD’ophtalmologie. 1973;8(1):114-118
  27. 27. Karthikeyan D, Bhowmick M, Pandey VP, Nandhakumar J, Sengottuvelu S, Sonkar S, et al. The concept of ocular inserts as drug delivery systems: An overview. Asian Journal of Pharmaceutics (AJP). 2014;2(4)
  28. 28. Vadnere M, Amidon G, Lindenbaum S, Haslam JL. Thermodynamic studies on the gel-sol transition of some pluronicpolyols. International Journal of Pharmaceutics. 1984;22(2-3):207-218
  29. 29. Worakul N, Robinson JR. Ocular pharmacokinetics/pharmacodynamics. European Journal of Pharmaceutics and Biopharmaceutics. 1997;44(1):71-83
  30. 30. McGhee PCN. An Overview of Topical Ophthalmic Drugs and the Therapeutics of Ocular Infection. CNJ Mc Ghee: Ocular Therapeutics. 2017
  31. 31. Jitendra SP, Banik A, Dixit S. A new trend: Ocular drug delivery system. International. Journal of Pharmaceutical Sciences. 2011;2(3):720-744
  32. 32. Patel A, Cholkar K, Agrahari V, Mitra AK. Ocular drug delivery systems: An overview. World Journal of Pharmacology. 2013;2(2):47
  33. 33. Urtti A. Challenges and obstacles of ocular pharmacokinetics and drug delivery. Advanced Drug Delivery Reviews. 2006;58(11):1131-1135
  34. 34. Galloway NR. Common eye diseases and their management. American Journal of Ophthalmology. 100(2):360
  35. 35. Ng JS. Ocular anatomy and physiology. Optometry & Vision Science. 2009;86(10):1208
  36. 36. Gaudana R, Ananthula HK, Parenky A, Mitra AK. Ocular drug delivery. The AAPS Journal. 2010;12(3):348-360
  37. 37. Khurana VK, Agrahari V. Recent advances in ocular drug delivery. Advanced Drug Delivery. 2014. (Chapter 19)
  38. 38. Barar J, Asadi M, Mortazavi-Tabatabaei SA, Omidi Y. Ocular drug delivery; impact of in vitro cell culture models. Journal of Ophthalmic & Vision Research. 2009;4(4):238
  39. 39. Achouri D, Alhanout K, Piccerelle P, Andrieu V. Recent advances in ocular drug delivery. Drug development and industrial pharmacy. 2013;39(11):1599-1617
  40. 40. Barar J, Javadzadeh AR, Omidi Y. Ocular novel drug delivery: Impacts of membranes and barriers. Expert Opinion on Drug Delivery. 2008;5(5):567-581
  41. 41. Ghate D, Edelhauser HF. Ocular drug delivery. Expert Opinion on Drug Delivery. 2006;3(2):275-287
  42. 42. Ruponen M, Urtti A. Undefined role of mucus as a barrier in ocular drug delivery. European Journal of Pharmaceutics and Biopharmaceutics: Official Journal of Arbeitsgemeinschaft fur PharmazeutischeVerfahrenstechnik eV. 2015;96:442-446
  43. 43. Gaudana R, Jwala J, Boddu SH, Mitra AK. Recent perspectives in ocular drug delivery. Pharmaceutical Research. 2009;26(5):1197-1216
  44. 44. Anand BS, Dey S, Mitra AK. Current prodrug strategies via membrane transporters/receptors. Expert Opinion on Biological Therapy. 2002;2(6):607-620
  45. 45. Kaur IP, Garg A, Singla AK, Aggarwal D. Vesicular systems in ocular drug delivery: An overview. International Journal of Pharmaceutics. 2004;269(1):1-14
  46. 46. Du Toit LC, Pillay V, Choonara YE, Govender T, Carmichael T. Ocular drug delivery—A look towards nanobioadhesives. Expert Opinion on Drug Delivery. 2011;8(1):71-94
  47. 47. Djebli N, Khier S, Griguer F, Coutant AL, Tavernier A, Fabre G. Ocular drug distribution after topical administration: Population pharmacokinetic model in rabbits. European Journal of Drug Metabolism and Pharmacokinetics. 2016
  48. 48. Siegal. Glaucoma Medical Therapy: Principles and Management: Principles and Management. 2008
  49. 49. Dua HS, Faraj LA, Said DG, Gray T, Lowe J. Human corneal anatomy redefined: A novel pre-Descemet’s layer (Dua’s layer). Ophthalmology. 2013;20(9):1778-1785
  50. 50. Almeida H, Amaral MH, Lobao P, Lobo JM. In situ gelling systems: A strategy to improve the bioavailability of ophthalmic pharmaceutical formulations. Drug Discovery Today. 2014;19(4):400-412
  51. 51. Salazar MM, Wainer I, Patil PN. Relevance of drug-melanin interactions to ocular pharmacology and toxicology. Journal of Ocular Pharmacology. 1994;10(1):217-239
  52. 52. Barot M, Patel M, Kwatra D, Mitra AK. 7—Transporter–metabolism interplay in the eye. In: Ocular Transporters and Receptors. Woodhead Publishing; 2013. pp. 229-248
  53. 53. Dhananjay P, Krishna RV, AswaniDutt V, Mitra AK. Biology of ocular transporters: Efflux and influx transporters in the eye. In: Ocular Transporters and Receptors. WoodheadPublishing; 2013. pp. 37-84
  54. 54. Maurice D. Practical issues in intravitreal drug delivery. Journal of Ocular Pharmacology and Therapeutics: The Official Journal of the Association for Ocular Pharmacology and Therapeutics. 2001;17(4):393-401
  55. 55. Raghava S, Hammond M, Kompella UB. Periocular routes for retinal drug delivery. Expert Opinion on Drug Delivery. 2004;1(1):99-114
  56. 56. Okada AA, Wakabayashi T, Morimura Y, Kawahara S, Kojima E, Asano Y. Trans-Tenon’sretrobulbar triamcinolone infusion for the treatment of uveitis. The British Journal of Ophthalmology. 2003;87(8):968-971
  57. 57. Johnson KS, Chu DS. Evaluation of sub-Tenon triamcinolone acetonide injections in the treatment of scleritis. American Journal of Ophthalmology. 2010;149(1):77-81
  58. 58. Chang DT, Herceg MC, Bilonick RA, Camejo L, Schuman JS, Noecker RJ. Intracameral dexamethasone reduces inflammation on the first postoperative day after cataract surgery in eyes with and without glaucoma, Clinicalophthalmology. 2009;3:345-355
  59. 59. Mishima S. Clinical pharmacokinetics of the eye: Proctor lecture. Investigative Ophthalmology & Visual Science. 1981;21(4):504-541
  60. 60. Le Bourlais C, Acar L, Zia H, Sado PA, Needham T, Leverge R. Ophthalmic drug delivery systems—Recent advances. Progress in Retinal and Eye Research. 1998;17(1):33-58
  61. 61. Urtti A, Salminen. Minimizing systemic absorption of topically administered ophthalmic drugs. Survey of Ophthalmology. 1993;37(6):435-456
  62. 62. Benedetti MS, Whomsley R, Poggesi I, Cawello W, Mathy FX, Delporte ML. Drug metabolism and pharmacokinetics. Drug Metabolism Reviews. 2009;41(3):344-390
  63. 63. Tangri P, Khurana S. Basics of ocular drug delivery systems. International Journal of Research in Pharmaceutical and Biomedical Sciences. 2011;2(4):1541-1552
  64. 64. Katragadda S, Gunda S, Hariharan S, Mitra AK. Ocular pharmacokinetics of acyclovir amino acid ester prodrugs in the anterior chamber: Evaluation of their utility in treating ocular HSV infections. International Journal of Pharmaceutics. 2008;359(1-2):15-24
  65. 65. Levison ME, Levison JH. Pharmacokinetics and pharmacodynamics of antibacterial agents. Infectious Disease Clinics of North America. 2009;23(4):791-815
  66. 66. Del Amo EM, Urtti A. Rabbit as an animal model for intravitreal pharmacokinetics: Clinical predictability and quality of the published data. Experimental Eye Research. 2015;137:111-124
  67. 67. Bartlett JD, Jaanus SD. Clinical Ocular Pharmacology. St. Louis: Butterworth-Heinemann/Elsevier; 2008
  68. 68. Novack GD, Robin AL. Ocular pharmacology. Journal of Clinical Pharmacology. 2016;56(5):517-527
  69. 69. Vellonen KS, Soini EM, Amo EM, Urtti A. Prediction of ocular drug distribution from systemic blood circulation. Molecular Pharmaceutics. 2015
  70. 70. Tojo K. Pharmacokinetic model for ocular drug delivery. Chemical & Pharmaceutical Bulletin. 2004;52(11):1290-1294
  71. 71. Chiou GC. Systemic delivery of polypeptide drugs through ocular route. Journal of Ocular Pharmacology. 1994;10(1):93-99
  72. 72. Makoid MC, Robinson JR. Pharmacokinetics of topically applied pilocarpine in the albino rabbit eye. Journal of Pharmaceutical Sciences. 1979;68(4):435-443
  73. 73. Fauser S, Kalbacher H, Alteheld N, Koizumi K, Krohne TU, Joussen AM. Pharmacokinetics and safety of intravitreally delivered etanercept. Graefe’sArchive for Clinical and Experimental Ophthalmology Albrecht von Graefes Archiv fur klinische undexperimentelleOphthalmologi. 2004;242(7):582-586
  74. 74. Iyer MN, He F, Wensel TG, Mieler WF, Benz MS, Holz ER. Intravitreal clearance of moxifloxacin. Transactions of the American Ophthalmological Society. 2005;103:76-81. discussion 3
  75. 75. Bakri SJ, Snyder MR, Reid JM, Pulido JS, Ezzat MK, Singh RJ. Pharmacokinetics of intravitrealranibizumab (Lucentis). Ophthalmology. 2007;114(12):2179-2182
  76. 76. Wu CY, Wu HM, Lee JD, Weng HH. Stroke risk factors and subtypes in different age groups: A hospital-based study. Neurology India. 2010;58(6):863-868
  77. 77. Xu L, Lu T, Tuomi L, Jumbe N, Lu J, Eppler S. Pharmacokinetics of ranibizumab in patients with neovascular age-related macular degeneration: A population approach. Investigative Ophthalmology & Visual Science. 2013;54(3):1616-1624
  78. 78. Gaudreault J, Fei D, Rusit J, Suboc P, Shiu V. Preclinical pharmacokinetics of Ranibizumab (rhuFabV2) after a single intravitreal administration. Investigative Ophthalmology & Visual Science. 2005;46(2):726-733
  79. 79. Proksch JW, Lowe ER, Ward KW. Ocular pharmacokinetics of mapracorat, a novel, selective glucocorticoid receptor agonist, in rabbits and monkeys. Drug Metabolism and Disposition: The Biological Fate of Chemicals. 2011;39(7):1181-1187
  80. 80. Le KN, Gibiansky L, Good J, Davancaze T, van Lookeren CM, Loyet KM. A mechanistic pharmacokinetic/pharmacodynamic model of factor D inhibition in cynomolgus monkeys by lampalizumab for the treatment of geographic atrophy. The Journal of Pharmacology and Experimental Therapeutics. 2015;355(2):288-296
  81. 81. Drolet DW, Nelson J, Tucker CE, Zack PM, Nixon K, Bolin R. Pharmacokinetics and safety of an anti-vascular endothelial growth factor aptamer (NX1838) following injection into the vitreous humor of rhesus monkeys. Pharmaceutical Research. 2000;17(12):1503-1510
  82. 82. Tucker CE, Chen LS, Judkins MB, Farmer JA, Gill SC, Drolet DW. Detection and plasma pharmacokinetics of an anti-vascular endothelial growth factor oligonucleotide-aptamer (NX1838) in rhesus monkeys. Journal of Chromatography B, Biomedical Sciences and Applications. 1999;732(1):203-212
  83. 83. Ng EW, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP. Targeted anti-VEGF aptamer for ocular vascular diseas. Nature Reviews Drug Discovery. 2006;5(2):123-132
  84. 84. Liu YC, Peng Y, Lwin NC, Wong TT, Venkatraman SS, Mehta JS. Optimization of subconjunctival biodegradable microfilms for sustained drug delivery to the anterior segment in a small animal model. Investigative Ophthalmology & Visual Science. 2013;54(4):2607-2615
  85. 85. Di Tommaso C, Bourges JL, Valamanesh F, Trubitsyn G, Torriglia A, Jeanny JC. Novel micelle carriers for cyclosporin A topical ocular delivery: In vivo cornea penetration, ocular distribution and efficacy studies. European Journal of Pharmaceutics and Biopharmaceutics: Official Journal of Arbeitsgemeinschaft fur PharmazeutischeVerfahrenstechnikeV. 2012;81(2):257-264
  86. 86. Hendrix DV, Stuffle JL, Cox SK. Pharmacokinetics of topically applied ciprofloxacin in equine tears. Veterinary Ophthalmology. 2007;10(6):344-347
  87. 87. Proksch JW, Granvil CP, Siou-Mermet R, Comstock TL, Paterno MR, Ward KW. Ocular pharmacokinetics of besifloxacin following topical administration to rabbits, monkeys, and humans. Journal of Ocular Pharmacology and Therapeutics. The Official Journal of the Association for Ocular Pharmacology and Therapeutics. 2009;25(4):335-344
  88. 88. Shen J, Durairaj C, Lin T, Liu Y, Burke J. Ocular pharmacokinetics of intravitreally administered brimonidine and dexamethasone in animal models with and without blood-retinal barrier breakdown. Investigative Ophthalmology & Visual Science. 2014;55(2):1056-1066
  89. 89. Kymionis GD, Bouzoukis DI, Diakonis VF, Siganos C. Treatment of chronic dry eye: Focus on cyclosporine. Clinical Ophthalmology. 2008;2(4):829-836
  90. 90. Mathews D, Mathews J, Jones NP. Low-dose cyclosporine treatment for sight-threatening uveitis: Efficacy, toxicity, and tolerance. Indian Journal of Ophthalmology. 2010;58(1):55-58
  91. 91. Tang-Liu DD, Acheampong A. Ocular pharmacokinetics and safety of ciclosporin, a novel topical treatment for dry eye. Clinical Pharmacokinetics. 2005;44(3):247-261
  92. 92. Bucolo C, Melilli B, Piazza C, Zurria M, Drago F. Ocular pharmacokinetics profile of different indomethacin topical formulations. Journal of Ocular Pharmacology and Therapeutics: The Official Journal of the Association for Ocular Pharmacology and Therapeutics. 2011;27(6):571-576
  93. 93. Amar T, Caillaud T, Elena PP. Ocular pharmacokinetic study following single and multiple azithromycin administrations in pigmented rabbits. Current Eye Research. 2008;33(2):149-158
  94. 94. Akpek EK, Vittitow J, Verhoeven RS, Brubaker K, Amar T, Powell KD. Ocular surface distribution and pharmacokinetics of a novel ophthalmic 1% azithromycin formulation. Journal of Ocular Pharmacology and Therapeutics: The Official Journal of the Association for Ocular Pharmacology and Therapeutics. 2009;25(5):433-439
  95. 95. Johannesson G, Moya MD, Asgrimsdottir GM, Lund SH, Thorsteinsdottir M, Loftsson T. Kinetics of gamma-cyclodextrin nanoparticle suspension eye drops in tear fluid. ActaOphthalmologica. 2014;92(6):550-556
  96. 96. Amon M, Busin M. Efficacy and safety for postoperative anti-inflammatory use. International Ophthalmology. 2012;32(5):507-517
  97. 97. Schopf L, Enlow E, Popov A, Bourassa J, Chen H. Ocular pharmacokinetics of a novel LoteprednolEtabonate 0.4% ophthalmic formulation. Ophthalmology and Therapy. 2014
  98. 98. Kapanigowda UG, Nagaraja SH, Ramaiah B, Boggarapu PR. Improved intraocular bioavailability of ganciclovir by mucoadhesive polymer based ocular microspheres: Development and simulation process in Wistar rats. Daru: Journal of Faculty of Pharmacy, Tehran University of Medical Sciences. 2015;23(1):49
  99. 99. Bakri SJ, Snyder MR, Reid JM, Pulido JS, Singh RJ. Pharmacokinetics of intravitrealbevacizumab (Avastin). Ophthalmology. 2007;114(5):855-859
  100. 100. Cholkar K, Vadlapudi AD, Trinh HM, Mitra AK. Compositions, formulation, pharmacology, pharmacokinetics, and toxicity of topical, periocular, and intravitreal ophthalmic drugs. In: Gilger CB, editor. Ocular Pharmacology and Toxicology. Totowa, N.J: Humana Press; 2014. pp. 91-118
  101. 101. Yang Y, Bailey C, Loewenstein A, Massin P. Intravitreal corticosteroids in diabetic macular edema: Pharmacokinetic considerations. Retina. 2015;35(12):2440-2449
  102. 102. Augustin A. Anecortave acetate in the treatment of age-related macular degeneration. Clinical Interventions in Aging. 2006;3:237-246
  103. 103. Edelhauser HF, Rendleman CL, Robinson MR, Dawson DG, Chader GJ, Grossniklaus HE. Ophthalmic drug delivery systems for the treatment of retinal diseases: Basic research to clinical applications. Investigative Ophthalmology & Visual Science. 2010;51(11):5403-5420
  104. 104. Hayden BC, Jockovich ME, Murray TG, Voigt M, Milne P, Kralinger M. Pharmacokinetics of systemic versus focal carboplatin chemotherapy in the rabbit eye: Possible implication in the treatment of retinoblastoma. Investigative Ophthalmology & Visual Science. 2004;45(10):3644-3649
  105. 105. Winter U, Buitrago E, Mena HA, Del Sole MJ, Laurent V, Negrotto S, et al. Pharmacokinetics, safety, and efficacy of intravitreal digoxin in preclinical models for retinoblastoma. Investigative Ophthalmology & Visual Science. 2015;56(8):4382-4393
  106. 106. Mannermaa E, Vellonen KS, Urtti A. Drug transport in corneal epithelium and blood–retina barrier: Emerging role of transporters in ocular pharmacokinetics. Advanced Drug Delivery Reviews. 2006;58(11):1136-1163
  107. 107. Dey S, Gunda S, Mitra AK. Pharmacokinetics of erythromycin in rabbit corneas after single-dose infusion: Role of P-glycoprotein as a barrier to in vivo ocular drug absorption. Journal of Pharmacology and Experimental Therapeutics. 2004;311(1):246-255
  108. 108. Hariharan S, Gunda S, Mishra GP, Pal D, Mitra AK. Enhanced corneal absorption of erythromycin by modulating P-glycoprotein and MRP mediated efflux with corticosteroids. Pharmaceutical Research. 2009;26(5):1270-1282
  109. 109. Hariharan S, Minocha M, Mishra GP, Pal D, Krishna R, Mitra AK. Interaction of ocular hypotensive agents (PGF2α analogs—bimatoprost, latanoprost, and travoprost) with MDR efflux pumps on the rabbit cornea. Journal of Ocular Pharmacology and Therapeutics. 2009;25(6):487-498
  110. 110. Majumdar S, Hingorani T, Srirangam R, Gadepalli RS, Rimoldi JM, Repka MA. Transcorneal permeation of L-and D-aspartate ester prodrugs of acyclovir: Delineation of passive diffusion versus transporter involvement. Pharmaceutical Research. 2009;26(5):1261-1269
  111. 111. Anand B, Katragadda S, Nashed Y, Mitra A. Amino acid prodrugs of acyclovir as possible antiviral agents against ocular HSV-1 infections: Interactions with the neutral and cationic amino acid transporter on the corneal epithelium. Current Eye Research. 2004;29(2-3):153-166
  112. 112. Jain-Vakkalagadda B, Pal D, Gunda S, Nashed Y, Ganapathy V, Mitra AK. Identification of a Na+−dependent cationic and neutral amino acid transporter, B0,+, in human and rabbit cornea. Molecular Pharmaceutics. 2004;1(5):338-346
  113. 113. Anand BS, Mitra AK. Mechanism of corneal permeation of L-valyl ester of acyclovir: Targeting the oligopeptide transporter on the rabbit cornea. Pharmaceutical Research. 2002;19(8):1194-1202
  114. 114. Gunda S, Hariharan S, Mitra AK. Corneal absorption and anterior chamber pharmacokinetics of dipeptide monoester prodrugs of ganciclovir (GCV): In vivo comparative evaluation of these prodrugs with Val-GCV and GCV in rabbits. Journal of Ocular Pharmacology & Therapeutics. 2006;22(6):465-476
  115. 115. Majumdar S, Nashed YE, Patel K, Jain R, Itahashi M, Neumann DM, et al. Dipeptide monoester ganciclovirprodrugs for treating HSV-1-induced corneal epithelial and stromal keratitis: In vitro and in vivo evaluations. Journal of Ocular Pharmacology & Therapeutics. 2005;21(6):463-474
  116. 116. Katragadda S, Talluri RS, Mitra AK. Modulation of P-glycoprotein–mediated efflux by prodrugderivatization: An approach involving peptide transporter–mediated influx across rabbit cornea. Journal of Ocular Pharmacology & Therapeutics. 2006;22(2):110-120
  117. 117. Kansara V, Hao Y, Mitra AK. Dipeptide monoester ganciclovirprodrugs for transscleral drug delivery: Targeting the oligopeptide transporter on rabbit retina. Journal of Ocular Pharmacology and Therapeutics. 2007;23(4):321-334
  118. 118. Janoria KG, Boddu SH, Wang Z, Paturi DK, Samanta S, Pal D, et al. Vitreal pharmacokinetics of biotinylatedganciclovir: Role of sodium-dependent multivitamin transporter expressed on retina. Journal of Ocular Pharmacology and Therapeutics. 2009;25(1):39-49
  119. 119. Vadlapudi AD, Vadlapatla RK, Earla R, Sirimulla S, Bailey JB, Pal D, et al. Novel biotinylated lipid prodrugs of acyclovir for the treatment of herpetic keratitis (HK): Transporter recognition, tissue stability and antiviral activity. Pharmaceutical Research. 2013;30(8):2063-2076
  120. 120. Mandal A, Agrahari V, Khurana V, Pal D, Mitra AK. Transporter effects on cell permeability in drug delivery. Expert Opinion on Drug Delivery. 2017;14(3):385-401
  121. 121. Vellonen KS, Soini EM, Del Amo EM, Urtti A. Prediction of ocular drug distribution from systemic blood circulation. Molecular Pharmaceutics. 2016;13(9):2906-2911
  122. 122. Boddu SH, Gunda S, Earla R, Mitra AK. Ocular microdialysis: A continuous sampling technique to study pharmacokinetics and pharmacodynamics in the eye. Bioanalysis. 2010;2(3):487-507
  123. 123. Danesh-Meyer HV, Zhang J, Acosta ML, Rupenthal ID, Green CR. Connexin43 in retinal injury and disease. Progress in Retinal and Eye Research. 2016;51:41-68
  124. 124. Kang-Mieler JJ, Rudeen KM, Liu W, Mieler WF. Advances in ocular drug delivery systems. Eye. 2020;34(8):1371-1379
  125. 125. Michael IP. Development of Novel Antiangiogenic Biologics. Canada: University of Toronto; 2011
  126. 126. Cholkar K, Dasari SR, Pal D, Mitra AK. Eye: Anatomy, physiology and barriers to drug delivery. In: Ocular Transporters and Receptors. Woodhead Publishing; 2013. pp. 1-36
  127. 127. Cholkar K, Trinh HM, Vadlapudi AD, Wang Z, Pal D, Mitra AK. Interaction studies of resolvin E1 analog (RX-10045) with efflux transporters. Journal of Ocular Pharmacology and Therapeutics. 2015;31(4):248-255
  128. 128. Juuti-Uusitalo K, Vaajasaari H, Ryhänen T, Narkilahti S, Suuronen R, Mannermaa E, et al. Efflux protein expression in human stem cell-derived retinal pigment epithelial cells. PLoS One. 2012;7(1):e30089
  129. 129. Mannermaa E, Vellonen KS, Ryhänen T, Kokkonen K, Ranta VP, Kaarniranta K, et al. Efflux protein expression in human retinal pigment epithelium cell lines. Pharmaceutical Research. 2009;26(7):1785-1791
  130. 130. Shah R, Amador C, Tormanen K, Ghiam S, Saghizadeh M, Arumugaswami V, et al. Systemic diseases and the cornea. Experimental Eye Research. 2021;204:108455
  131. 131. Sánchez-López E, Espina M, Doktorovova S, Souto EB, García ML. Lipid nanoparticles (SLN, NLC): Overcoming the anatomical and physiological barriers of the eye—Part I—Barriers and determining factors in ocular delivery. European Journal of Pharmaceutics and Biopharmaceutics. 2017;110:70-75
  132. 132. Bhujbal SS, Wale KK, Mahale NB, Landage AD, Sayyed SF, Chaudhari SR. Ocular drug delivery system: A review. World Journal of Pharmaceutical Research, RamaiyanDhanapal. 2014;3:320-343
  133. 133. Chien YW. Ocular drug delivery and delivery systems. In: Novel Drug Delivery Systems. Vol. 2nd ed. New York: Marcel Dekker; 1992. pp. 269-270
  134. 134. Saettone MF, Salminen L. Ocular inserts for topical delivery. Advance Drug Delivery Review. 1995;16:95-106
  135. 135. Khar RK, Vyas SP. Targeted and Controlled Drug Delivery Novel Carrier Systems. New Delhi: CBS Publishers and Distributors; 2002. p. 384

Written By

Prakriti Diwan, Rajendra Jangde, Sulekha Khunte, Harish Bhardwaj and Preeti K. Suresh

Submitted: 15 February 2022 Reviewed: 13 May 2022 Published: 07 October 2022