throbber
Drug Discovery Today  Volume 21, Number 6  June 2016
`
`REVIEWS
`
`Given that cyclosporine (CsA) is poorly water soluble and currently marketed
`products are not well tolerated, novel approaches for safe and efficient CsA
`delivery to the eye are of great interest.
`
`ReviewsKEYNOTEREVIEW
`
`Modern approaches to the ocular
`delivery of cyclosporine A
`
`Priyanka Agarwal and Ilva D. Rupenthal
`
`Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of
`Medical and Health Sciences, University of Auckland, New Zealand
`
`Cyclosporine A (CsA) has long been the mainstay treatment for dry eye
`syndrome (DES), one of the most common disorders of the eye. However,
`the poor water solubility of CsA renders it difficult to formulate it into
`
`topical ocular dosage forms. Restasis1 is currently the only US Food and
`Drug Administration (FDA)-approved CsA formulation, while Ikervis1
` has
`recently been launched in Europe, with both commonly associated with
`severe ocular discomfort. Therefore, several CsA formulations have been
`investigated with the aim to improve bioavailability while reducing
`adverse effects associated with the marketed formulations. In this review,
`we summarize recent advances in ocular CsA delivery that provide safer
`and more effective alternatives for the management of DES and other
`ocular inflammatory conditions.
`
`Introduction
`CsA is a metabolite of the fungi Tolypocladium inflatum and Beauveria nevus that was initially
`suggested for use as an antifungal agent. Its immunosuppressive activity soon became evident
`and, because of the reduced incidence of associated myelotoxicty, CsA eventually became the
`mainstay treatment after organ transplantation [1,2]. Systemic CsA is still used, although to a
`lesser extent, to treat several other autoimmune diseases, including those with eye involvement
`[3]. CsA is the preferred immunomodulatory agent for topical treatment of several immune-
`mediated ocular surface disorders [4], and its prevalence in the treatment of these ocular disorders
`is second only to corticosteroids, whose adverse effects are well documented [5]. CsA therapy has
`been approved for the treatment of keratoconjunctivitis sicca (KCS), more commonly known as
`dry eye syndrome (DES). It is also frequently used off-label to treat several other ophthalmic
`conditions, such as posterior blepharitis [6,7], ocular rosacea [8,9], vernal keratoconjunctivitis
`[10–14], atopic keratoconjunctivitis [15–17], acute corneal graft rejection [18], and conjunctival
`graft versus host disease [19–21].
`DES is one of the most prevalent ocular surface disorders, and is usually characterized by
`increased evaporation or decreased production of tear fluid, resulting in damage to the inter-
`palpebral ocular surface and moderate to severe discomfort [22]. Symptoms of DES have been
`reported in approximately one out of seven individuals above the age of 48, with its prevalence
`nearly doubling after 59 years of age [23–25]. A recent study estimated that nearly 20% of
`
`Priyanka Agarwal is
`currently pursuing a
`doctorate within the
`Buchanan Ocular
`Therapeutics Unit,
`Department of
`Ophthalmology, University
`of Auckland, and is
`investigating novel
`cyclosporine A (CsA) formulations for topical
`administration. She completed her BPharm at the
`University of Mumbai, India, and subsequently
`obtained a Postgraduate Diploma in Health Sciences
`from the School of Pharmacy at the University of
`Auckland. Priyanka has extensive industrial research
`experience in formulation development and
`pharmacokinetics and worked as a Veterinary
`Formulation Development Scientist for several years.
`During her time in industry, she primarily worked on
`the development of new platforms for safe drug
`delivery to animals and she is currently one of the
`inventors on a series of patents filed in this field.
`
`Ilva Rupenthal is a senior
`lecturer in the Department
`of Ophthalmology, New
`Zealand National Eye
`Center, University of
`Auckland, and the
`inaugural Director of the
`Buchanan Ocular
`Therapeutics Unit, which
`aims to translate ocular therapeutic-related scientific
`research into the clinical setting, whether
`pharmaceutical, cell, or technology based. Her
`current research, funded by a Sir Charles Hercus
`Health Research Fellowship from the New Zealand
`Health Research Council, focuses mainly on the
`development of stimuli-response devices, with
`projects investigating ocular implants responsive to
`light or a small electrical current. Moreover, Dr
`Rupenthal’s group is developing tailored controlled
`delivery systems that specifically target the drug to the
`site of action with projects around dry eye, optic
`neuropathy, diabetic retinopathy, and age-related
`macular degeneration treatment.
`
`Corresponding author: Rupenthal, I.D.
`
`(i.rupenthal@auckland.ac.nz)
`
`1359-6446/ß 2016 Elsevier Ltd. All rights reserved.
`
`http://dx.doi.org/10.1016/j.drudis.2016.04.002
`
`www.drugdiscoverytoday.com 977
`
`1
`
`ALL 2011
`MYLAN PHARMACEUTICALS V. ALLERGAN
`IPR2016-01128
`
`

`

`REVIEWS
`
`Drug Discovery Today  Volume 21, Number 6  June 2016
`
`N
`
`O
`
`HN
`
`NH
`
`O
`
`O
`
`O
`
`N
`
`O
`
`HN
`
`O
`
`N
`
`O
`
`N
`
`N
`
`O
`
`Drug Discovery Today
`
`N
`
`O
`
`N
`
`O
`
`HN
`
`OH
`O
`
`FIGURE 1
`Molecular structure of cyclosporine A (CsA), a cyclic undecapeptide with very
`low aqueous solubility.
`
`neutrally charged and hydrophobic molecule (Fig. 1) with an
`aqueous solubility of less than 10 mg/ml (0.001%) at physiological
`temperature [42], formulation of aqueous eye drops at these con-
`centrations is difficult. Attempts have been made to improve the
`aqueous solubility of CsA using surfactants and/or penetration
`enhancers, such as macrogolglycerol ricinoleate (Cremophor
`EL1) and benzalkonium chloride, with the latter also commonly
`used as a preservative in ocular formulations [43]. Although these
`excipients can improve the solubility and penetration of CsA, their
`use is limited by their high irritation potential because these
`molecules typically function by compromising the integrity of
`the ocular tissues [44–46].
`CsA solutions in vegetable oils were considered as the next best
`alternative for topical administration to the eye and concentra-
`tions as high as 2% could be achieved [47,48]. Despite their poor
`absorption, oily CsA solutions have demonstrated success in the
`treatment of DES by improving tear production and inducing
`in a canine model
`regression of corneal neovascularization
`[49–54]. Similar results were also observed in humans after topical
`application of 2% CsA in olive oil [55,56]. However, one of the
`major limitations of vegetable oils in ophthalmic preparations is
`the increased incidence of ocular toxicity after frequent use, with
`blurring of vision also commonly observed because of the high
`viscosity of the carrier oils. BenEzra et al. [38,57,58] showed that
`penetration of CsA from olive oil drops was negligible in normal or
`slightly inflamed eyes, but increased significantly over time be-
`cause the corneal barrier was compromised as a result of toxic
`effects of the oily vehicle. Thus, the use of topical CsA oily drops
`for the management of ophthalmic conditions has largely been
`discontinued.
`CsA ointments [59–61] and oil-in-water (o/w) emulsions [62]
`have also been used to reduce the toxicity associated with oily
`solutions, although they have not been able to eliminate it
`completely. Currently, Restasis1
` (Allergan) is the only CsA for-
`mulation approved by the FDA for DES therapy in humans. It is a
`0.05% CsA emulsion of castor oil in water and is often associated
`with severe adverse effects, such as ocular burning (most com-
`mon), conjunctival hyperemia, discharge, epiphora, eye pain,
`
`hospitalized patients above the age of 50 had DES, with old age and
`illiteracy being major predictors of the disorder [26]. On the
`in
`recommendation of the International Dry Eye Workshop
`2007, DES was classified as a multifactorial disease of the tears
`and ocular surface that can be triggered by a variety of underlying
`causes. However, recently, there has been increasing evidence that
`inflammation has a key role in the manifestation of DES. Ocular
`surface abnormalities, such as the appearance of inflammatory cell
`intermediates in the lacrimal gland and an increase in immune-
`related antigens and cytokines at the conjunctival epithelium,
`are commonly demonstrated by both autoimmune (Sjo¨gren’s
`(non-Sjo¨gren’s syndrome)-
`syndrome) and non-autoimmune
`mediated DES [27–30], making treatment of the underlying cyto-
`inflammatory processes
`the primary
`kine–receptor-mediated
`‘causative therapeutic approach’ [31,32]. A combination of symp-
`tomatic therapy, which includes modification of the ocular envi-
`ronment (by increasing humidity, occlusion of lacrimal canaliculi,
`or simulation of tears), and pathogenic treatments, including the
`use of antibacterial and anti-inflammatory agents (corticosteroids,
`antihistamines, tetracyclines, and CsA), is currently recommended
`for DES therapy [32].
`CsA is the anti-inflammatory agent of choice for the treatment
`of DES, because it can be used long term without adverse effects
`commonly associated with other anti-inflammatory agents, such
`as steroids [5]. Furthermore, unlike corticosteroids, the activity of
`CsA results from specific and reversible action on T cells, making it
`safe for prolonged use. For example, corneal epitheliopathy and
`eyelid maceration associated with long-term CsA therapy were
`found to be reversible with complete cessation on discontinuation
`of the drug [33]. The immunomodulatory activity of CsA helps in
`reducing the inflammation associated with subconjunctival and
`lacrimal glands, resulting in increased goblet cell density and tear
`production [34,35]. CsA tends to bind with specific nuclear pro-
`teins that initiate the activation of T cells, thus preventing T cell
`production of inflammatory cytokines and disrupting the im-
`mune-mediated inflammatory response [31,36]. CsA is a hydro-
`phobic molecule and, therefore, is difficult to formulate into
`conventional topical ocular delivery systems. Significant research
`has been performed over recent years to develop safe and effective
`ocular delivery systems for CsA. In this review, we highlight recent
`efforts to improve the ophthalmic delivery of CsA in terms of its
`bioavailability and ocular tolerability while reducing adverse
`effects.
`
`Conventional CsA formulations
`Systemic CsA is generally not considered for the treatment of
`ocular pathologies because of severe systemic adverse effects, such
`as nephrotoxicity and hypertension [2,37], although significant
`concentrations of CsA have been reported in tears and lacrimal
`glands after oral administration [38]. Thus, the topical route is
`generally preferred because, as well as reducing systemic adverse
`effects, it also helps to achieve improved bioavailability and spe-
`cific targeting to the ocular tissues [39,40].
`Dose-ranging randomized clinical trials have shown that topi-
`cally applied CsA is effective at concentrations between 0.05 and
`0.1% (w/v). No additional benefits were observed at higher con-
`centrations; hence, clinical trials are generally recommended at a
`maximum concentration of 0.1% [4,41]. However, because CsA is a
`
`978 www.drugdiscoverytoday.com
`
`Reviews  KEYNOTE REVIEW
`
`2
`
`

`

`Drug Discovery Today  Volume 21, Number 6  June 2016
`
`REVIEWS
`
`*
`
`*
`
`120
`
`100
`
`20
`
`60
`
`40
`
`20
`
`*
`
`Cornea
`
`Conjunctiva
`
`0
`
`Aqueous
`humor
`
`CsA in oil 0.50% formulation
`Prodrug in water 0.50% formulation
`
`ReviewsKEYNOTEREVIEW
`
`Iris-Ciliary
`body
`
`
`*
`
`*
`
`Vitreous
`humor
`
`Retina
`
`Converted CsA
`Unconverted CsA
`
`Drug Discovery Today
`
`8000
`
`7000
`
`6000
`
`5000
`
`4000
`
`3000
`
`2000
`
`1000
`
`0
`
`
`
`Total CsA (ng/g)
`
`FIGURE 2
`Ocular distribution of cyclosporine A (CsA) in oil and CsA prodrug in water.
`Prodrug formulations significantly increased the amount of CsA absorbed by,
`and accumulated in, the cornea and conjunctiva; however, no improvement
`was observed in the penetration of CsA across the cornea.
`Source: Adapted from [75].
`
`a significant
`in DES
`in basal tear production
`improvement
`[71,72]. Aqueous prodrug solutions have also been evaluated for
`their efficacy in the treatment of corneal graft rejection and it was
`found that prodrug eye drops applied five times a day were
`therapeutically equivalent to a 10 mg/kg/day intramuscular injec-
`tion in rats [73]. Recent studies have shown that 2% prodrug
`solutions have a 200–500-fold higher conjunctival permeability
`than the conventional 2% CsA in oil formulation. Accumulation
`of CsA prodrug formulations in the cornea to form large tissue
`deposits that provide a sustained release effect over prolonged
`periods of time was also observed. However, prodrug formulations
`did not show much improvement in the permeability across the
`cornea and into the aqueous humor compared with the conven-
`tional CsA emulsion, probably because of the rapid conversion of
`the prodrug into CsA at the corneal surface (Fig. 2). This depot
`formation could have the added advantage of overall poor system-
`ic absorption of prodrug formulations, reducing the incidence of
`systemic complications and immunosuppression. Prodrug formu-
`lations with CsA concentrations higher than 2% are currently
`under investigation for safety and toxicity [74,75].
`
`Novel application of excipients
`Several new excipients have been introduced to improve CsA
`solubility in ocular formulations and enhance their bioavailability
`(Table 1).
`Cyclodextrins
`The introduction of cyclodextrins (CDs) in ophthalmic drug de-
`livery is relatively recent; however, they have rapidly gained
`popularity because of their pronounced benefits in drug solubili-
`zation and stabilization. CDs are cyclic oligosaccharides of six
`(a-CD), seven (b-CD), or eight (g-CD) glucopyranose units with
`a hydrophobic cavity in the center. In aqueous solutions, water
`from the hydrophobic cavity is displaced by hydrophobic mole-
`cules, resulting in the formation of large water-soluble complexes.
`CDs have been used on several occasions to prepare aqueous CsA
`
`www.drugdiscoverytoday.com 979
`
`foreign body sensation, pruritus, stinging, and visual disturbance
`[63]. Devecı et al. [64] recently showed that ocular irritation
`associated with Restasis usually decreases within 1 week of con-
`tinuous therapy and significant resolution of DES can be observed
`at the 1-month follow-up. A veterinary ointment of 0.2% CsA
`(Optimmune1, Schering-Plough Animal Health) has also been
`approved for DES therapy in dogs and, although it has been shown
`to resolve the symptoms of DES, associated ocular toxicity, proba-
`bly because of the oily base, is significant [65,66]. Ointments are
`generally more viscous and have a cloudy appearance and, thus,
`tend to blur the vision, which, in addition to their tolerability
`issues, reduces patient acceptability. Recently, a cationic nanoe-
`mulsion containing 0.1% CsA was launched in Europe for the
`treatment of severe DES under the brand name Ikervis1
` (Santen).
`Unlike emulsions and ointments, this system does not cloud
`vision because of its low viscosity; however, adverse effects, such
`as stinging and pain, have frequently been reported [67].
`The limitations of currently approved formulations leave tre-
`mendous scope for the development of improved ocular formula-
`tions for the safe and efficient delivery of CsA to the eye. The focus
`of new CsA technologies has predominantly been the improve-
`ment of solubility, transcorneal penetration, and precorneal resi-
`dence time, with a simultaneous reduction in the frequency of
`dosing and irritation potential. Such CsA formulations could
`significantly improve patient comfort and compliance, thus in-
`creasing the quality of life of patients with DES.
`
`Recent approaches in CsA delivery
`Over the past few years, several novel strategies have been sug-
`gested for delivering CsA to the ocular tissues, with topical,
`episcleral, subconjunctival, and intravitreal routes being investi-
`gated. Of these, topical administration remains the most preferred
`route for the treatment of DES, because it is non-invasive, painless,
`and convenient. For the purpose of this review, the key approaches
`to improving ocular drug delivery of CsA have been classified into
`three major areas of research: (i) chemical modification of the
`drug; (ii) novel application of excipients; and (iii) novel ophthal-
`mic dosage forms.
`
`Chemical modification of the drug
`Reversible chemical modification of CsA to obtain prodrugs with
`improved aqueous solubility was first suggested by Hammel et al.
`[68] when they coupled CsA with diketopiperazine to synthesize
`dipeptide esters with improved oral bioavailability. Monomethox-
`y(polyethyleneglycol) derivatives of CsA have also been suggested
`for improving solubility and absorption of CsA in oral prepara-
`tions. However, esterification of the free hydroxyl group of CsA
`with a solubilizing moiety is the generally accepted approach for
`prodrug synthesis for ocular applications. Lallemand et al. [69,70]
`developed a series of amphiphilic acidic prodrug molecules (pat-
`ented by DeBiopharm, Switzerland) having an approximately
`25 000 times higher solubility than CsA in isotonic phosphate
`buffer solution (PBS) at pH 7. These prodrugs are quantitatively
`hydrolyzed in artificial tears to release CsA within 1 min. Prodrug
`conversion into the parent molecule was significantly faster in tear
`fluid than in a buffer at physiological pH, indicating that the
`hydrolysis is enzyme mediated. Aqueous formulations of these
`esterified CsA prodrugs were well tolerated and have shown
`
`3
`
`

`

`REVIEWS
`
`Drug Discovery Today  Volume 21, Number 6  June 2016
`
`TABLE 1
`Novel excipients used in ocular CsA delivery systems.
`
`Excipients
`
`Dosage form
`
`Advantages
`
`Limitations
`
`Cyclodextrins
`
`Aqueous eye drops
`
`Solubilize CsA in aqueous formulations; show
`improved bioavailability and low toxicity at
`optimized concentrations
`
`Cannot be used in concentrated
`solutions; ocular toxicity can be
`observed at higher concentrations
`because of complexation of
`cellular components
`
`Refs
`
`[76–79]
`
`Semifluorinated
`alkanes
`
`Eye drops (CyclASolW)
`
`Safe and well tolerated; solubilize hydrophobic
`drugs to form clear eye drops; improved
`penetration; no preservative required;
`lubricating nature
`
`Cationic vectors
`(amines, chitosan,
`poly-L-lysine, Eudragit)
`
`Emulsions, liposomes,
`nanoparticles
`
`Improve precorneal residence time because
`of mucoadhesion
`
`No sustained release effect
`
`[80,81]
`
`Amine vectors can cause stability
`problems; safety and toxicity is a
`concern because ocular irritation
`is usually observed with most
`cationic moieties
`
`[95]
`
`Reviews  KEYNOTE REVIEW
`
`eye drops with significant improvement in transcorneal penetra-
`tion and retention [76–79]. a-CDs have been found to be the most
`efficient in solubilizing CsA, because the cavity size is a good fit for
`the cyclic CsA molecule. When used to prepare concentrations
`below 0.025%, a-CDs improved CsA bioavailability without dem-
`onstrating any toxic effects in vivo [77,79].
`Semi-fluorinated alkanes (SFAs)
`SFAs are a class of amphiphilic fluorinated compounds that are
`physically, chemically, and biologically inert and capable of dis-
`solving several hydrophobic drugs. Although they have been used
`intraocularly as intravitreal tamponades for several years, their use
`in drug delivery to the front of the eye is relatively recent [80].
`Given their established safety profile, excellent spreading proper-
`ties, and ability to solubilize and stabilize several hydrophobic
`drugs, SFAs can be used as suitable carriers for ocular preparations.
`The added advantages of SFAs include their ability to form clear
`solutions that do not cause any blurring of vision or ocular
`discomfort while also providing lubrication to the ocular surface,
`which is particularly useful in DES treatment. Moreover, because
`these solutions are nonaqueous, they do not require any preser-
`vatives, surfactants, or pH modifiers, which are frequently impli-
`cated in ocular toxicity. Recent studies showed that SFA-based
`solutions containing 0.05% CsA were well tolerated and signifi-
`cantly increased CsA penetration across the cornea and into the
`aqueous humor when compared with Restasis, rendering SFA-
`based formulations promising candidates for the treatment of
`ocular inflammatory conditions [81]. This technology is currently
`registered as CyclASol1
` (Novaliq GmbH, Germany) and recently
`conducted Phase I clinical trials have shown promising results
`[82], with the company now recruiting patients for a Phase II
`clinical trial [83].
`Positively charged vectors
`One of the major objectives of topical CsA delivery has been to
`improve ocular bioavailability of the drug by increasing corneal
`penetration and precorneal residence time. Therefore, positively
`charged ocular formulations have received much interest because
`of their higher mucoadhesion and, thus, longer precorneal resi-
`dence time. Studies performed by Daull et al. [84] showed that
`cationic emulsions have significantly greater bioavailability than
`Restasis, which is an anionic o/w emulsion. Safety profiles of
`
`980 www.drugdiscoverytoday.com
`
`cationic and anionic emulsions were further compared and, while
`their tolerability was similar, only the cationic emulsion was able to
`maintain the normal healing rate of the human corneal epithelium
`in vitro and reduce inflammation in vivo [85]. Similar to most epithe-
`lia, corneal and conjunctival cells are negatively charged at physio-
`logical pH and, therefore, cations can adhere to and penetrate them
`more easily [86,87]. Their cell membranes are further coated with a
`layer of mucin containing negatively charged sialic acid groups,
`which develop electrostatic interactions with positively charged
`vectors and thus improve the precorneal residence time [88].
`Stearylamine has been used extensively to impart a positive
`charge to liposomes [89–91] and ocular emulsions [92–94] to
`increase mucoadhesion and, thus, retention. Oleylamine is anoth-
`er cationic lipid that has been used for preparation of cationic
`ophthalmic emulsions [88]. However, a major disadvantage of
`these amines is their poor stability and compatibility with other
`excipients, while ocular tolerability of amines remains a signifi-
`cant concern. Hence, chitosan has emerged as the cationic agent of
`choice in ocular formulations. Chitosan is a biodegradable and
`linear polysaccharide with
`several positively
`biocompatible
`charged free amino groups that interact with mucin. There is
`some evidence that, in addition to the positive charge, the specific
`nature of chitosan might also be responsible for improving the
`uptake of drug molecules. Calvo et al. [95] showed that the
`bioavailability of nanocapsules coated with chitosan was at least
`twofold higher than for poly-L-lysine-coated nanocapsules. How-
`ever, commercialization of chitosan formulations might be diffi-
`cult, because raw materials of natural origin often demonstrate
`high batch-to-batch variability. Recently, several studies also
`reported that, despite its biodegradability, chitosan can display
`some toxicity, especially when administered as nanoparticles
`[96,97], while chitosan hydrogels of a certain molecular weight
`have also shown to initiate an inflammatory response and, thus,
`delay wound closure in a corneal scrape wound model [98]. There-
`fore, Eudragit1, a synthetic polymer frequently used for the prepa-
`ration of enteric oral dosage forms, has been suggested as an
`alternative for ophthalmic formulations [99]. Eudragit is a cationic
`copolymer based on dimethylaminoethyl methacrylate, butyl
`methacrylate, and methyl methacrylate groups. Eudragit-based
`ocular colloidal formulations have shown good tolerability and
`
`4
`
`

`

`Drug Discovery Today  Volume 21, Number 6  June 2016
`
`TABLE 2
`Novel ophthalmic dosage forms for CsA delivery to the front of the eye.
`
`Drug delivery system
`
`Advantages
`
`Limitations
`
`REVIEWS
`
`Refs
`
`[103–106]
`
`Improved uptake into all layers of the cornea;
`sustained release over extended periods
`
`Generally poor shelf life; irritation potential and
`toxicity can be significant
`
`ReviewsKEYNOTEREVIEW
`
`Form depots in cornea for sustained drug
`release over prolonged periods
`Improved stability and simple manufacturing
`procedure
`
`Short half-life at the corneal surface results in
`poor transcorneal penetration
`Safety and irritation potential need to be assessed
`
`[113,114,118]
`
`[119]
`
`Improved retention and uptake into cornea;
`reduced drug toxicity can be observed
`Improved uptake because of transport by
`transcellular pathway
`Improved bioavailability; improved shelf-life
`compared with nanoemulsions
`
`Difficult to scale up; can show some long-term
`toxicity
`Generally poor stability and manufacturability
`
`Generally poor long-term stability and
`manufacturability
`
`[133,134]
`
`[142]
`
`[143–145]
`
`Increased precorneal residence time and
`sustained release
`Sustained release of the drug over prolonged
`periods
`
`High burst release; blurring of vision can reduce
`patient compliance
`Discomfort and foreign body sensation; adverse
`effects because of poor oxygen permeability
`
`[154,157,160]
`
`[164,165,171]
`
`Micelles
`
`Liposomes
`Liposomes
`
`Proliposomes
`
`Nanoparticles
`Nanospheres/nanocapsules
`
`Nanoemulsions
`
`Solid lipid nanoparticles
`
`Other
`In situ gelling systems
`
`Hydrogels
`
`low incidence of ocular irritation [100–102], while decomposition
`of Eudragit is typically slow, providing controlled drug release with a
`low burst effect.
`
`Novel ophthalmic dosage forms
`The field of ocular therapeutics has seen the development of
`several novel delivery systems in the form of micelles, liposomes,
`nanoparticles, in situ gelling systems, and hydrogels, which have
`also been evaluated for the delivery of CsA (Table 2 and Fig. 3).
`
`Micelles
`Micelles are self-assembling spherical colloidal systems that are
`frequently used for the solubilization of hydrophobic molecules.
`Typically, micelles are formed around a hydrophobic drug in
`aqueous solution because of the orientation of surfactant mole-
`cules to form a hydrophobic core enclosing the drug within a
`hydrophilic shell. N-Octyl chitosan has been used as a surfactant
`for preparation of CsA micelles for DES therapy [103]. CsA uptake
`from these micelles was found to be significantly higher than from
`
`In situ gelling systems
`
`+ Improved retention
`+ Sustained release
`
`- Blurring
`- Burst release
`
`Hydrogels
`+ Improved retention
`+ Sustained release
`
`- Patient discomfort
`- Poor oxygen permeability
`
`Micelles
`+ Improved absorption
`+ Deeper penetration
`
`- Surface toxicity
`- Poor stability
`
`Liposomes
`+ Corneal accumulation
`+ Sustained release
`
`- Poor scalability
`- Low entrapment
`
`Nanoparticles
`+ Improved penetration
`+ Improved absorption
`
`- Poor scalability
`- Poor stability
`
`Drug Discovery Today
`
`FIGURE 3
`Schematic representation of novel ophthalmic dosage forms used to improve the bioavailability of topically applied cyclosporine A (CsA).
`
`www.drugdiscoverytoday.com 981
`
`5
`
`

`

`REVIEWS
`
`Drug Discovery Today  Volume 21, Number 6  June 2016
`
`from a saturable interaction with mucin [116] and, thus, concen-
`trations could be optimized to achieve maximal mucoadhesion
`with minimal toxicity. Li et al. [117] prepared liposomes coated
`with low-molecular-weight chitosan and observed a significant
`improvement in the ocular bioavailability of CsA. The number of
`free positively charged groups available in low-molecular-weight
`chitosan is considerably reduced, resulting in better ocular tolera-
`bility of these formulations.
`Despite their many benefits, the fast degradation on the corneal
`surface, poor entrapment efficacy, and difficulty in manufacturing
`sterile formulations at a large scale are disadvantageous to the use
`of liposomal formulations. To counter these problems, Karn et al.
`[118] developed CsA-loaded multilamellar liposomes from egg
`lecithin and phosphatidyl choline from soybean using supercriti-
`cal fluid of carbon dioxide as the antisolvent. Liposomes, thus
`prepared, showed better stability on the corneal surface, had an
`improved shelf life, and were easy to manufacture on a larger scale.
`Considerable improvements were also seen in the precorneal
`residence time, corneal absorption, and tolerance in comparison
`to Restasis [112]. Recently, the same method was also used to
`prepare proliposomes [119], which are dry free-flowing powders
`that assemble spontaneously in an aqueous medium to form
`liposomes. These proliposomes had excellent stability and were
`easy to scale up. However, some additional studies might be
`required to establish their efficacy in ocular CsA delivery.
`Nanoparticles
`A large body of research has also been dedicated to the develop-
`ment of nanoparticle-based systems for targeted CsA delivery to
`the eye. Nanoparticles are submicron particles ranging from 10 to
`1000 nm in size that can have drug molecules dissolved, adsorbed,
`or entrapped in the nanoparticle matrix. Nanoparticles have been
`shown to accelerate drug penetration and significantly improve
`corneal absorption of drugs, primarily by the transcellular path-
`way [120–123]. They have also been reported to decrease the
`incidence of ocular irritation and toxicity because of their small
`particle size [124,125]. Moreover, because they are similar to
`aqueous eye drops in appearance and viscosity, nanoparticle for-
`mulations are easy to handle and ensure convenient ocular appli-
`cation [126–128]. However, despite their advantages, products
`based on nanotechnology rarely reach the market [129] as a result
`of technical issues involved in scale up and manufacturing, use of
`new excipients or nonpharmacopoeial solvents [130], as well as
`the relatively poor stability of these colloidal systems [131].
`CsA nanosuspensions have been prepared using zirconia beads
`in a water/polyvinyl alcohol system and were found to show
`significantly lower irritation compared with Restasis [132]. How-
`ever, biodegradable polymers, such as poly-D,L-lactide-co-glycolide
`(PLGA) and poly-e-caprolactone-co-lactide (PCL), are usually pre-
`ferred for ophthalmic applications because of their established
`safety profile. PLGA nanoparticles prepared by solvent evaporation
`have shown a 11–16% drug-loading capacity and have been used
`for sustained release of CsA for up to 65 days [133]. It was found
`that coating these nanoparticles with a surface-modifying poly-
`mer, such as PEG, could influence the penetration behavior of CsA
`across the cornea. PCL nanoparticles have also been used for ocular
`CsA delivery in conjugation with penetration-enhancing surfac-
`tants, such as benzalkonium chloride [134]. Although the bio-
`availability from these nanospheres was 10–15 times higher than
`
`conventional CsA oil drops, and the bioavailability could be
`further increased by coating the micelles with carbopol to give
`them mucoadhesive properties. Penetration of micellar formula-
`tions can also be enhanced by reducing the size of the micelles. In a
`recent study using an optimized blend of hydrogenated castor oil-
`40 and octoxynol-40, Cholkar et al. [104] prepared a clear aqueous
`nanomicellar solution containing 0.025–0.1% CsA with an aver-
`age particle size of 22.4 nm. High corneal and conjunctival levels
`were observed with low ocular toxicity. One of the most significant
`findings of this study was that this formulation could also deliver
`CsA to the posterior segment of the eye, with as much as 53.7 ng/g
`of CsA recovered from the retina/choroid in vivo. Polyvinyl capro-
`lactam–polyvinyl acetate–polyethylene glycol nanomicelles have
`also shown improved penetration of CsA along with excellent
`ocular tolerability and safety [105]. CsA uptake from these micelles
`is predicted to be an energy-dependent intracellular pathway,
`leading to improved efficacy in vivo. Recently, in vivo studies
`performed with nano-sized micellar systems based on methoxy
`poly(ethylene) glycol-hexyl substituted poly(lactide) (mPEGhex-
`PLA) copolymers showed increased uptake and prolonged release
`of CsA because of better penetration into, and accumulation in, all
`layers of the cornea [106]. Significant advantages of this mPE-
`GhexPLA nanomicellar system included the absence of ocular
`toxicity and a longer shelf life compared with conventional mi-
`celle systems [107,108]. Further improvement in shelf life by
`lyophilization of mPEGhexPLA micellar systems has also been
`suggested; however, long-term studies still need to be performed
`to verify this [109]. Poor long-term stability is the major drawback
`of most micelle systems. Therefore, in an attempt to overcome this
`limitation, CsA-loaded self-assembling micellar systems were pre-
`pared using two PEG-fatty alcohol ether-type surfactants [110].
`The micellar formulation had good entrapment efficiency and
`showed a threefold increase in corneal CsA concentration com-
`pared with Restasis. However, the toxicity and irritation potential
`of this formulation still needs to be evaluated.
`Liposomes
`Over

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket