throbber
ARTICLE IN PRESS
`
`European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`www.elsevier.com/locate/ejpb
`
`Review article
`
`Cyclosporine A delivery to the eye: A pharmaceutical challenge
`
`F. Lallemanda, O. Felt-Baeyensa, K. Besseghirb, F. Behar-Cohenc, R. Gurnya,*
`
`aSchool of Pharmacy, University of Geneva, Geneva, Switzerland
`bDebiopharm S.A., Lausanne, Switzerland
`cINSERM U450, Paris, France
`
`Received 5 February 2003; accepted in revised form 5 August 2003
`
`Abstract
`
`Systemic administration of cyclosporine A (CsA) is commonly used in the treatment of local ophthalmic conditions involving cytokines,
`such as corneal graft rejection, autoimmune uveitis and dry eye syndrome. Local administration is expected to avoid the various side effects
`associated with systemic delivery. However, the currently available systems using oils to deliver CsA topically are poorly tolerated and
`provide a low bioavailability. These difficulties may be overcome through formulations aimed at improving CsA water solubility (e.g.
`cyclodextrins), or those designed to facilitate tissue drug penetration using penetration enhancers. The use of colloidal carriers (micelles,
`emulsions, liposomes and nanoparticles) as well as the approach using hydrosoluble prodrugs of CsA have shown promising results. Solid
`devices such as shields and particles of collagen have been investigated to enhance retention time on the eye surface. Some of these topical
`formulations have shown efficacy in the treatment of extraocular diseases but were inefficient at reaching intraocular targets. Microspheres,
`implants and liposomes have been developed to be directly administered subconjunctivally or intravitreally in order to enhance CsA
`concentration in the vitreous. Although progress has been made, there is still room for improvement in CsA ocular application, as none of
`these formulations is ideal.
`q 2003 Published by Elsevier B.V.
`
`Keywords: Cyclosporine A; Ocular delivery; Intraocular; Topical; Delivery system; Prodrug; Review
`
`1. Introduction
`
`is a cyclic undecapeptide
`Cyclosporine A (CsA)
`produced by Tolypocladium inflattum Gams and other
`fungi imperfecti. This drug is now routinely used as an
`oral immunosuppressor for organ transplantation. It acts by
`selective inhibition of interleukin-2 release during the
`activation of T-cells and causes suppression of the cell-
`mediated immune response [1]. The resultant
`immuno-
`suppression is non-toxic and reversible when treatment is
`stopped. Therefore, most of the diseases that
`involve
`cytokines or immune – related disorders are potential targets
`of CsA.
`Over the past years, CsA has been evaluated for
`numerous potential applications in ophthalmology. It is
`effective in the treatment of severe intraocular inflam-
`mations affecting the posterior segment of the eye, when
`
`* Corresponding author. School of Pharmacy, University of Geneva, 30,
`quai E. Ansermet, CH-1211 Geneva 4, Switzerland. Tel.: þ 41-22-379-61-
`46; fax: þ 41-22-379-65-67.
`
`E-mail address: robert.gurny@pharm.unige.ch (R. Gurny).
`
`0939-6411/$ - see front matter q 2003 Published by Elsevier B.V.
`doi:10.1016/S0939-6411(03)00138-3
`
`administered systemically by i.v. injection [2] or orally [3].
`Systemic CsA has also shown efficacy in peripheral
`ulcerative keratitis associated with Wegener’s granuloma-
`tosis [4], in severe Grave’s ophthalmopathy [5] and it was
`also effective in preventing recurrence of graft rejection
`after keratoplasty, and this for a long period of time [6]. In
`fact, intraocular fluids (aqueous or vitreous humor) and
`extraocular organs or annexes (cornea, conjunctiva and
`lachrymal glands) can be reached through the systemic
`pathway after oral or i.v. administration. CsA concen-
`trations of 25 – 75 mg/ml were measured in human tears after
`an oral daily administration of 5 mg/kg [7] but deleterious
`side effects such as nephrotoxicity and hypertension may
`occur [8 – 10]. Although it has met numerous difficulties,
`topical ocular delivery should offer a good alternative.
`Despite a poor intraocular penetration, topical CsA has been
`successfully used in a variety of immune-mediated ocular
`surface phenomena like vernal conjunctivitis [11], dry eye
`syndrome [12] and the prevention of corneal allograft
`rejection [13]. An ideal topical ocular formulation must
`fulfill several requirements: the formulation must be well
`
`1
`
`ALL 2014
`MYLAN PHARMACEUTICALS V. ALLERGAN
`IPR2016-01128
`
`

`

`ARTICLE IN PRESS
`
`2
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`tolerated and easy to administer, increase CsA residence
`time in the eye and avoid systemic absorption (the toxic
`concentration in blood is above 300 ng/ml [14]). In addition,
`the formulation should have a long shelf life and be
`manufactured easily. The main difficulty is that CsA cannot
`be prepared in formulations based on the commonly used
`aqueous ophthalmic vehicles because of both its hydro-
`
`phobicity (log P ¼ 3.0 [15]) and its extremely low aqueous
`
`solubility (6.6 mg/ml [16]). Therefore, in most studies, CsA
`was dissolved and administered in vegetable oils [17 – 19].
`However,
`these media are poorly tolerated, result
`in
`relatively low ocular availability, and have short shelf
`lives. The concentration that has been mostly investigated
`for an eyedrop solution is 1% w/v [20,21] but concentrations
`ranging from 2% w/v [22] to 0.05% w/v [23] have also been
`explored. In all cases, the concentration of the formulation
`remains secondary as long as therapeutic levels in the ocular
`tissues are achieved by the dosage form, immune response
`and inflammation being suppressed at a concentration of
`50 – 300 ng/g of
`tissue [24]. When high intraocular
`concentrations are needed, the drug is also injected directly
`into the eye or periocularly (by the subconjunctival route).
`Numerous formulations were developed to avoid repeated
`injections and achieve controlled release of CsA or to
`enhance efficacy of topical administration. This review
`summarizes the main pharmaceutical systems and devices
`that have been described for topical and intraocular delivery
`of CsA to the eye. It will first present and discuss the topical
`systems developed during the last 15 years. In the second
`part,
`intraocular and subconjunctival devices will be
`reviewed.
`
`2. Topical administration
`
`Most ocular medications may be administered topically
`in order to treat surface as well as intraocular disorders. This
`route is often preferred for the management of various
`pathological diseases that affect the anterior chamber of the
`eye,
`for
`two main reasons:
`it
`is more conveniently
`administered and provides a higher ratio of ocular to
`systemic drug levels. To be administered topically and to
`achieve the necessary patient compliance, CsA must present
`a good local tolerance. Topical CsA in olive oil solution
`induces a burning sensation and an irritative effect on the
`conjunctiva. These side effects have been attributed to the
`vehicles used [25]. Patients did not complain of such
`disorders after application of a 2% w/w CsA ointment, and
`ocular examination revealed no significant lesions [26]. A
`recent study [27] showed that formulations of CsA in peanut
`oil were non-toxic to rabbit eyes. However, an unusual
`corneal deposit [28] was reported in a patient after 5 days of
`topical use of a 1% w/v CsA olive oil eyedrop. This deposit
`was probably due to the precipitation of CsA on the corneal
`surface. No long-term study of surface toxicity is yet
`available.
`
`New developments in the topical delivery of CsA can be
`divided in two general areas of research: new delivery
`systems (solutions, ointments, colloidal carriers and drug-
`impregnated contact lenses) and chemical modifications of
`the drug (prodrugs).
`
`2.1. Solutions and ointments
`
`2.1.1. Oil solutions and ointments
`Several vegetable oils such as arachis [29], castor [30],
`olive [7] and peanut oils [31] have been used to solubilize
`CsA. Petrolatum oils (Cremophor [32]) and ointments
`([23,33] have also been investigated as CsA vehicles for
`topical eye administration. Some authors [24,34] have
`reported that such formulations could achieve, after topical
`administration, therapeutic levels in ocular tissues: Kaswan
`[24] reported concentrations of 4 mg/g in the cornea and 60
`ng/g in the iris 2 h after application. On the other hand, a
`majority of authors [7,17,30,35] have reported none or
`negligible intraocular penetration. Furthermore, oils are
`known to be poorly tolerated by the eye and are therefore
`rapidly evacuated from the ocular surface. Due to its
`lipophilicity [15], CsA has a greater affinity for the vehicle
`than for the cornea, providing a low local availability. Also,
`these vegetable oils may present problems of stability such
`as rancidity [36]. Despite these drawbacks, olive oil is still
`tested in the prevention of corneal graft rejection [13] and is
`still the most frequent reference vehicle cited. A marketed
`ointment formulation for veterinary use (Optimmunew,
`0.2% Cyclosporine USP Ophthalmic Ointment, Schering
`Plough, Welwyn, Herts, UK) is available for the treatment
`of keratoconjunctivitis sicca and ocular surface inflamma-
`tory diseases in dogs [37]. This formulation has not reached
`the human field mainly because of its poor acceptability by
`patients. Tolerance in the veterinary area is evaluated by
`tests (Draize, slit lamp) that do not take into account blurred
`vision and patient discomfort; very important criteria in the
`human field.
`
`2.1.2. Aqueous solutions
`Attempts have been made to improve the solubility of
`CsA in water by complexation of CsA with cyclodextrins or
`penetrations enhancers.
`
`2.1.2.1. Cyclodextrins. Cyclodextrins are complex sugars of
`cyclo-malto-hexose type, exhibiting a lipophilic center
`hidden by an external hydrophilic layer [38]. These
`physicochemical characteristics enable cyclodextrins to
`combine with lipophilic molecules and increase their
`water solubility. CsA combined to a-cyclodextrin was
`solubilized up to 750 mg/ml
`in water [39], which is
`approximately 100-fold higher than for CsA alone. Four
`different formulations were tested and the optimal concen-
`tration for maximum corneal permeability and lowest
`toxicity was found to be 0.025% w/v CsA in 40 mg/ml
`a-cyclodextrin solution. After the application of one drop
`
`2
`
`

`

`ARTICLE IN PRESS
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`3
`
`every 2 h four times a day on the rabbit eye, this solution
`achieved concentrations of 4.1 ^ 0.4 mg/g in the cornea,
`which was five to 10 times higher than those obtained with a
`10% w/w CsA ointment, and above therapeutic levels. This
`study was confirmed by Cheeks [33], who showed on
`excised rabbit corneas that CsA bound to cyclodextrins
`resulted in higher corneal penetration than an application of
`corn oil solutions. This formulation resulted, however, in a
`very small reservoir effect in the cornea, because of the low
`intrinsic quantity of drug in the formulation and the short
`residence time on the eye surface.
`
`2.1.2.2. Penetration enhancers. Penetration enhancers are
`chemicals that can help solubilize CsA and transiently
`modify the corneal epithelium to promote drug penetration
`through the cornea. Azonew (laurocapram) [40] was used as
`a CsA solvent in order to solubilize the drug and improve its
`delivery to the eye. Clinically significant concentrations of
`CsA were measured in the grafted corneas of rabbits but
`little or no drug was found either in the aqueous humor or in
`w
`resulted in
`the blood of the treated animals. CsA in Azone
`suppression of the severity and incidence of graft rejection.
`This penetration enhancer has, since, been shown to induce
`cytotoxicity on corneal epithelium [41].
`The effect of three other penetration enhancers on the
`transcorneal permeation of CsA was evaluated [42]. Flux
`rates of radiolabeled-CsA across human excised corneas
`were measured in the presence and absence of aqueous
`solutions of benzalkonium chloride (0.01%), dimethyl-
`sulfoxide (DMSO) (20%) or Cremophor (10% and 20%)
`(concentrations expressed in w/v). Cremophor and benzal-
`konium significantly increased flux rates of CsA across
`cornea (Fig. 1) while no change was observed with DMSO.
`Benzalkonium presented a very good tolerance at
`the
`concentration used in eye drops as preservative (0.01% w/v)
`[43], but induced ocular irritation at higher concentration
`(1% w/v) [44]. The topical application of Cremophor has
`been associated with changes of corneal surface structure
`[45] while severe anaphylactic hypersensitivity reactions,
`hyperlipidemia, abnormal lipoprotein patterns, aggregation
`of erythrocytes and peripheral neuropathy were observed
`
`Fig. 1. Influence of benzalkonium (0.01%) on the ex vivo flux values of
`CsA (B) across excised fresh (K) and frozen (A) human corneas after
`topical application of CsA aqueous solution [43].
`
`after systemic absorption [46]. The use of penetration
`enhancers represents a potentially interesting approach, but
`with the serious limitation of the low tolerance of these
`molecules that act by modifying the corneal properties
`(mostly a disruption of the epithelial cell layer of the
`cornea).
`
`2.2. Colloidal carriers
`
`Colloidal carriers are small particles of 100 – 400 nm in
`diameter, suspended in an aqueous solution. Calvo [47] has
`shown that colloidal particles were specifically taken up by
`the epithelial cells of the cornea by endocytosis. The cornea
`then acts as a reservoir, releasing the drug to the surrounding
`tissues. These carriers represent a means of delivering
`lipophilic drugs into hydrophilic tissues. They include
`micelles, emulsions, nanoparticles, nanocapsules and
`liposomes.
`
`2.2.1. Micelles
`CsA was solubilized at 0.1% w/v in isotonic and neutral
`aqueous solution by micelles of the non-ionic surfactant,
`polyoxyl 40 stearate, at a concentration of 2% w/v [48].
`After a single administration on the rabbit eye (50 ml), this
`suspension provided a 60-fold higher concentration in the
`cornea than the 0.1% w/v CsA castor oil control solution.
`Although these results are promising, a certain number of
`points remain to be further investigated. Indeed, polyoxy-
`ethylene stearates are widely used in pharmaceutical
`formulations and cosmetics and are generally regarded as
`essentially non-toxic and non-irritant materials [49], but
`ocular tolerance of the surfactant is not known and has not
`been evaluated in this work. In addition, micelles are often
`unstable and their shelf life must be investigated.
`
`2.2.2. Emulsions
`in the
`Oil-in-water emulsions are particularly useful
`delivery of lipophilic drugs. In vivo data from early studies
`confirmed that emulsions could be effective topical
`ophthalmic drug delivery systems [50], with a potential
`for sustained drug release [51]. With the recent improve-
`ments in aseptic processing, and the availability of new
`well-tolerated emulsifiers (polysorbate-80), emulsion tech-
`nology is currently under evaluation for topical CsA
`delivery. Ding and colleagues have developed a castor oil-
`in water microemulsion [52]. This emulsion, stabilized by
`polysorbate 80, solubilizes up to 0.4% w/w CsA and
`remains stable over 9 months at room temperature. It was
`found to cause only mild discomfort and slight hyperemia
`on the rabbit eyes when applied eight times a day during
`7 days. CsA penetrated into rabbit extraocular tissues
`(cornea, lachrymal glands, conjunctiva) at concentrations
`adequate for local immunosuppression while penetration
`into intraocular tissues was much lower and absorption into
`blood was minimal [53]. These encouraging results allowed
`the formulation to undergo clinical trials of phase II and III
`
`3
`
`

`

`ARTICLE IN PRESS
`
`4
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`in dry eye disease [23,54]. The phase II trial performed on
`162 patients demonstrated good tolerance of the emulsion
`and significant improvement of ocular signs and symptoms
`of moderate-to-severe dry eye disease [23]. CsA formu-
`lations of 0.05% and 0.1% w/w were selected for evaluation
`in phase III trials. In this pivotal study, RESTASISw
`demonstrated statistically significant and clinically relevant
`increases in Schirmer wetting versus vehicle at 6 months. It
`has received approval (December 2002) from the United
`States Food and Drug Administration (FDA) for RESTA-
`SISw (cyclosporine ophthalmic emulsion, 0.05%) as the
`first and only therapy for patients with keratoconjunctivitis
`sicca whose lack of tear production is presumed to be due to
`ocular inflammation.
`Since epithelial corneal cells exhibit negative charges on
`their surface, Klang [55] hypothesized that a positively
`charged emulsion would interact with the corneal cells and
`prolong residence time on the surface of cornea. As Ding’s
`emulsion [52] was negatively charged, Abdulrazik and
`coworkers [56] made a positively charged emulsion loaded
`with CsA. Positive charges were introduced in the emulsion
`through the insertion of stearylamine (0.12% w/w). Conse-
`quently, the spreading coefficient of this emulsion on the
`cornea was four times higher than that of the negatively
`charged emulsion. It was therefore deduced that
`the
`positively charged submicron emulsion has better wett-
`ability properties on the cornea. After a single dose of the
`positively charged emulsion on the rabbit eye, CsA yielded
`higher maximum concentrations in the conjunctiva and the
`cornea, compared to the emulsion of Ding [52] (Fig. 2).
`Tolerance evaluation and stability tests have to be
`performed but so far the results are encouraging. The
`emulsion should soon be submitted to a phase I clinical trial.
`
`2.2.3. Liposomes
`Liposomes are membrane-like vesicles consisting of
`one or more concentric phospholipid bilayers alternating
`
`aqueous or lipophilic compartments, making them potential
`carriers for lipophilic drugs. Milani [57] applied that
`technology to the ocular delivery of CsA. He obtained a
`40% trapping efficiency of CsA into such vesicles. The
`formulation was tested topically on corneal rat allografts: a
`liposome suspension at a CsA concentration of 0.21 mg/ml
`was administered five times daily on grafted corneas. After
`60 days, a 77% rate of graft survival was achieved with the
`CsA loaded liposomes group while only 45% survival rate
`was observed in the olive oil CsA solution control group
`(Fig. 3). As serum levels were undetectable, the authors
`concluded that the graft was reached only by the topical
`route. However, the potential of liposomes as a topical CsA
`delivery system remains limited because of their short half-
`life on the corneal surface and relatively poor stability.
`A charge-inducing agent
`like stearylamine could be
`introduced in order to improve intraocular penetration and
`drug availability, as shown by Law [58]. Furthermore, large-
`scale manufacture of sterile liposomes is expensive and
`technically challenging, which make liposomes secondary
`candidates for CsA delivery. Subconjunctival and intra-
`ocular injections of CsA loaded liposomes have also been
`investigated (see Section 3).
`
`2.2.4. Nanoparticles
`Nanoparticles, primarily developed for i.v. adminis-
`tration, have demonstrated promising results over the last 10
`years in ophthalmology. These systems are able to
`encapsulate and protect
`the drug against chemical and
`enzymatic degradation, improve tolerance, increase corneal
`uptake and intraocular half-lives. Three main studies have
`been undertaken to evaluate aqueous suspensions of CsA
`loaded nanoparticles.
`Calvo and coworkers [59] have made nanocapsules
`composed of an oily phase (Mygliolw) surrounded by a
`poly-e-caprolactone (PCL) coat. CsA was loaded in the oil
`
`Fig. 2. Concentrations (ng/g or ml) of CsA in different ocular tissues, 60
`min after topical application of the positively charged (B) and negatively
`charged (A) emulsions containing CsA on the rabbit eye [57].
`
`Fig. 3. Percentage of non-rejected grafted corneas after topical treatment by
`CsA-bounded liposomes (A), CsA in olive oil (W) and empty liposomes (K)
`on a rat model and comparison to controls non-treated grafts (X) and
`syngeneic grafts (B) over 60 days [58].
`
`4
`
`

`

`ARTICLE IN PRESS
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`5
`
`at a concentration of up to 50% (w/w CsA/PCL ratio) to give
`a 10 mg/ml CsA concentration in the final formulation.
`After topical administration, these capsules were taken up
`by corneal epithelial cells [60] and achieved corneal levels
`of CsA that were five times higher than a 10 mg/ml CsA oily
`solution. The delivery system was well tolerated but did not
`provide significant CsA levels at the ocular mucosa for an
`extended period of time. Consequently, this formulation
`failed to prolong corneal graft survival in an experimental
`rat model [61]. Moreover, PCL nanocapsules are degraded
`after autoclaving and g-irradiation [62] and thus must be
`sterilized by aseptic filtration. However, since these
`nanocapsules have a mean size in the range of
`210 – 270 nm the only practical alternative is a totally
`sterile manufacturing process. Although PCL nanoparticles
`did not succeed in prolonging corneal graft survival they
`may be useful in the treatment of extraocular diseases, as the
`cornea would represent a CsA reservoir.
`The ex vivo corneal absorption of CsA loaded
`polyisobutylcyanoacrylate (PACA) nanoparticles and
`nanoparticles in poly(acrylic) acid (carbopol) gel was
`evaluated in bovine corneas [63]. The authors found that
`CsA concentrations in corneas were significantly higher
`with nanoparticles in gel than nanoparticles alone and CsA
`olive oil solution. However, one limitation of the ex vivo
`model is that it does not take into account tear wash and
`lachrymal drainage. Further characterization of nanoparti-
`cles (encapsulation efficiency, size and zeta potential,
`release rates studies) would help an understanding of the
`underlying physiological processes involved in transcorneal
`absorption. PACA nanoparticles are known to penetrate into
`the outer layers of the corneal epithelium causing a
`disruption of the cell membranes [64]. In vivo tolerance
`of these CsA loaded nanoparticles should be investigated.
`Chitosan is a biopolymer obtained by deacetylation of
`chitin that is extracted from crab shells. This polymer is
`positively charged and biodegradable. These properties
`make it a good candidate for ocular delivery. Recently, an
`innovative colloidal drug carrier, made by ionic gelation of
`chitosan, has been described [65]. These nanoparticles
`encapsulated CsA at levels up to 9% of their weight. After
`four applications of 10 ml, the formulation achieved high
`concentrations in vivo (rabbit model) in external ocular
`tissues (cornea and conjunctiva) from 2 to 48 h after
`application (Fig. 4), while other tissues (aqueous humor, iris
`ciliary body, and blood) presented negligible CsA levels.
`These relatively large CsA concentrations in the periocular
`tissues are explained by the corneal and conjunctival surface
`retention due to the positive charges of chitosan. It should be
`noted that no Draize or tolerance tests have been performed
`to evaluate this formulation. However, Felt [66] demon-
`strated excellent tolerance of a topically applied chitosan
`gel. Since steam and dry heat sterilization affect physical
`properties of chitosan [67], and since sterile filtration is not
`possible for such nanoparticles (mean size 283 ^ 24 nm),
`g-sterilization should be investigated for these carriers.
`
`Fig. 4. CsA concentrations (ng/g) in the cornea and conjunctiva after topical
`application of CsA loaded chitosan nanoparticles (B) and control
`formulations over 48 h in a rabbit model (* statistically significant
`differences) [66].
`
`Since, chitosan is a polymer of natural origin, batch to batch
`heterogeneity, with respect
`to molecular weight may
`complicate manufacturing.
`The nanoparticle approach is not yet completely
`satisfactory as the precorneal clearance is still too rapid.
`Of the three CsA colloidal carriers described, the most
`promising, so far, is the chitosan carrier mainly because of
`the therapeutic levels achieved in periocular tissues and its
`good tolerance.
`
`2.3. Solid formulations
`
`Since most solutions are eliminated from the ocular
`surface within a few minutes by normal tear turnover and
`lachrymal drainage, solid systems have been developed in
`order to enhance contact
`time of the drug with the
`extraocular tissues. These include collagen-based systems.
`
`2.3.1. Collagen shields
`This approach consists of the application on the cornea of
`a collagen shield loaded with CsA. Reidy and coworkers
`
`5
`
`

`

`ARTICLE IN PRESS
`
`6
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`[68] tested such a shield loaded with 4 mg CsA on rabbit
`cornea to enhance drug penetration into the anterior
`chamber by increasing contact time on the cornea. These
`shields are directly applied on the rabbit cornea, and the
`eyelids are kept closed by a piece of tape to avoid rejection
`of the system. The device achieved therapeutic concen-
`trations in the cornea and aqueous humor from 2 to 8 h after
`application with a maximum peak at 4 h; no CsA was
`detected in the blood. Both the corneal and aqueous humor
`concentrations of CsA achieved with the shield were 10-fold
`higher than those obtained with topical CsA-olive oil drops
`and over a time period which was twice longer than the
`control. Collagen shields are useful as drug delivery systems
`and also as bandages after corneal surgery. However,
`several drawbacks of the system must be kept in mind. The
`shield may blur the vision. It is also applied dry on the eye, a
`maneuver that may be poorly tolerated especially in some
`pathologies such as dry eye. Also, the collagen matrix is
`disaggregated by tear fluid and after a few hours fragments
`separate from the main part and may be expulsed. There-
`fore, the collagen shield requires a delicate and experienced
`handling in order not to be adversely applied to the ocular
`surface. Consequently, this device may be difficult for self –
`administration by patients.
`
`2.3.2. Liposomes loaded with CsA incorporated in collagen
`shields
`Topical CsA liposomes are able to enhance corneal graft
`survival [57] but have a too short corneal retention time.
`Combining the positive features of collagen shields with the
`advantages of liposomes may provide a synergistic action
`on the eye. Pleyer [69] showed that such a device was able
`to significantly improve CsA aqueous humor concentration
`at 1 and 3 h when compared to CsA loaded liposomes. But,
`this device did not achieve the aim of prolonging corneal
`and aqueous humor levels over 6 h. In addition, this system
`is complex to manufacture and retains the already
`mentioned drawbacks of the collagen shields.
`
`2.3.3. Collagen particles
`To overcome the disadvantages of shields but maintain
`the benefits of collagen devices, small collagen particles
`loaded with CsA were manufactured and suspended in a
`methylcellulose hydrogel. The particle size is relatively
`
`large: 2 mm2 ^ 0.1 mm2 £ 0.1 mm. A volume of the
`
`suspension containing 0.5 mg CsA was administered on the
`cornea of rabbits. The particles were more effective than
`corn oil
`in delivering CsA to the cornea and anterior
`chamber. Higher concentrations of CsA were found in both
`the cornea and aqueous humor of eyes treated with collagen
`particles. Kinetics of drug penetration were different for
`drug in the collagen vehicle compared to the corn oil. The
`particles produced a CsA peak concentration at 4 h and
`maintained a high level until 8 h (Fig. 5) while corn oil gave
`a peak at 1 h that almost disappeared at 8 h [70]. This
`formulation improved significantly corneal allograft
`
`Fig. 5. Concentrations curves of CsA over 24 h in the cornea and aqueous
`humor after topical application on the rabbit eye of different CsA
`formulations: collagen particles (B), collagen shield (O) and corn oil
`solution (B) [72].
`
`survival compared to corn oil [71] but showed no significant
`improvement compared to the collagen shield. The hydrogel
`exerts a viscosifying effect and enhances retention time on
`the cornea. The main advantage of collagen particles is the
`ease with which the formulation can be applied to the ocular
`surface and its possibility for self-administration. Whether
`the sustained release effect
`is due to the collagen
`formulation or to the methylcellulose hydrogel remains to
`be determined. Kinetic studies comparing CsA collagen
`particles alone and CsA suspended in the methylcellulose
`hydrogel would be very welcome. Tolerance evaluation is
`not mentioned and should be evaluated since the particles
`are quite large.
`
`2.4. Chemical modifications of the molecule: prodrug
`approach
`
`Prodrugs are defined as pharmacologically inactive
`derivatives of drug that are chemically or enzymatically
`converted to the active drug. Such molecules are used to
`modify the physico-chemical properties of the drug or its in
`vivo behavior. To improve its water solubility, a water-
`solubilizing moiety was grafted on a free hydroxyl function
`of CsA [72]. The resulting prodrug could be solubilized in
`isotonic and neutral solutions at a concentration equivalent to
`
`6
`
`

`

`ARTICLE IN PRESS
`
`F. Lallemand et al. / European Journal of Pharmaceutics and Biopharmaceutics xx (2003) xxx–xxx
`
`7
`
`1% CsA w/v that is approximately 10,000 times the solubility
`of CsA alone in water. After topical administration of 25 ml
`of this solution in the rabbit eye, the CsA prodrug is cleaved
`in vivo by tear enzymes and releases free CsA. This resulted
`in CsA concentrations in tears above experimental thera-
`peutic levels for up to 20 min. Objective and subjective
`animal (rabbit) tolerance tests showed an improved tolerance
`over that of classical CsA-in-oil solution. This prodrug is a
`promising candidate in the topical treatment of dry eye
`disease and corneal graft rejection.
`Most of the topical delivery systems discussed here
`did not succeed in achieving aqueous humor therapeutic
`levels but corneal and conjunctival levels were sufficient
`to suppress T cell activation. This route will be useful
`for extraocular diseases. As all devices yielded thera-
`peutic levels, choice between systems will be influenced
`by the degree of tolerance and ease of administration and
`manufacture. Altogether,
`three topical
`systems are
`noteworthy: chitosan nanoparticles, positively charged
`emulsions and CsA prodrugs as they are innovative, well
`tolerated and result
`in therapeutic concentrations
`in
`extraocular tissues in animal models.
`Recently, new non-aqueous solvents were proposed
`for the topical delivery of lipophilic drugs. Perfluorocar-
`bons or fluorinated silicone liquids are chemically and
`biologically inert compounds, and present
`low surface
`tension, excellent spreading characteristics and close-to-
`water refractive indices [73]. These solvents could be an
`alternative to complex topical drug delivery systems.
`The insert approach has not yet been proposed to deliver
`CsA. Inserts are composed of a polymeric support contain-
`ing a drug. They are usually placed in the lower fornix and
`may increase contact time between the preparation and
`the conjunctival tissue to ensure a sustained release. As
`lipophilic drugs can be incorporated in inserts [74], CsA
`could be a good candidate for this delivery system.
`
`3. Other administration sites
`
`The other main routes of administration for ocular
`therapeutics include the subconjunctival, intraocular and
`systemic pathways. They are employed when drugs are not
`absorbed by the topical route or when the vitreous humor is
`the target. We can also briefly mention retrobulbar injection
`of CsA [75] that was reported to enhance effect on transplant
`survival, and direct injections of CsA into the anterior
`chamber [76].
`
`3.1. Subconjunctival administration
`
`The subconjunctival route is an alternative to the topical
`and intraocular delivery of CsA. Following subconjunctival
`injections, the administered drug passes through the sclera
`and into the eye by simple diffusion. Before the injection or
`implantation, the eye is anaesthetized by a topical local
`
`anesthetic. Microspheres and implants have been developed
`and tested after subconjunctival administration.
`
`3.1.1. Microspheres
`In order to maintain high levels of CsA in the cornea and
`aqueous humor, Harper [77] employed microspheres made
`of 50:50 D,L-lactide/glycolide copolymer (PLGA) and
`loaded with CsA. Experimental therapeutic concentrations
`were reached in the cornea and aqueous humor 6 h after
`subconjunctival injection, in a rabbit model, of 0.13 ml of
`the microsphere solution containing a total dose of 2 mg of
`CsA (approx. 15 mg/ml in the final formulation). These
`concentrations were maintained for 2 weeks in the cornea
`after injection. Although these results were encouraging,
`efficacy tests we

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