throbber
Progress on New Therapeuties for Fungal
`Nail Infections
`Stephen J. Bakera, Xiaoying Huib and Howard I. Maibachb
`
`aAnacor Pharmaceuticals, 1060 East Meadow Circle, Palo Alto, CA 94303, USA
`bDepartment of Dermatology, Surge Building, Room 110, 90 Medical Center Way, University
`of California at San Francisco, San Francisco, CA, 94143, USA
`
`Contents
`1. Introduction
`2. Drug Therapy
`2.1. Systemic treatments
`2.2. Topical treatments
`2.3. Combination and booster treatments
`3. Drug Penetration Through the Nail
`3.1. Composition of the nail plate
`3.2. Nail penetration models
`4. Factors Affecting Drug Penetration
`4.1. Physicochemical properties of the drug
`4.2. External factors
`5. Infection Models
`5.1. In vitro infection models
`5.2. In vivo infection models
`6. Preclinical/Clinical Pipeline
`7. Conclusion
`References
`
`1. INTRODUCTION
`
`333
`334
`335
`336
`337
`337
`337
`338
`340
`340
`340
`342
`342
`342
`343
`344
`344
`
`Onychomycosis is a fungal infection of the toe and finger nails, with the majority of
`cases involving infection of the toe nails [1]. The disease is mostly caused by a class
`of fungi known as the dermatophytes, which are also responsible for skin fungal
`infections. Dermatophytes flourish on dead keratinized tissue and normally infect
`the stratum corneum layer of skin, scalp hair and nails [2]. Non-dermatophyte
`species including yeasts and molds can also be involved. The dermatophytes ac-
`count for around 90% of all cases of onychomycosis [1,3] and include Trichophyton,
`Microsporum and Epidermophyton species. However, Trichophyton rubrum and
`Trichophyton mentagrophytes are by far the major causative agents accounting for
`60–70% of the cases [1,3,4]. The fungi can infect the nail plate, nail bed and sur-
`rounding skin folds (proximal fold at the cuticle and lateral folds on either side of
`the nail plate). Onychomycosis damages the nail plate causing thickening and
`discoloration. In more advanced cases, the nail plate lifts away from the nail
`bed, termed onycholysis, which causes discomfort and sometimes can be painful.
`
`ANNUAL REPORTS IN MEDICINAL CHEMISTRY, VOLUME 40
`ISSN: 0065-7743 DOI 10.1016/S0065-7743(05)40021-4
`
`r 2005 Elsevier Inc.
`All rights reserved
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 1 of 13
`
`

`
`334
`
`S.J. Baker et al.
`
`Clinical presentations of onychomycosis have been divided into four categories:
`distal subungual (infection occurs at or near the tip of the nail plate and involves the
`underlying nail bed), proximal subungual (infection is at or near the cuticle and
`involves the underlying nail bed), superficial (infection is in the nail plate only with
`no nail bed involvement) and total dystrophic onychomycosis (whole nail involve-
`ment and considered a combination of the other types) [5]. Between 6.5 to 13.8% of
`the population in North America is reported to be infected with this disease and the
`prevalence increases with age [1,4,6,7]. One study reported 48% of 70 year olds are
`infected with onychomycosis [6].
`
`2. DRUG THERAPY
`
`Onychomycosis is difficult to permanently cure. Treatment failures and relapses are
`common, which exacerbate the problem [8–10].
`In order for an antifungal drug to be effective, it must presumably disseminate
`throughout the nail plate, nail bed and other locations occupied by the fungi, and
`reach concentrations that will eliminate the pathogen. This can be especially dif-
`ficult when the nail plate has lifted from the bed (onycholysis). Unlike damaged skin
`that can repair itself, the nail plate cannot, therefore results of therapeutic treatment
`are not evident until new nail growth occurs and is clear of infection. Toe nails
`typically take about 1 year to fully grow out.
`Because of the length of time required to observe new nail growth, clinical trials
`typically take around 9–12 months (either 3 months systemic treatment with 6–9
`months follow up or 6–9 months topical treatment with 3–6 months follow up).
`During this time, the infected nails can be monitored for growth of new clear nail
`and for presence of viable dermatophytes. Efficacy is usually recorded in one of
`three ways: mycological cure, clinical cure or complete cure. Standard definitions of
`these cures are not completely uniform; each report usually provides the criteria
`that were used in the study. A mycological cure is defined by the extent of erad-
`ication of the fungi. It is assessed by removing a section of nail and screening for the
`presence of dermatophytes by microscopy and by culturing the nail for growth of
`dermatophytes in vitro. A clinical cure is defined by the extent of new nail growth at
`the proximal fold which is visibly clear of infection. A complete cure is defined when
`a patient has a mycological cure and clinical cure. Obviously, a complete cure is
`most desirable but hindering this is the fact that in many cases more than one digit
`is infected and not every digit may be cleared of infection.
`Onychomycosis is treated both systemically and topically. Current systemic
`treatments include terbinafine (1), itraconazole (2) and griseofulvin (3). Current
`topical treatments include ciclopirox (4), amorolfine (5) and tioconazole (6).
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 2 of 13
`
`

`
`Fungal Nail Infections
`
`335
`
`OMe
`
`O
`
`Me
`
`O
`
`MeO
`
`O
`
`Cl
`
`OMe
`
`Cl
`
`S
`
`3
`
`Cl
`
`6
`
`N
`
`N
`
`Cl
`
`Cl
`
`Cl
`
`O
`
`N
`
`N
`
`O
`
`O
`
`N
`
`Me
`
`Me
`
`Et
`
`O
`
`N N
`
`N
`
`Me
`
`N
`
`2
`
`N
`
`N
`
`Me
`
`O
`
`Me
`
`NM
`
`e
`
`1
`
`Me
`
`O
`
`N O
`
`H
`
`4
`
`5
`
`2.1. Systemic treatments
`
`is the most effective method of curing ony-
`Currently, systemic treatment
`chomycosis. Even so, between 20–25% of patients fail to respond [11]. Terbina-
`fine (1) and itraconazole (2) are the two systemic treatments of choice with ter-
`binafine showing greater efficacy than itraconazole and lower rates of recurrence
`[11–15].
`Terbinafine (1), a representative of the allylamine class of antifungal agents,
`inhibits squalene epoxidase [16,17] and thereby prevents the biosynthesis of ergos-
`terol, a key ingredient in the fungal cell wall. Terbinafine is active against de-
`rmatophytes, M. furfur, Aspergillus species and some Candida species including
`C. parapsilosis; however, it is fungistatic against C. albicans [2]. A single oral dose of
`250 mg terbinafine given to humans produces peak plasma concentrations of 1 mg/
`mL within two hours [14]. It is 499% protein bound and has a half-life of about 36
`hours. It is administered at a dose of 250 mg once daily for 6 weeks for finger nails
`or 12 weeks for toe nails [14]. One study showed that terbinafine localizes in the
`stratum corneum via sebum [18]. Terbinafine has a cLogP of 6.5 and a molecular
`weigh of 292 Da.
`Itraconazole (2), which is from the azole class of antifungal agents, inhibits la-
`nosterol 14 a-demethylase and thus stops the biosynthesis of ergosterol. It has
`broad spectrum activity against species including dermatophytes, Candida species,
`Aspergillus species and M. furfur [2]. Blood levels of itraconazole after a single
`200 mg dose given to humans reached a peak level of 0.2–0.3 mg/mL after 4–5 hours
`[15]. It is 99.8% protein bound and has a half-life of 21 hours. It is administered
`either 200 mg once daily for 12 weeks or 200 mg twice daily for 7 days followed
`by 3 weeks with no treatment and repeated for three months. Like terbinafine,
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 3 of 13
`
`

`
`336
`
`S.J. Baker et al.
`
`itraconazole also localizes in the stratum corneum via sebum but at much lower
`levels [18,19]. Itraconazole has a cLogP of 3.3 and molecular weight of 706 Da.
`Griseofulvin (3), isolated from Penicillium griseofulvin in 1939 [20], has a limited
`spectrum of activity. It is fungistatic against dermatophytes only and works by
`binding to microtubular proteins thus inhibiting cell mitosis. It has a cLogP of 2.2
`and a molecular weight of 353 Da.
`The commonly used antifungal agent, fluconazole, has also been prescribed, off-
`label, for the treatment of onychomycosis.
`
`2.2. Topical treatments
`
`Treatment of onychomycosis by topical methods has been met with limited success
`and reasons for this will be explored in more detail in Section 3. As with treating
`skin fungal
`infections such as tinea pedis (athletes foot), topical application
`for onychomycosis would seem the obvious choice. However, unlike the stratum
`corneum, the nail plate is a more difficult barrier to penetrate, requiring the drug
`to have much different physicochemical properties than are required for skin
`penetration. The two main topical treatments used today are ciclopirox and am-
`orolfine, both of which are formulated in lacquers that are painted onto the infected
`nails. The lacquer dries to leave a water-insoluble film on top of the infected nail,
`which then acts like a drug depot releasing the drug into the nail plate [21,22].
`Tioconazole has also been used but has been largely replaced by ciclopirox and
`amorolfine.
`Ciclopirox (4) is a hydroxypyridone antifungal agent and is believed to work by
`inhibiting metal dependant enzymes that degrade intracellular toxic peroxides. It
`does this by chelating the polyvalent cations (Fe3+ or Al3+) required by these
`enzymes [23–25]. Ciclopirox has antifungal, antibacterial and anti-inflammatory
`activities [25]. It is administered to the infected nails daily and due to the slow
`growth of nails, this treatment continues for at least 6 months. Ciclopirox has a
`cLogP of 2.5 and a molecular weight of 207.
`Amorolfine (5) is a morpholine antifungal agent and works by inhibiting ergos-
`terol biosynthesis. Amorolfine is administered once or twice weekly to the infected
`nails for 6 to 12 months. Amorolfine has a cLogP of 5.8 and molecular weight of
`317.
`The relative lack of clinical efficacy seen by topical antifungal treatments has led
`to a substantial research effort to understand the reasons for this failure. The most
`common belief
`is that
`treatment
`failure following topical
`therapy for on-
`ychomycosis results from the inability of the drug to penetrate and disseminate
`throughout the nail. This topic will be explored in more depth in Sections 3 and 4.
`Other factors that have been implicated include lack of microbiological activity in
`the presence of keratin [26,27], lack of microbiological activity against the dormant
`dermatophytes in the nail keratin [28] and poor penetration of drug into the de-
`rmatophytoma, a thick mass of fungi and nail debris, that builds up between the
`nail plate and nail bed [29].
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 4 of 13
`
`

`
`Fungal Nail Infections
`
`337
`
`2.3. Combination and booster treatments
`
`Since there are no current antifungal treatments available that will provide a com-
`plete cure, practitioners are attempting combination therapy and/or booster thera-
`py in an attempt to improve efficacy rates [30–32]. Combination therapy includes
`the use of oral plus oral therapy e.g. oral teribinafine plus oral itraconazole either in
`parallel or sequentially; oral plus topical therapy, e.g. oral terbinafine plus topical
`ciclopirox lacquer; or other dual, triple or quadruple combinations. However, these
`studies show only marginal improvement at best and further studies are warranted.
`Booster therapy involves giving a second course of systemic treatment, terbinafine
`or itraconazole, 6–9 months after systemic treatment began [30].
`
`3. DRUG PENETRATION THROUGH THE NAIL
`
`3.1. Composition of the nail plate
`
`The human nail anatomy consists of nail plate, nail bed and nail matrix. The nail
`plate consists of three layers: the dorsal and intermediate layers derived from the
`matrix, and the ventral layer derived from the nail bed [33,34]. The upper (dorsal)
`layer is a few cell layers thick and consists of hard keratin. It constitutes the main
`barrier to drug diffusion into and through the nail plate. The intermediate layer
`constitutes three quarters of the whole nail thickness, and consists of soft keratin.
`Below the intermediate layer is the ventral layer of soft keratin, a few cells thick,
`that connects to the underlying nail bed, in which many pathological changes can
`occur. Thus, in the treatment of nail diseases, achieving an effective drug concen-
`tration in the ventral nail plate is of great importance. The nail bed consists of non-
`cornified soft tissue under the nail plate, and is highly vascularized. Beneath the nail
`bed at the proximal fold is the nail matrix, which is a heavily vascularized thick
`layer of highly proliferative epithelial tissue that forms the nail plate.
`The human nail is approximately 100 times thicker than the stratum corneum of
`the skin, and both are rich in keratin. However, they exhibit some physical and
`chemical differences [35,36]. The nail possesses high sulphur content (cystine) in its
`hard keratin domain, whereas the stratum corneum does not. The total lipid con-
`tent of the nail ranges from 0.1% to 1%, as opposed to approximately 10% for the
`stratum corneum.
`Under average conditions, the nail contains 7% to 12% water, in comparison to
`25% in the stratum corneum. At 100% relative humidity, the maximum water
`content in the nail is approximately 25%, in sharp contrast to that in the stratum
`corneum, which can increase to 200–300%.
`The nail’s unique properties, particularly its thickness and relatively compact
`construction, make it a formidable barrier to the entry of topically applied agents
`[37]. In one study, the concentration of an applied drug across the nail dropped
`about 1000-fold from the outer surface to the inner surface [38]. As a result, the
`drug concentration presumably had not reached a therapeutically effective level in
`the inner ventral layer.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 5 of 13
`
`

`
`338
`
`S.J. Baker et al.
`
`The existing clinical evidence suggests that a key to successful treatment of ony-
`chomycosis by a topical antifungal product lies in effectively overcoming the nail
`barrier.
`
`3.2. Nail penetration models
`
`To achieve an effective drug concentration into and through the human nail plate,
`development of an appropriate in vitro method to explore the physicochemical
`characteristics and permeability of the nail is of importance.
`Walters and his colleagues pioneered the study of the permeability characteristics
`of the human nail plate in the early 1980’s. They designed an in vitro method
`utilizing a stainless steel diffusion cell that permitted the exposure of nail plate to a
`bathing medium that was stirred by small motors mounted above the cell. Their
`most important research finding is that, as a permeable membrane, the hydrated
`human nail plate behaves more like a hydrophilic gel membrane in its barrier
`properties than as a lipophilic membrane such as stratum corneum [37]. This finding
`also explains the behavior of some solvents that promote diffusion through the skin
`horny layer but have little promise as accelerants of nail plate permeability [39].
`Mertin and Lippold [35,36,40] modified Franz diffusion cells to measure nail per-
`meability characteristics and drug uptake into nails. For onychomycosis treatment,
`they indicated, not only the flux of an antifungal drug through the nail plate is of
`importance, but also the antifungal potency, which is expressed as minimum inhib-
`itory concentration (MIC). An efficacy coefficient, E, was therefore introduced, which
`should be maximized for high therapeutic effectiveness. Thus, for maximum efficacy,
`a high flux of drug through the nail and a low MIC are desired characteristics.
`E ¼ Flux=MIC
`Kobayashi and his coworkers [34] investigated the permeation characteristics of
`drugs with different lipophilicity through three layers of the human nail plate (the
`dorsal, intermediate, and ventral nail layers), using a modified side-by-side diffusion
`cell. The data suggested that the upper (dorsal) layer functions as the main nail barrier
`to drug permeation, exhibiting low drug diffusivity.
`However, most published in vitro nail study methods required the human nail
`sample to be in contact with an aqueous solution on either or both sides during
`incubation. Consequently, the human nail plate is artificially hydrated beyond
`normal levels. Methods of nail sample preparation have also used scalpel or sand
`paper to remove nail samples, which is not only time consuming, but also may not
`be accurate [34,41]. Recently Hui et al. [42–44] developed a novel experimental
`system that simulates the in vivo conditions of therapeutic, non-occluded applica-
`tion of drug to a human nail. In this device, the human nail (top center) surface was
`open to air, while the inner (ventral) surface made contact with a small saline-
`wetted cotton ball, which acted as both a nail supporting bed and a moisture
`supplier. The average of hydration of the wetted cotton balls, 11879.4 AU, re-
`sembles the average hydration of a human nail bed, 99.978.9 AU, as measured in
`fresh human cadavers. After completion of the dosing and incubation phase, the
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 6 of 13
`
`

`
`Fungal Nail Infections
`
`339
`
`nail plate was transferred to a micrometer-controlled nail sampling instrument that
`enables accurate and reproducible sampling of the inside of the nail with high mass
`balance efficiency (Fig. 1).
`Data shows that the average depth of nail sampling from the inner center surface
`was well controlled at 0.2670.05 mm (corresponding with the ventral/intermediate
`layer), which was close to the expected depth of 0.24 mm. With this in vitro nail study
`system, Hui et al. examined antifungal drugs delivered into the dorsal/intermediate
`and ventral/intermediate nail layers, and into the support bed (cotton ball) to de-
`termine the flux and the efficacy coefficient (E) of drugs in nail (Table 1). This study
`shows both ciclopirox and econazole penetrate to the deep layer of the nail plate in
`
`Dorsal layer
`
` Topical dose
`
`Dorsal/intermediate center
`
`Intermediate layer
`
`Remainder nail
`
`Ventral layer
`
`Ventral/intermediate center
`
`Cutting tip
`for nail sampling
`
`Fig. 1. Nail and nail drilling tip.
`
`Table 1. Two antifungal drugs, econazole and ciclopirox, concentration and rela-
`tive antifungal efficacy
`
`Parameter
`
`Nail Lacquera
`
`Control
`Formulation
`
`p Value (t Test)
`
`Econazole in the deeper
`layer (mg/cm3)b
`Efficacy coefficient E
`(MIC ¼ 1 mg/mL)
`Ciclopirox in the deeper
`layer (mg/cm3)b
`Efficacy coefficient E
`(MIC ¼ 0.04 mg/mL)
`
`14,830 (341)
`
`2,371 (426)
`
`0.008
`
`14,830
`
`2,371
`
`0.008
`
`407 (106)
`
`10,175
`
`—
`
`—
`
`—
`
`—
`
`Sources: econazole data is from reference [43]; ciclopirox data is from reference [44].
`aThe data represent the mean (SD) of each group (n ¼ 5). The nail lacquer group of econ-
`azole contains 18% 2-n-nonyl-1,3-dioxolane and the control formulation contains no
`2-n-nonyl-1,3-dioxolane.
`bThe deeper layer is the center of the ventral/intermediate layer of the nail plate. The data
`represent the amount drug in the sample after a 14-day dosing period.
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 7 of 13
`
`

`
`340
`
`S.J. Baker et al.
`
`concentrations above the MIC and that econazole penetration is significantly im-
`proved when formulated with a lacquer containing a penetration enhancer.
`
`4. FACTORS AFFECTING DRUG PENETRATION
`
`Using in vitro nail penetration models, studies have been performed to help un-
`derstand the physicochemical properties that allow a molecule to penetrate into and
`through the human nail plate. External factors also have a major influence upon
`nail penetration and they are: nail thickness, the vehicle within which the drug is
`formulated, pH of the vehicle and addition of permeation enhancing agents to the
`vehicle. These factors have been recently reviewed [21,45].
`
`4.1. Physicochemical properties of the drug
`
`A recent study [46] investigated the relationship of molecular weight and lipophili-
`city of benzoic acid derivatives upon nail penetration. Using p-hydroxybenzoic
`esters ranging from methyl to hexyl, they found that permeation through the nail
`was mostly influenced by molecular weight and little, if any, by lipophilicity, which
`is in agreement with earlier studies [35,36]. This finding can be understood after
`consideration nail plate morphology. Because the nail plate is composed of many
`strands of keratin held together through disulfide bonds, the space between the
`strands must have a finite size causing the nail plate to act like a molecular sieve or
`size exclusion medium. Small molecules can weave through these spaces while larger
`molecules are unable to pass [21].
`The molecular weight of most antifungal agents is4300 Da. Accordingly, these
`drugs will have difficulty penetrating the nail plate, a likely reason for the low
`clinical efficacy observed [47].
`
`4.2. External factors
`
`4.2.1. Nail plate effect
`
`Nails infected with onychomycosis are thicker than healthy nails due to the pres-
`ence of the dermatophyte and the damage they have caused. The effect of nail
`thickness on penetration has been investigated and found to have an inverse re-
`lationship; as the nail increases in thickness drug penetration is reduced [46]. In the
`same study, the authors compared penetration of 5-fluorouracil though healthy
`nails versus fungal infected nails. They concluded that there was no significant
`difference between healthy and infect nails. However, they did not investigate
`heavily infected nail plates because the thickness of the uneven plate could not be
`accurately measured and that the uneven nail plate would collapse in water. They
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 8 of 13
`
`

`
`Fungal Nail Infections
`
`341
`
`speculated that penetration through this type of nail plate should increase due to the
`destruction of the plate caused by the fungi.
`
`4.2.2. Excipients
`
`The nail plate acts like a hydrogel and swells in the presence of water resulting in
`increased pore size [34]. This has the overall effect of increasing permeation [21]. A
`study into the penetration of ciclopirox (4) in three formulations concluded that
`when ciclopirox was formulated in an aqueous gel, its penetration was far superior
`than when formulated in a lacquer [44]. The effect of lipophilic vehicles on drug
`penetration through the nail plate has been investigated [40]. The authors concluded
`that providing the formulation does not affect the hydration level of the nail plate
`and the lipophilic vehicle does not penetrate the nail plate, then penetration of the
`drug is independent of the vehicle medium.
`
`4.2.3. Vehicle pH
`
`Antifungal agents have a range of pKa values and so studies have been reported that
`compare the penetration of the ionic and non-ionic forms of the parent. These studies
`investigated the penetration of miconazole [48] (pK a ¼ 6:7), benzoic acid [35]
`(pK a ¼ 4:2), pyridine [35] (pK a ¼ 5:3) and 5-fluorouracil [49] (pK a ¼ 7:9) in vehicles
`over a pH range from 2 to 8.5. In the case of miconazole, it was reported that
`penetration was independent of the pH of the vehicle. However, in all the other cases,
`the ionic forms of the parent did not penetrate as well as the non-ionic forms. A recent
`study [46] investigating the penetration of ionic and non-ionic compounds and the
`relationship with molecular weight also found non-ionic compounds penetrate better.
`These authors speculated that the decrease in penetration of ionic drugs may be due to
`an apparent increase in molecular weight of around 100 Da from ion hydration.
`
`4.2.4. Penetration enhancers
`
`Efficacy rates of onychomycosis agents are widely believed to improve if penetra-
`tion of antifungal agents through the nail plate is increased. Initial research has
`focused upon modifying environmental conditions, using penetration enhancers, to
`allow larger molecules to penetrate more easily through the nail plate. The most
`common method is to add a chemical enhancer to the vehicle. This enhancer can be
`a keratolytic agent [50], such as urea or salicylic acid, which break down the nail
`keratin; mercaptans [50], which break disulfide bonds in the nail keratin; solvent
`carrier, such as DMSO [42]; or 2-n-nonyl-1,3-dioxolane [43], which has been shown
`to fluidize stratum corneum lipids [51] (although its effect on nails is unknown). In
`most cases reported, an increase in penetration of the nail plate was observed.
`Mechanical methods to increase penetration have also been attempted. Lacquer
`formulations are the most common method to enhance penetration and are used in
`commercial preparations of ciclopirox and amorofine. After the lacquer dries, the
`drug impregnated film remaining creates a large drug gradient across the nail plate,
`which may force the drug through the nail plate. The lacquer also increases
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 9 of 13
`
`

`
`342
`
`S.J. Baker et al.
`
`in the nail plate, which will assist dissemination of the drug
`hydration level
`throughout the nail plate [21]. Another report described the use of pressure sensitive
`adhesives to enhance ciclopirox penetration [52].
`
`5. INFECTION MODELS
`
`Since a complete cure is highly improbable without good nail penetration, in vitro nail
`penetration studies are becoming one of the first screening tools used in the selection
`of compounds for treatment of onychomycosis. Once a compound has been identified
`that exhibits good penetration properties, the next step in lead optimization is to
`assess efficacy in a model of onychomycosis. This is a relatively new area of research
`and few models of onychomycosis exist. Several of these are summarized below.
`
`5.1. In vitro infection models
`
`One recent model of infection tested the minimum fungicidal activity (MFC) of
`antifungal agents against T. rubrum in a medium containing human nail powder,
`which the authors termed nail-MFC [53]. The medium by itself would not support
`the growth of T. rubrum but addition of the pulverized keratin allowed the der-
`matophytes to grow. The antifungal agents were incubated with T. rubrum in this
`nail medium for four weeks, after which the viability of T. rubrum was assessed.
`Terbinafine had a nail-MFC of 1 mg/mL, but other antifungal agents did not show
`efficacy at the highest concentrations tested (amorolfine 1 mg/mL, ciclopiroxolamine
`128 mg/mL, clotrimazole 64 mg/mL, fluconazole 128 mg/mL, griseofulvin 64 mg/mL,
`itraconazole 4 mg/mL and naftifine 8 mg/mL).
`Two groups have reported models of dermatophyte infection in human nail
`plates. In one study, nail clipping were applied to the top of an agar plate cultured
`with T. mentagrophytes and after a few days the infection spread onto the nail plate.
`Drugs were assessed by applying the formulation to the top of the nail and com-
`paring the extent of dermatophyte infection that had spread to the nail plate with an
`untreated control [54]. In the second model, T. mentagrophytes was applied to the
`nail plate directly without a supporting medium and invasion occurred without
`addition of nutrients [55].
`
`5.2. In vivo infection models
`
`Two in vivo efficacy models in guinea pigs have been reported. In the first study, the
`authors infected the nails and toes of guinea pigs creating both onychomycosis and
`tinea pedis (fungal infection of the surrounding skin). They then used this model to
`show the efficacy of a topically applied triazole in comparison with amorolfine and
`terbinafine. All three were effective in clearing the tinea pedis, but only the exper-
`imental triazole showed efficacy against onychomycosis [27]. The second model was
`developed as an optimized in vivo model for dermatophytosis. The authors shaved
`and abraded the skin on the back of guinea pigs and infected the site with T.
`mentagrophytes. After the infection was established, the animals were treated with
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 10 of 13
`
`

`
`Fungal Nail Infections
`
`343
`
`oral and topical formulations of terbinafine and observed for improvement of the
`infection [56]. In this study, 1% topical terbinafine treatment had 100% clinical and
`mycological efficacy. Although this is a model for dermatophytosis, this model was
`used to determine the efficacy of a topical lacquer formulation of terbinafine.HCl,
`currently in clinical development for onychomycosis [57].
`
`6. PRECLINICAL/CLINICAL PIPELINE
`
`New treatments in development for onychomycosis are listed in Table 2. Currently,
`most treatments that are in clinical trials represent re-formulations of known an-
`tifungal agents combined with penetration enhancers in an effort to increase pen-
`etration through the nail plate. There are two treatments in clinical trials that
`contain novel antifungal agents. The first, in Phase 2, is a topical treatment that
`generates nitric oxide. The second, in Phase 3, is a topical treatment containing
`abafungin (7), a membrane integrity antagonist that has antibacterial and antifun-
`gal activity. In addition to the treatments listed, azoline and ravuconazole, two oral
`triazole antifungal agents are in clinical trials for fungal infections and include
`onychomycosis as a potential indication.
`
`N
`
`H N
`
`H N
`
`N
`
`S
`
`O
`
`Me
`
`7
`
`Me
`
`Table 2. Onychomycosis treatments in preclinical and clinical development
`
`Status
`
`Discovery
`
`Phase 1
`
`Phase 2
`
`Phase 3
`
`Drug
`
`Further information
`
`Ciclopirox (4)
`Ketoconazole
`Econazole
`
`Formulated in a metered dose transdermal spray
`Formulation contains a topical carrier
`Formulation contains the penetration enhancer
`2-n-nonyl-1,3-dioxolane
`Formulation contains an absorption enhancer
`Clotrimazole
`Topical NO donor Novel topical treatment that generates nitric
`oxide
`Formulation contains a penetration enhancer
`Oral treatment, once daily regime
`A membrane integrity antagonist formulated as
`a topical treatment for bacterial and fungal
`skin infections
`
`Terbinafine
`Itraconazole
`Abafungin (7)
`
`CFAD v. Anacor, IPR2015-01776, CFAD EXHIBIT 1066 - Page 11 of 13
`
`

`
`344
`
`7. CONCLUSION
`
`S.J. Baker et al.
`
`Onychomycosis is a common disease, especially in the older generation, that is
`difficult to treat using current medicines. In efforts to understand why current
`topical treatments work so poorly, research studies have focused on the nail plate
`and factors affecting movement of organic molecules through this barrier. The
`major problem that must be overcome is penetration and dissemination of the drug
`throughout the nail plate. Several models of nail penetration have been developed
`in order to assess this parameter. Data from these models suggest the optimal
`properties for effective penetration are: low molecular weight, low polarity, activity
`in the presence of keratin and optimal vehicle formulation (favorable excipients, pH
`and/or penetration enhancers). Infection models have also been developed that
`provide additional screening tools during lead optimization studies. These studies
`represent a major advancement in the search for new and effective treatments.
`However, as this remains a young field, it is unknown how success in these pre-
`clinical models will translate into clinical efficacy.
`
`REFERENCES
`
`[1] A. K. Gupta, H. C. Jain, C. W. Lynde, P. MacDonald, E. A. Cooper and R. C.
`Summerbell, J. Am. Acad. Dermatol., 2000, 43, 244.
`[2] M. D. Richardson and D. W. Warnock, Fungal Infections, Diagnosis and Management,
`3rd edition, Blackwell Publishing, Malden, MA, 2003.
`[3] R. C. Summerbell, J. Kane and S. Krajden, Mycoses, 1989, 32, 609.
`[4] M. A. Ghannoum, R. A. Hajjeh, R. Scher, M. Konnikov, A. K. Gupta, R. Summerbell,
`S. Sullivan, R. Daniel, P. Krusinski, P. Fleckman, P. Rich, R. Odom, R. Aly, D. Pariser,
`M. Zaiac, G. Rebell, J. Lesher, B. Gerlach, G. F. Ponce-de-leon, A. Ghannoum, J.
`Warner, N. Isham and B. Elewski, J. Am. Acad. Dermatol., 2000, 43, 641.
`[5] J. Faergemann and R. Baran, Br. J. Dermatol., 2003, 149 (Suppl. 65), 1.
`[6] B. Elekski and M. A. Charif, Arch. Dermatol., 1997, 133, 1172.
`[7] G. E. Pie´ rard, Dermatology, 2001, 202, 220.
`[8] J. E. Arrese and G. E. Pie´ rard, Dermatology, 2003, 207, 255.
`[9] R. K. Scher and R. Baran, Br. J. Dermatol., 2003, 149 (Suppl. 65), 5.
`[10] A. K. Gupta and L. E. Lynch, Cutis, 2004, 74 (Suppl. 1), 10.
`[11] B. Sigurgeirsson, S. Billstein, T. Rantanen, T. Ruzicka, E. Di Fonzo, B. J. Vermeer, M.
`J. D. Goodfield and E. G. V. Evans, Br. J. Dermatol., 1999, 141 (Suppl. 56), 5.
`[12] D. T. Roberts, Br. J. Dermatol., 1999, 141 (Suppl. 56), 1.
`[13] C. De Cuyper and P. H. F. B. Hindryckx, Br. J. Dermatol., 1999, 141 (Suppl. 56), 15.
`[14] Lamisil package insert.
`[15] Sporanox package insert.
`[16] N. S. Ryder, G. Seidl, G. Petranyi and A. Stuetz, Recent Adv. Chemother., Proc. Int.
`Congr. Chemother., 14th (1985) (Antimicrobial Sect. 3), 2558-9, Publisher, Univ. Tokyo
`Press, Tokyo, Japan.
`[17] M. A. Ghannoum and L. B. Rice, Clin. Microbiol. Rev., 1999, 12, 501.
`[18] J. Faergemann, H. Zehender, J. Denoue¨ l and L. Millerioux, Acta Dermatol. Venereol.
`(Stockh.), 1993, 73, 305.
`[19] G. Cauwenbergh, H. Degreef, R. Woestenborghs, P. Van Rooy and K. Haeverans,
`J. Am. Acad. Dermatol., 1988, 18, 263.
`[20] E. D. Weinberg, in Principles of Medicial Chemistry, (ed

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