Penetracion Ocular

Embed Size (px)

Citation preview

  • 8/11/2019 Penetracion Ocular

    1/36

    Molecular Design for Enhancement of Ocular Penetration

    YOSHIHISA SHIRASAKISenju Pharmaceutical Co., Ltd., 1-5-4 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan

    Received 22 June 2007; revised 21 August 2007; accepted 23 August 2007

    Published online in Wiley InterScience (www.interscience.wiley.com). DOI 10.1002/jps.21200

    ABSTRACT: Over the past two decades, many oral drugs have been designed inconsideration of physicochemical properties to attain optimal pharmacokinetic proper-

    ties. This strategy significantly reduced attrition in drug development owing to inade-

    quate pharmacokinetics during the last decade. On the other hand, most ophthalmic

    drugs are generated from reformulation of other therapeutic dosage forms. Therefore,the modification of formulations has been used mainly as the approach to improve ocular

    pharmacokinetics. However, to maximize ocular pharmacokinetic properties, a specific

    molecular design for ocular drug is preferable. Passive diffusion of drugs across the

    cornea membranes requires appropriate lipophilicity and aqueous solubility. Improve-

    ment of such physicochemical properties has been achieved by structure optimization or

    prodrug approaches. This review discusses the current knowledge about ophthalmic

    drugs adapted from systemic drugs and molecular design for ocular drugs. I propose the

    approaches for molecular design to obtain the optimal ocular penetration into anterior

    segment based on published studies to date. 2007 Wiley-Liss, Inc. and the American

    Pharmacists Association J Pharm Sci 97:24622496, 2008

    Keywords: tissue partition; drug design; permeability; solubility; structure-property

    relationship (SPR); epithelial delivery/permeability

    INTRODUCTION

    Over the past two decades, oral drugs have been

    designed in consideration of physicochemical

    properties to maximize their pharmacokinetic

    properties. Numerous papers and reviews

    describe the design of molecules to improve the

    pharmacokinetic properties such as oral absorp-tion, bioavailability and the duration of action.19

    At present, in silico screening is widely used forthe selection of drug-like compounds from combi-

    natorial libraries and is based on physicochemical

    parameters such as the rule-of-five,10 which is a

    qualitative absorption/permeability predictor.1113

    Physicochemical property-based drug design

    can reduce attrition due to inappropriate phar-

    macokinetics in the drug development process.

    Inappropriate pharmacokinetics accounted for

    Abbreviations: 17-Ph-PGF2a

    , 17-phenyl-18,19,20-trinorprostaglandin F2

    a; 17-Ph-PGF2

    a-IE, isopropyl ester of 17-Ph-

    PGF2a

    ; ACD logD7, distribution coefficient in pH 7 was calcu-lated using ACD/Labs software; API, active pharmaceuticalingredient; AUC, area under the tissue concentration-timecurve; BBB, blood brain barrier; BCRP, breast cancer resistantprotein; Caco-2, human colon adenocarcinoma cells; CAI, car-bonic anhydrase inhibitor;Cmax, maximum observed concentra-tion in tissues after instillation; CNS, central nervous system;EGTA, ethylene glycol-bis(2-aminoethylether)-N,N,N0,N0-tetra-acetic acid; FDA, U.S. Food and Drug Administration; ICB, iris-

    ciliary body; IOP, intraocular pressure; LAT, large neutralamino acid transporter; log Dxx, logarithm of n-octanol/bufferdistribution coefficient at pH xx; logP, logarithm ofn-octanol/water partition coefficient; MDCK, MardinDarby caninekidney cells; MDR, multidrug resistance protein; MP, meltingpoint; MRP, multidrug resistance-associated protein; NSAID,nonsteroidal anti-inflammatory drug; Papp, apparent cornealpermeability coefficient; PepT1, intestinal peptide transporter1; PGF2

    a, prostaglandin F2

    a; P-gp, P-glycoprotein.

    Correspondence to: Yoshihisa Shirasaki (Telephone: 81-78-997-1010; Fax: 81-78-997-1016;E-mail: [email protected])

    Journal of Pharmaceutical Sciences, Vol. 97, 24622496 (2008)

    2007 Wiley-Liss, Inc. and the American Pharmacists Association

    2462 JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

  • 8/11/2019 Penetracion Ocular

    2/36

    about 40% of the reasons for attrition in 1992, but

    decreased to about 10% in 2000.14,15 Thus, in oral

    drugs, physicochemical-based drug design can

    successfully decrease attrition caused by pharma-

    cokinetic reasons. On the other hand, in ophthal-

    mology, only a few drugs are designed exclusively

    for ocular use.1620 Systemic administration oftenresults in insufficient ocular drug concentration

    because the drug penetration from the blood

    stream to the eye tissues is limited by a blood-

    aqueous and a blood-retinal barrier located in iris

    and retina-choroid, respectively.21,22 To maximize

    drug penetration to the target tissues in parti-

    cular the anterior segment of the eye, topical

    instillation of eye drops are used mainly in clinical

    therapy. The periocular injection methods such as

    sub-conjunctival, sub-tenon and intravitreal

    injection can attain high levels of drugs in

    intraocular tissues, but they are invasive andinconvenient.23 Therefore topical instillation is

    the most useful method because it delivers the

    drug easily and noninvasively to the external and

    intra ocular tissues.

    Ophthalmic drugs in current use originate from

    oral drugs mainly because the adaptation of oral

    drugs to ophthalmic drugs is very efficient in drug

    developments. However, most oral drugs gener-

    ally have low aqueous solubility for ophthalmic

    solutions. The ocular bioavailability of eye drops is

    generally low,21 so high aqueous solubility is

    desirable for eye drops to attain the high drug

    concentration in the formulation, unless it pro-

    duces toxicity such as ocular irritation and

    hyperemia. An alternative option is suspension

    formulation but this is accompanied with pro-

    blems such as inconvenience and technical

    difficulties in manufacturing processes. Ophthal-

    mic suspensions need to be resuspended when

    they are administered. The active pharmaceutical

    ingredients (API) of an ophthalmic suspension

    have to be sterilized. Since most APIs for injection

    have enough aqueous solubility as formulate eye

    drops and are sterilized, such difficulties would

    not be problematic. However, the majority of themis unstable in an aqueous solution for a long time

    and often has low lipophilicity, which may lead

    to low corneal permeability. Because ocular bio-

    availability depends mainly upon pharmaceutical

    formulation, the modification of formulations has

    so far been used as the major approach to improve

    the ocular pharmacokinetics of eye drops.24

    However, if the drug only has poor corneal

    permeability and aqueous solubility, it is difficult

    to deliver sufficient amounts of drugs to intrao-

    cular target tissues using the modification of

    formulation. Therefore, the molecular design with

    consideration of ocular pharmacokinetic and phy-

    sicochemical properties is desirable for ophthal-

    mic drugs to obtain optimal ocular bioavailability

    and efficacies.

    This review summarizes the current state ofknowledge about molecular design for ocular

    drugs and compounds for ophthalmic use origi-

    nating from systemic drugs. I will also consider

    the molecular design to maximize the penetration

    into the anterior segment based on published

    studies to date.

    ABSORPTION ROUTE OF DRUG AFTERTOPICAL OCULAR ADMINISTRATION

    Various anatomic and physiologic barriers limit

    the drug absorption to the anterior segment of the

    eye (cornea, aqueous humor, iris, ciliary body and

    lens). The structure of the eye is shown in

    Figure 1. In general, only 17% of the dose of

    the drugs after topical instillation is able to attain

    the aqueous humor.24 The instilled drug is diluted

    by tear fluid and rapidly removed from the ocular

    surface by tear turnover and blinking. These

    resulted in only a short contact time on the ocular

    surface. The large fraction of the instilled drug

    will be transferred to systemic circulation via thenasolacrimal duct in a few minutes. In the case of

    lipophilic drugs (logP >0), more than 5080% ofinstilled doses are absorbed into the systemic

    circulation.25

    Figure 1. Cross sectional view of the eye.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2463

  • 8/11/2019 Penetracion Ocular

    3/36

    The drug in tear fluid is absorbed via two routes:corneal route and noncorneal route (conjunctiva-

    sclera route).24 Most drugs penetrate to the cornea

    via transcellular absorption as a major route,

    because the corneal epithelium cells form tight

    junctions that limit paracellular drug permea-

    tion.21 The drugs pass through the conjunctivaand the sclera via not only transcellular absorp-tion, but also paracellular absorption, because

    these tissues are more leaky than the cornea. The

    permeability of drugsviatranscellular absorptiondepends mainly on lipophilicity. Most ophthalmic

    drugs with modest lipophilicity and low-molecular

    weight are predominately absorbed via thecorneal route. Animal studies of topical instilla-

    tion have shown that corneal route/noncorneal

    route ratio is 70:1, 12:1, and 5:1 in the case of

    hydrocortisone, timolol and pilocarpine, respec-

    tively.26

    Therefore, it is considered that the corneais the most principle route for ocular drug

    penetration from tear fluid to the anterior

    segment.

    The cornea is very tight tissue, more than the

    intestine, lung, bronchus, and nasal mucosa, and

    the drug penetration is difficult.27 The cornea is

    composed of five membranes: epithelium, Bow-

    mans membrane, stroma, Desmets membrane,

    and endothelium. Among these layers, epithe-

    lium, stroma, and endothelium are the substan-

    tial barriers. The corneal epithelium is a lipophilic

    membrane, which forms tight and gap junctions,

    and the most prominent barrier for corneal

    absorption.28 Therefore, most of the lipophilic

    compounds can pass through the corneal epithe-

    liumvia transcellular absorption. Recent studiesshow that various uptake and efflux trans-

    porters such as oligopeptide transporters, amino

    acid transporters, monocarboxylate transporters,

    nucleoside transporters, P-glycoprotein (P-gp,

    MDR1), MRP1MRP6, and BCRP are expressed

    in cornea epithelium and actively uptake and

    efflux their substrates.21,2934 The stroma is in

    hydrophilic environment and limits the pene-

    tration of highly lipophilic or large molecularweight compounds. The endothelium is a leaky

    lipophilic barrier and partially resists the pene-

    tration of lipophilic compounds, but not hydro-

    philic compounds.

    In the case of the conjunctiva-sclera route,

    drugs can be directly accessed to iris and ciliary

    body through conjunctiva and sclera without

    diffusion to aqueous humor. The conjunctiva is

    a mucous membrane and has many capillary blood

    vessels. The area of human conjunctiva is

    approximately 17-fold larger than that of the

    cornea.26 The conjunctival epithelium forms tight

    junction and limits drug penetration. In conjunc-

    tival epithelium, expression of efflux transporters

    such as P-gp has been reported.26 The sclera,

    which is constructed of collagen bundle and elastic

    fiber, is also a leaky tissue. The scleral perme-ability of polyethylene oligomer is 2-fold less than

    that of conjunctiva and 10-fold more than that of

    the cornea. The conjunctiva and sclera are ever

    leakier than the cornea and permeate the drugs

    through paracellular absorption in addition to

    transcellular absorption.35 The conjunctiva-sclera

    route is generally considered as nonproductive

    route because the vessels in conjunctiva rapidly

    absorbed most of the instilled drug into the

    systemic circulation. However, several reports

    suggest that the conjunctiva-sclera route is the

    main route of penetration to the anterior segmentfor carbonic anhydrase inhibitors (CAIs), hydro-

    philic compounds and large molecules.24

    EVALUATION OF OCULAR PENETRATIONAND REQUIRED PHYSICOCHEMICALPROPERTIES

    Corneal Permeability

    For the sufficient drug penetration into aqueous

    humor, both high corneal permeability and

    aqueous solubility are generally required.36 The

    corneal permeability correlates with lipophilicity

    like Caco-2 and MDCK cell permeability. It is

    reported that the optimal lipophilicity for corneal

    permeation is logP23 in the case of steroids andb-blockers.37,38 Excess lipophilicity would lead to

    a reduction in permeability of corneal stroma,

    which is hydrophilic tissue and limits the pene-

    tration of highly lipophilic compounds. The

    corneal permeability has been evaluated gener-

    ally by using rabbit corneas.39 The corneal

    thickness of rabbits is similar to that of humans.

    Good correlation has been observed betweenrabbit and human corneal permeability in the

    case of both cyclophosphamide40 and CAIs.41 The

    Ussing chamber usually has been used in the

    assessment of corneal permeability. In this

    system, the apparent corneal permeability coeffi-

    cient (Papp) is calculated.36 ThePappvalue can be

    used to evaluate the relative permeability.

    Recently, the in vitro cell systems for predictingcorneal permeability are by determination of

    permeability of compounds through rabbit, bovine

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2464 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    4/36

    and human corneal epithelium cells.4247 The

    permeability of the corneal epithelium cell sys-

    tems is positively correlated with the corneal

    permeability. In addition to passive diffusion, the

    active transport and metabolism may be partially

    evaluated by these systems. The expression of

    various transporters4851 and the enzymes forester hydrolysis on corneal cell lines46,52 has been

    reported. These systems may be useful for screen-

    ing of compound libraries to predict the corneal

    permeability and metabolism.

    Aqueous Solubility

    The high aqueous solubility is also important

    because only the dissolved drug is capable of

    permeating cornea membrane. The instilled drug

    is diluted by tear fluid and contacts with the

    cornea in a very short time.22,24,27,36 The ocular

    bioavailability is generally quite low (17%) and

    high drug concentration is important for ophthal-

    mic solution. Therefore, ophthalmic drugs need

    high aqueous solubility in tears at neutral pH

    (pH 6.57.6) to permeate across the cornea.

    On the other hand, the enhancement of aqueous

    solubility may reduce drug lipophilicity and

    corneal permeability. Although the incorporation

    of a strong ionizable center, such as sulfonate,

    phosphate, and guanidine moieties, into a struc-

    tural template can provide highly soluble mole-

    cules, excessive aqueous solubility would cause amarked reduction in corneal permeability. The

    compounds having ionizable centers should be

    designed not to have excess ionic strength at

    physiological pH. After all, it is desirable to

    possess adequate aqueous solubility without loss

    of lipophilicity for corneal penetration. An excep-

    tion to this is ampholyte CAI, which show higher

    permeability at the higher rate of the ionic species.

    The ionic form more readily sequestered in the

    cornea, which led to the higher drug levels in

    cornea, aqueous humor and iris-ciliary body than

    the unionized form.

    53

    Assessment of Intraocular Penetration

    In vivo ocular pharmacokinetic studies for asses-sing intraocular penetration have most commonly

    been performed with rabbits, because the rabbits

    have relatively large eyeballs in spite of their

    small bodies.39 The corneal permeability for

    lipophilic compounds in rabbits is approximately

    equivalent to that in humans, but when the same

    dosage is topically administrated, intraocular

    drug concentration in rabbits is often higher than

    that in humans. The faster precorneal loss of

    instilled ophthalmic drugs in human ascribed to

    blinking with about three times more frequency.

    The drug penetration across the cornea is higher

    in rabbits than in humans, but it is probable thatrabbits can be used for comparison of intraocular

    penetration for a set of compounds. The aqueous

    humor drug concentrations and area under the

    curve (AUC) can be used as an indicator of

    intraocular tissues drug concentration. The drug

    concentration in aqueous humor would be easy to

    determine due to fluid. Aqueous humor contains

    only low protein content (0.2 mg/mL) compared to

    plasma (50 mg/mL)54 and is approximated to the

    free fraction of drugs in iris and ciliary body

    located around the anterior chamber, which parti-

    cipates in pharmacodynamic effects in intraoculartissues. The intraocular drug levels after instilla-

    tion would be overestimated when the levels are

    obtained in rabbit experiment. Therefore, it is

    suggested that several-times higher ocular levels

    in rabbit are required for the sufficient efficacy in

    human.55

    REPRESENTATIVE EXAMPLES

    Representative examples of corneal permeability

    and ocular penetration of sets of compounds and

    molecular designs for improved ocular pharma-

    cokinetics are described below.

    Anti-Glaucoma Agents

    Carbonic Anhydrase Inhibitors (CAIs)

    Oral CAIs such as acetazolamide and ethoxzola-

    mide have been used for the treatment of

    glaucoma and hyper intraocular pressure (IOP),

    but their systemically adverse effects resulted in

    the discontinuation of the drugs in about 50% of

    patients.56 Therefore, topical instillation of CAIshas been investigated. It is expected that topical

    instillation of CAIs is capable of providing the IOP

    lowering effect without systemically adverse

    effects. However, topical instillation of oral CAIs

    such as acetazolamide, methazolamide and ethox-

    zolamide, did not show sufficient efficacy. For the

    IOP lowering effect, CAIs have to reach the ciliary

    process where aqueous humor is produced by

    carbonic anhydrase. To deliver CAIs to the ciliary

    process, ocular CAIs need to possess the high

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2465

  • 8/11/2019 Penetracion Ocular

    5/36

    corneal permeability and aqueous solubility,

    which lead to high intraocular drug concentration.

    Two major approaches have been used for the

    design of topical CAIs for eye drops. The

    approaches, categorized into ring approach and

    tail approach for enhancing ocular penetration,

    have been tried to obtain the topically effectiveCAIs.5759 The ring approach implies that a

    modification of ring system of lipophilic oral CAIs

    such as ethoxzolamide will improve the aqueous

    solubility. Two topical CAIs presently available

    (dorzolamide18 and brinzolamide)60 were design-

    ed by this approach. On the other hand, in the tail

    approach, the functionalities, which can enhance

    corneal permeability and aqueous solubility, are

    attached to the ring of oral CAIs such as

    acetazolamide and methazolamide without mod-

    ification of the ring.

    Ethoxzolamide, an oral CAI, shows high cornealpermeability, but its aqueous solubility is very

    low.61 The low solubility resulted in insufficient

    intraocular tissue drug concentration for the

    efficacy after topical instillation of aqueous

    suspension. To improve its poor properties, Merck

    Research Laboratories group has designed and

    synthesized many CAI inhibitor for topical instil-

    lation based on ethoxzolamide using the ring

    approach.57 Finally, they identified a topical CAI,

    dorzolamide, which is approved as the first topical

    CAI from FDA. The physicochemical and ocular

    pharmacokinetic properties of several examples of

    these CAIs are shown in Table 1.

    The Merck group focused on conversion of 6-

    position functionalities on ethoxzolamide. The

    conversion of the ethoxy group into the hydroxy

    group led to an increase in aqueous solubility, and

    this phenol L-643,799 demonstrated a weak IOP

    lowering effect in a rabbit model.61 Since this

    conversion lowered the corneal permeability,

    L-643,799 was esterified to the corresponding

    O-pivaloyl derivative L-645,151 to increase cor-neal permeability like dipivefrine62 (dipivaloyl

    ester prodrug of epinephrine).63 This compound is

    efficacious in rabbits, but its repeated instillationfor 3 months induced ocular irritation. This may

    be due to a drug mediated allergic reaction caused

    by the formation of an allergen generated by a

    reaction between the reactive sulfamoyl group at

    2-position on the benzothiazole ring and biomo-

    lecular nucleophiles. Thus, less reactive ben-

    zothiophene analogs have been synthesized.64

    The lipophilicity of the benzothiophene derivative

    L-650,719 is similar to the corresponding ben-

    zothiazole derivative L-643,799. The area under

    the curve of the time-drug concentration profile

    in aqueous humor (AUC) after instillation for

    benzothiophene phenol L-650,719 is approxi-

    mately equal to that for L-643,799.65 The acetyl

    analog of L-650,719 (L-651,465) showed roughly

    twofold higher AUC than the pivaloyl analog of

    L-643,799 (L-645,151). Even though the benzo-thiophenes and benzothiazoles show relatively

    high aqueous solubility, they were only formu-

    lated as suspension at 12%, not ophthalmic

    solution. Thus, the discovery of compounds with

    aqueous solubility exceeding 1% was continued

    and the efforts led to the thienothiopyran

    derivative L-654,230 with more than 1% solubility

    at pH 7.4.66 The 4-hydroxy-thienothiopyran L-

    654,230 showed the 8.1 mg/g ofCmaxafter topicalinstillation in iris-ciliary body (ICB) of pigmented

    rabbits far exceeded the values of benzothiazole

    L-645,151 and benzothiophene L-651,465. Theconversion of the benzothiophene ring into

    the 5,6-dihydro-4H-thieno[2,3-b]thiopyran 7,7-dioxide ring greatly increased aqueous solubility

    without the loss of adequate lipophilicity. The

    substitution of 4-OH functionality in L-654,230

    with isobutylamine moiety provided MK-927 with

    higher Cmax value in ICB than alcohol L-654,230.67 This increase in Cmax value in ICB is

    most likely ascribed to its basic functionality,

    which increases the binding affinity for melanin.

    The further modification of MK-927 identified

    dorzolamide, which demonstrated the similar

    drug levels in ICB after instillation and was

    approved for the treatment of glaucoma by FDA in

    1994.57,58 Thereafter, brinzolamide, which is a

    6-(methoxypropyl)aza analog and is more lipo-

    philic structure than dorzolamide, also received

    FDA approval in 1999 as an anti-glaucoma

    agent.60 The compound shows lower solubility

    at neutral pH than dorzolamide and formulated

    as an aqueous suspension. The drug levels of

    brinzolamide are lower than that of dorzolamide,

    but showed the longer duration of the action with

    the approximately same efficacy as dorzolamide.

    This longer duration may be attributed to thesuspension formulation and the increase in

    lipophilicity caused by the introduction of the

    methoxypropyl group.

    The tail approach consisted of modifying the

    side chains of well-known oral aromatic/

    heterocyclic sulfonamide derivatives, such as

    acetazolamide and itsN-methyl derivative metha-zolamide, to improve the physicochemical and

    ocular pharmacokinetic properties.58,59,68 These

    examples are shown in Table 2. Scozzafava et al.69

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2466 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    6/36

    obtained the topical effective CAIs with highintraocular penetration better than dorzolamide

    by attaching tails such as protonable nitrogen

    atom (EGTA, 2,3-pyridinedicarboxyimido,70 gua-

    nidino,71 and pycolinoyl72) and perfluoroalkyl/

    aryl moieties,73 which provided the molecules

    adequate lipophilicity and aqueous solubility. The

    condensation of 2,6-pyridinedicarboxy acid with

    the C4-amino ethyl group in dorzolamide yielded

    the ocularly permeable CAI with higher intrao-

    cular drug levels than dorzolamide.70 Sharir

    et al.74 reported that the acetyl group of acet-azolamide was substituted with dicarboxylic acids

    such as oxalate, succuinate, adipate and succeed

    in increasing corneal permeability and demon-

    strating the IOP lowering effect (Tab. 3).

    b-Blocker

    All ophthalmic b-blocker drugs on the market

    have been generated by the reformulation of oral

    b-blocker drugs. This class contains various drugs

    Table 1. Physicochemical and Ocular Pharmacokinetic Properties of CAI Inhibitors (Ring Approach)

    Compound log D7.4a Solubility (mg/mL) Cmax (mg/mL or g)

    b Refs.

    Acetazolamide

    0.85 0.71 (pH 7.4) 57,60

    Ethoxzolamide RC2H5, XN 2.06 (pH 7.2) 0.024 (pH 7.4) 61,66

    L-643,799 RH, XN 1.11 7.92 (pH 7.65) 2.67/2.21 (AHc/ICBd)e 57,61,65

    L-645,151 RCOC(CH3)3, XN 2.45 0.058 (pH 7.4) 3.91/4.45 (AHc/ICBd)e 57,63,65

    L-650,719 RH, XCH 1.28 1.16/1.44 (AHc/ICBd)f 57,64,65

    L-651,465 RCOCH3, XCH 1.28 3.05/2.76 (AHc/ICBd)f 57,64,65

    L-654,230 ROH 0.35 12.5 (pH 7.4) 8.1 (ICBd)g 57,66,67

    MK-927 RNHCH2CH(CH3)2 0.90 >20 (pH 7.4) 27.8 (ICBd)g 57,67

    Dorzolamide (L-671,152)

    0.18 6.7 (pH 7.4) 7.8/27.0 (AHc/ICBd)g 18,57,60

    Brinzolamide (AL-4623A)

    0.50 (pH 7.4) 3.85 (ICBd)h 60

    aDistribution coefficient in pH 7.4.bTheCmaxafter topical instillation.cAqueous h umor.dIris-ciliary body.e2% suspension.f0.5% suspension.g2% solution.h1% suspension.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2467

  • 8/11/2019 Penetracion Ocular

    7/36

    with diverse physicochemical properties. Among

    these drugs, the major ophthalmic drugs aretimolol, levobunolol, betaxolol, metipranolol, car-

    teolol, and so on.75 Corneal permeability and

    ocular pharmacokinetics of various oral b-block-

    ers has been evaluated. The lipophilicity and

    pharmacokinetic parameters of these drugs are

    included in Table 4. Schoenwald et al. investi-

    gated the correlations between lipophilicity and

    corneal permeability of 12 b-blockers.38 This

    study revealed that the corneal permeability

    and lipophilicity (logD7.65) exhibited a parabolic

    relationship having the optimal logD7.65at about

    2.5 (Fig. 2). An increase in lipophilicity of thecompound enhances the permeability to the

    cornea epithelium, whereas it does not increase

    the permeability to the corneal stroma due to its

    hydrophilic environment.76 Wang et al.77 also

    investigated the relationship between corneal

    permeability and lipophilicity of 13 b-blockers.

    This study demonstrated that the corneal perme-

    ability varied with lipophilicity according to a

    sigmoidal relationship and was saturated at about

    log P3. However, acebutolol was an outlier in

    Table 2. Physicochemical and Pharmacokinetic Parameters After Topical Instillation of CAI Inhibitors

    (Tail Approach)

    Compound

    logD7.4(CHCl3)

    aSolb (mM)

    (pH 7.4) kinc (103 h1) C1h/C2h

    d (mM)c Refs.

    Acetazolamide (COCH3) 0.001 3.2 0.37 70

    1.45 52 4.6268/53 (AH),

    59/37 (CP) 69

    0.449 81 (HCl) 3.8280/42 (AH),

    50/12 (CP) 70

    1.620 73 4.1308/50 (AH),

    54/18 (CP) 71

    1.944 75 4.7 325/45 (AH), 69/21 (CP) 71

    0.589 78 (HCl) 2.7283/39 (AH),

    51/10 (CP) 72

    2.113 62 4.5324/42 (AH),

    45/13 (CP) 73

    Dorzolamide 2.0 60 (HCl; pH 5.8) 3.0 32/21 (AH), 15/6 (CP) 73

    aDistribution coefficient between chloroform and buffer in pH 7.4.bAqueous solubility.cThe rate constant of transfer across the cornea.dDrug concentration in aqueous humor and ciliary process at 1 and 2 h following instillation of 2% solution.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2468 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    8/36

  • 8/11/2019 Penetracion Ocular

    9/36

    Table 4. Physicochemical and Ocular Pharmacokinetic Properties ofb-Blockers

    Compound log P

    ACD

    logD7a

    Pappb

    (106 cm/s)

    Cmaxc

    (mg/mL)

    AUCc

    (mg h/mL) Refs.

    Penbutolol 4.15 2.05 45 38,76

    Bufuranol 3.65 1.43 57 22.86

    (Ref. 80)

    10.1

    (Ref. 80)

    38,76,80

    Betaxolol 3.44 0.56 27 77

    Propranolol 3.21 1.00 48 5.32

    (Ref. 81)

    3.80

    (Ref. 81)

    38,76,81

    Alprenolol 2.37 0.77 29 38,77

    Levobunolol 2.40 0.77 16

    (Ref. 76),

    23

    (Ref. 77)

    5.92

    (Bunolol)

    (Ref. 83)

    4.56

    (Bunolol)

    (Ref. 83)

    38,76,

    77,83

    Oxprenolol 2.37 0.17 25 (Ref. 76),

    32 (Ref. 77)

    38,76,77

    Metoprolol 1.88 0.33 22

    (Ref. 76),28

    (Ref. 77)

    2.89

    (Ref. 81)

    2.01

    (Ref. 81)

    38,76,

    77,81

    Timolol 1.91 1.77 12

    (Ref. 76),

    12

    (Ref. 77)

    30.65

    (Ref. 80),

    5.86

    (Ref. 81)

    25.4

    (Ref. 80),

    3.73

    (Ref. 81)

    38,76,77,

    80,81

    (Continued)

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2470 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    10/36

    prodrugs might be superior in pharmacological

    activity to nonprodrugs ascribed to the presence of

    much free fraction. Conversion to aliphatic acid

    ester prodrugs of tilisolol, a hydrophilic b-blocker,

    also provided an improved corneal permeability

    and intraocular penetration. The propionyl and

    butyryl ester prodrugs demonstrated approxi-

    mately sixfold higher tilisolol concentration at1 h after instillation in aqueous humor than the

    parent compound.87

    Oxime/methoxime analogs of b-blocker have

    been reported as classes of prodrugs, except for

    esters. Bodor et al. synthesized oxime/methoxime

    analogs of alprenolol, betaxolol, carteolol, propra-

    nolol, timolol, etc. Some of these compounds

    demonstrated higher and longer IOP reducing

    efficacy than the parent drugs.20 For instance,

    after instillation the oxime analog of propranolol

    Table 4. (Continued )

    Compound log P

    ACD

    logD7a

    Pappb

    (106 cm/s)

    Cmaxc

    (mg/mL)

    AUCc

    (mg h/mL) Refs.

    Acebutolol 1.77 0.11 0.85

    (Ref. 76),1.1

    (Ref. 77)

    1.26

    (Ref. 80)

    2.93

    (Ref. 80)

    38,76,

    77,80

    Pindolol 1.75 0.18 10 77

    Nadolol 0.93 0.83 1.0

    (Ref. 76)

    38,76

    Atenolol 0.16 2.02 0.67

    (Ref. 76)

    2.22

    (Ref. 81)

    0.93

    (Ref. 81)

    38,76,81

    Sotalol 0.62 1.82 1.6

    (Ref. 76)

    38,76

    aDistribution coefficient in pH 7 was calculated using ACD/Labs software.bCorneal permeability.cThe values in aqueous humor after single81,83 or triple80 instillation normalized to 1% solution dosing.

    Figure 2. Relationship between logarithm of cor-

    neal permeability coefficient (Papp) and lipophilicity

    (logD7.65) in the rabbit intact cornea. This graph was

    reconstructed from the graph in Ref. 38.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2471

  • 8/11/2019 Penetracion Ocular

    11/36

    Table 5. Physicochemical and Ocular Pharmacokinetic Properties of Timolol86 and Tilisolol Prodrugs87

    Compound log D7.4a Papp (10

    6 cm/s)b C20minc or C1h

    d (mM)

    Timolol prodrugs

    RH 0.04 8.1 7.7c

    RCOCH3 1.1 23 23c

    RCOCH2CH3 1.6 29 19c

    RCO(CH2)2CH3 2.1 32 22c

    RCOC(CH3)3 2.7 13 17c

    RCO(CH2)3CH3 2.7 31 26c

    RCO(CH2)4CH3 3.3 21 21c

    RCO(CH2)6CH3 4.4 8.9 12c

    Tilisolol prodrugs

    RH 0.27 2.7 3.1d

    RCOCH3 1.02 8.9 4.0 (vs. parent)d

    RCOCH2CH3 1.56 8.4 5.1 (vs. parent)d

    RCO(CH2)2CH3 2.02 15 6.2 (vs. parent)d

    RCO(CH2)3CH3 2.47 13 5.9 (vs. parent)d

    aDistribution coefficient in pH 7.4.bCorneal permeability. The data were constructed from the graph in Ref. 86.cDrug concentration in aqueous humor at 20 min after instillation. The data were constructed from the graph in Ref. 86.

    dDrug concentration in aqueous humor at 1 h after instillation.

    Figure 3. Relationship between corneal permeability

    coefficient (Papp) and prodrug lipophilicity (logD7.4) in

    the rabbit intact cornea. This graph was reconstructed

    from the graph in Ref. 86.

    Figure 4. Relationship between aqueous humor

    timolol concentration at 20 min and prodrug lipophili-

    city (logD7.4) in the rabbit intact cornea. This graph was

    reconstructed from the graph in Ref. 86.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2472 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    12/36

    showed higher intraocular penetration and longer

    duration of action by sustainable release of the

    parent drug (Tab. 6).88 Since release of parent

    compound for oxime/methoxime analogs is slower

    than that of ester prodrugs, conversion to oxime/

    methoxime is useful for continuation of the

    duration of action.

    In the case ofb-blockers that possess relatively

    low lipophilicity and hydrophilic functionalities,

    such as hydroxyl and secondary amine group,

    increasing lipophilicity of the molecule would

    improve the ocular penetration. However, exces-

    sive lipophilicity would cause an increase in

    ocular tissue binding, leading to a decrease in

    unbound fraction and an increase in resistance

    to hydrophilic corneal stroma. For this reason,

    the highly lipophilic molecule is unsuitable for

    ocular drugs as ophthalmic solution. It is most

    likely to be effective for ocular drugs to in-

    troduce a heterocycle, which allows the moleculesto increase lipophilicity without loss of water

    solubility. The ester prodrug approach is also

    desirable because the molecule will be converted

    into the less lipophilic parent compound in corneal

    epithelium.

    a2-Agonist

    a2-Agonist showed an IOP lowering effect and is

    used for the treatment of glaucoma. Clonidine, a

    representative selective a2-agonist, has been used

    as an ocular hypertensive agent via topicaladministration. However, it can also penetrate

    to the central nervous system (CNS) through the

    blood-brain barrier (BBB) due to its high lipo-

    philicity and causes centrally mediated cardio-

    vascular side effects.89 Therefore, to reduce the

    adverse effect, p-aminoclonidine (apraclonidine),which possess higher polarity than clonidine, was

    synthesized and is used for reducing the IOP. The

    introduction of the p-amino group resulted in areduction of adverse effects, but also a decrease of

    corneal permeability because of an increase in

    polarity.

    Brimonidine, a quinoxaline derivative, is more

    lipophilic than apraclonidine but less lipophilic

    than clonidine.90 Conversion of 2,6-dicloro-p-aminobenzene into 5-bromoquinoxaline provided

    more than 20-fold higher corneal permeability

    and 10-fold higher drug levels in aqueous humorafter instillation (Tab. 7). Although this drug

    passes BBB, its side effects are tolerable.91 This

    may be due to less penetration to the CNS than

    clonidine in addition to the receptor subtype

    selectivity. On the other hand, to minimize access

    the CNS without a loss of ocular penetration,

    derivation of brimonidine has been reported.

    Munk et al. synthesized brimonidine analogs that

    possessed the 5-methyl group instead of the 5-

    bromo group.92 The methyl derivative AGN

    Table 6. Physicochemical and Ocular Pharmacokinetic Properties of Propranolol and its Oxime Prodrug88

    Compound ACD log D7a

    Concentration (mg/mL or g)b

    0.5 h 1 h

    Propranolol

    3.21 1.28 (AHc)

    8.05 (ICBd)

    0.26 (AHc)

    0.00 (ICBd)

    Propranolone oxime

    3.27 0.86e (0.04f) (AHc)

    9.90e (2.11f) (ICBd)

    1.51e (0.71f) (AHc)

    1.79e (1.79f) (ICBd)

    a

    Distribution coefficient in pH 7 was calculated using ACD/Labs software.bDrug concentration after instillation at 0.5 or 1 h.cAqueous h umor.dIris-ciliary body.eTotal concentration (prodrugpropranolol).fPropranolol concentration.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2473

  • 8/11/2019 Penetracion Ocular

    13/36

    191103 and its corresponding benzodioxane deri-

    vative AGN 192836 crossed the BBB, but the

    structurally related benzoxazin derivative AGN

    193080 did not. This benzoxazin derivative had

    higher IOP lowering efficacy than clonidine. This

    suggests that AGN 193080 shows good corneal

    penetration with low BBB penetration. The

    tetrahydroquinoxaline derivative AGN 192172

    demonstrated neither IOP lowering efficacy nor

    BBB penetration. These data indicated that the

    introduction of morpholine ring may be useful for

    decreasing BBB penetration without corneal

    penetration.

    Prostaglandin (PG) F2aDerivatives

    Topical administration of PGF2a lowered the

    elevated IOP of glaucoma patients.89,93 Ocular

    Table 7. Physicochemical and Ocular Pharmacokinetic Properties ofa2 Agonists

    Compound log D7.4a Papp

    b (106 cm/s) C1h (mg/mL or g)c (Aqueous Humor/Iris) Refs.

    Chlonidine

    0.52 36 11.53/13.87 90

    Aprachlonidine

    0.96 0.44 0.59/0.87 90

    Brimonidine (AGN190432)

    0.17 9.8 7.67/7.84 90

    AGN 191103

    3.40 91

    AGN 192836

    91

    AGN 193080

    0.80 92

    AGN 192172

    3.90 92

    aDistribution coefficient values in pH 7.4.bCorneal permeability.cDrug concentration in aqueous humor at 1 h after instillation.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2474 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    14/36

    penetration of PGF2a is limited because it will

    exhibit an ionized form in tears at neutral pH due

    to the presence of the carboxylic acid function-

    ality.94 Esterification of the carboxyl moiety at

    1-position significantly increased the corneal

    permeability and the IOP lowering activity

    (Tab. 8).9598 Since esters of PGF2a are metabo-lized to the free acid in the cornea, they act as

    prodrugs. Esterifications of alcohol moiety at

    11- and/or 15-position of PGF2a also provided

    prodrugs, which showed an enhanced corneal

    permeability.98 However, 11,15-diester had lower

    corneal permeability than 11- or 15-monoester.

    Di-esterification may cause the decline of perme-

    ability to the corneal epithelium and hydrophilic

    stroma because it leads to an increase inmolecular weight and an excessive lipophilicity

    causing strong interaction between the molecule

    Table 8. Corneal Permeability and ACD logD7 of PGF2a and Related Compounds

    Compound

    Papp (106 cm/s)a

    ACD logD7bRabbit Human Porcine

    PGF2a derivatives

    PGF2a R1R

    11OH, R15OH 0.2

    c/0.13d 1.7e 0.13f 0.09

    R1R11OH, R

    15OCOCH3 1.0c

    R1R11OH, R

    15OCOC(CH3)3 5.2c

    R1OCH3, R11H, R

    15OH 8.9g 2.70

    PGF2a isopropyl ester (PGF2a) R1OCH(CH3)2, R

    11OH, R15OH 19

    c 3.3e 29f 3.58

    R1OCH2Ph, R11OH, R

    15OH 26g 4.45

    R1

    R11

    1,11-Lactone, R15

    OH 18c 2.80

    R1OH, R11COC(CH3)3, R

    15OH 7.7e 2.27

    R1OH R11R

    15OCOC(CH3)3 4.0c 2.6e 4.33

    S-1033 (15-deoxy PGF2a) R1R

    11R15OH 0.58

    d 1.77

    S-1033 methyl ester R1OCH3, R11R

    15OH 1.3d 4.55

    Isopropyl unoprostone R1OCH(CH3)2 0.95h 4.63

    Unoprostone R1OH NDh,i 0.97

    aCorneal permeability.bDistribution coefficient was calculated using ACD/Labs software.cRef. 97.dRef. 102.eRef. 98.fRef. 96.gRef. 95.hRef. 104.iThe drug was not detected in the receiver cell.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2475

  • 8/11/2019 Penetracion Ocular

    15/36

    and membrane lipids of corneal epithelium. The

    9-pivaloyl ester and 9,11-lactone of PGF2a were

    unsuitable for prodrugs, because they were not

    substantially hydrolyzed by ocular tissue homo-

    genates.99

    S-1033, a 15-deoxy derivative of PGF2a, showed

    higher permeability than PGF2a. Removal of thepolar hydroxyl group led to the increased perme-

    ability. Additionally, esterification of the 1-car-

    boxyl moiety resulted in a further increase in

    corneal permeability (Tab. 8).100

    PGF2a 1-ester analogs show high corneal

    permeability and IOP-lowering activity, but are

    not suitable for clinical use due to side effects such

    as irritation and conjunctival hyperemia. There-

    fore, to reduce the side effects, several v-side

    chain (1320 position) analogs of PGF2a have been

    synthesized. The addition of two carbons at the

    20-position of a PGF2ametabolite (15-keto-13,14-dihydro PGF2a) provided a compound (unopros-

    tone) with an improved side-effect profile in the

    eye and without a loss of IOP-lowering activ-

    ity.101103 Unoprostone is a carboxylic acid and

    showed quite low corneal permeability (Tab. 8).

    Therefore, its isopropyl ester (isopropyl unopros-

    tone, UF-021) is marketed as an anti-glaucoma

    agent.104 Substitution of carbons at positions

    1820 of PGF2a with phenyl or at positions

    1720 with phenoxy moiety also improved the

    side-effect profile.105107 17-phenyl-18,19,20-tri-

    nor PGF2a

    (17-Ph-PGF2a

    ) and 16-(3-chlorophe-

    noxy)-17,18,19,20-tetranor PGF2a (cloprostenol)

    demonstrated a comparable corneal permeability

    to PGF2a(Tab. 9).108 The isopropyl ester of 17-Ph-

    PGF2a (17-Ph-PGF2a-IE) exhibited lower corneal

    permeability than the isopropyl ester of PGF2a(PGF2a-IE). Latanoprost, the 13,14-dyhydro

    analog of 17-Ph-PGF2a-IE, showed improved

    pharmacological profiles and is widely used as

    anti-glaucoma agent.10 Amidation of 17-Ph-

    PGF2a is also effective for increasing corneal

    permeability, but showed lower impact on corneal

    permeability than esterification.109 This may be

    because an amide group is a hydrogen bonddonor. Since the hydrolysis rate of amides is

    generally slower than that of esters, amide

    derivation is not suitable for prodrugs except for

    nonsubstituted amide (CONH2), which showed a

    somewhat higher hydrolysis rate than mono-

    substituted amides (CONHR).110 Bimatoprost,

    an ethyl amide derivative of 17-Ph-PGF2a, is used

    for the treatment of glaucoma and is most likely to

    show an ocular hypotensive effect without meta-

    bolism to the corresponding free acid.109 Con-

    version of the hydroxyl group at position 15 of

    17-Ph-PGF2a-IE into a carbonyl group resulted in

    an increase in corneal permeability. Since the con-

    version did not significantly change the lipophi-

    licity, the reduction of the number of hydrogen

    bond donor would enhance the permeability.108

    To enhance the corneal permeability of PGF2aand its analogs, esterification and amidation of

    the C1-carboxyl functionality is the most desirable

    derivation. Esterification of the alcohol moiety

    can also increase the corneal permeability due to

    an increase in lipophilicity and a reduction in the

    number of hydrogen bond donors. All ocular

    PGF2aanalog prodrugs on the market (isopropyl

    unoprostone, latanoprost and travoprost) are

    isopropyl esters, which would show suitable stabi-

    lity in aqueous solution due to its bulky structural

    nature.104,107,108 Because amide derivatives are

    also generally stable for hydrolysis, they are sup-erior to ester derivatives in the self-life.

    Anti-Infective Agents

    Fluoroquinolones

    Fluoroquinolone is a class of anti-bacterial agents,

    which are widely used for the treatment of ocular

    infection as eye drops.111115 Increasing the

    lipophilicity of fluoroquinolones has a tendency

    to increase corneal penetration (Tab. 10). Robert-

    son et al. reported that the corneal permeability ofseven fluoroquinolones (which are used as

    eye drops) is positively correlated with their

    MardinDarby canine kidney (MDCK) cell perme-

    ability.112 Moxifloxacin demonstrates the highest

    ocular penetration in commercial products used as

    eye drops.112115

    Antiviral Agents (Acyclovir/Ganciclovir)

    The acyclic guanosine analogs acyclovir (ACV)

    and ganciclovir (GCV) are clinically used in the

    treatment of various infections caused by the

    herpes family of viruses.116,117 However, thesedrugs have low ocular permeability due to their

    hydrophilic nature and did not show sufficient

    efficacy for intraocular infection via topical

    instillation.118,119 Therefore, the lipophilic pro-

    drug approach of these drugs was investigated.

    Esterification of ACV and GCV with fatty acids

    has improved the corneal permeability (Tabs. 11

    and 12).120125 These esters are rapidly hydro-

    lyzed to the parent drugs in the cornea. It is shown

    that increasing the length of an alkyl side chain of

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2476 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    16/36

    acyclovir ester prodrug from propionyl to hex-

    anoyl leads to the gradually enhancement of

    ocular penetration in vivo in addition to in vitrocorneal permeability.126

    The conversion of ACV and GCV into their L-

    valine ester (valacyclovir127 and valganciclovir,128

    respectively) or into dipeptide esters having

    specific amino acid sequences remarkably

    enhance the corneal permeability.129,130 These

    derivatives possess a free amino group and show

    high aqueous solubility but low lipophilicity.

    However, their corneal permeability is higher

    than that expected from their lipophilicity. This

    may arise from an active uptake by amino acid or

    Table 9. Corneal Permeability and ACD logD7 of 17-Ph PGF2a and Related Compounds

    Compound

    Papp (106 cm/s)a

    ACD logD7bHuman Porcine

    17-Ph PGF2a derivatives

    17-Ph PGF2a(17-Ph-18,19,20-trinor PGF2a)

    R1OH

    0.696c 0.10

    Bimatoprost

    R1NHC2H5 3.24d 1.98

    R1

    OCH(CH3)2 5.9e

    3.56

    15-Keto-17-Ph PGF2a 11.0e 3.34

    Latanoprost (13,14-dihydro-17-Ph PGF2a) 6.8e 3.65

    Cloprostenolol 1.49c 0.09

    aCorneal permeability.bDistribution coefficient was calculated using ACD/Labs software.cRef. 98.dRef. 110.eRef. 108.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2477

  • 8/11/2019 Penetracion Ocular

    17/36

    oligopeptide transporters in corneal epithelium

    such as LAT 1, LAT 2, and PepT1.131 Therefore,

    such amino acid and dipeptide prodrug appro-

    aches are useful for increasing ocular penetration.

    The corneal permeability of these derivatives is

    higher than that of aliphatic ester prodrugs. The

    rank order of ocular penetration was Val-Gly >

    Val>Tyr-Val>Val-Val in the case of acyclo-

    vir amino acid or dipeptide esters.132 In the case of

    ganciclovir amino acid or dipeptide esters, the

    rank order of ocular penetration was Val-Tyr >

    Val-Val>Val>Gly-Val.122

    Table 10. Physicochemical and Ocular Pharmacokinetic Properties of Fluoroquinolone Derivatives

    Compound Sol. (%)a

    Papp (107 cm/s)

    logD7.4

    AQCmax (mg/mL)d

    AUC01 (mg h/mL)e

    MDCKb Corneac Ref. 115 Ref. 114 Ref. 113

    Norfloxacin

    0.05 3.3 1.63 1.60 0.21,

    0.92

    0.22,

    0.92

    Ciprofloxacin

    0.02 4.5 2.46 1.52 0.64,

    3.18

    Lomefloxacin

    0.13 6.6 3.58 1.45 1.25,

    4.22

    Ofloxacin

    0.35 15.1 6.78 0.45 1.15,

    3.20

    1.59,

    5.08

    0.87,

    2.24

    Gatifloxacin

    0.21 10.3 4.6 0.97 2.30,

    5.90

    1.26,

    2.52

    Moxifloxacin

    >6.43 35.2 15.8 0.23 5.42f,

    7.34f

    aAqueous solubility.bMDCK cell permeability (Ref. 112).cCorneal permeability (Ref. 112).dDrug concentration in aqueous humor after three times topical dosing to rabbit at 15 min interval.eThe area under the curves for 0 h to infinity.fAQCmaxand AUC values of moxifloxacin (0.5% solution) were normalized to 0.3% solution.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2478 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    18/36

    Anti-Inflammatory Agents

    Nonsteroidal Anti-Inflammatory Drugs (NSAIDS)

    Most of NSAIDS for eye drops have arisen from

    oral NSAIDS such as diclofenac,133 bromfenac,134

    flurbiprofen,135 pranoprofen,136 and ketorolac

    tromethamine.137 Ocular penetration of these

    drugs is not very high due to the incorporationof a carboxyl functionality that will completely

    ionize in tears at a neutral pH. In addition to these

    NSAIDS adapted from oral drugs, nepafenac, the

    NSAID used only for eye drops, was recently

    approved by FDA in 2005. Nepafenac is nonsub-

    stituted amide prodrug of amfenac,138 which is

    used as an oral drugs for the treatment of

    rheumatoid in Japan, and was designed to

    improve the corneal permeability and tissue

    distribution profile.139 Nepafenac showed about

    4- to 30-fold higher corneal permeability than

    conventional NSAIDS such as diclofenac, bromfe-

    nac and ketorolac (Tab. 13).140 The permeability

    data of amfenac were not given, but amidation of

    carboxylic acid in amfenac would result in at least

    a 28-fold increase in permeability than the parent

    drug, based on the estimation from permeability

    of ketorolac and ACD logD7 value of ketorolac andamfenac. Since nepafenac showed low aqueous

    solubility, it was formulated as an aqueous

    suspension. Its high permeability enables it to

    deliver to posterior segment of the eye in addition

    to the anterior segment. Moreover, its duration of

    action is longer than that of diclofenac.141 Con-

    version of carboxylic acid into nonsubstituted

    amide is useful for the enhancement of corneal

    permeability, although it leads to a decrease in

    aqueous solubility. Amides are most likely to be

    Table 11. Physicochemical and Pharmacokinetic Properties of Ganciclovir Ester Derivatives

    Compound

    Solba

    (mM)

    logD7.4b or

    ACD logPcPapp

    (106 cm/s)dCmax

    (mM)e,fAUCinf

    (mM min)e,g Refs.

    Aliphatic fatty acid esters

    RH 15.7 1.55b 3.8 119

    RCOCH3 15.2 1.08b 4.9 119

    RCOCH2CH3 11.9 0.92b 5.7 119

    RCO(CH2)2CH3 8.4 0.30b 7.7 119

    RCO(CH2)3CH3 4.1 0.07b 24 119

    a-amino acid or dipeptide esters

    RH 3.4 2.07c 4.1 201 42259 130,132

    RVal 92 1.28c 32 647 82112 130,132

    RValVal 82 0.73c 31 943 301370 130,132

    RValTyr 74 0.55c 1458 536278 130,132

    RTyrVal 68 0.54c 130

    RValGly 63 1.95c 130

    RGlyVal 66 1.95c 109 34460 130,132

    RTyrGly 68 1.77c 130

    RGlyTyr 74 1.78c 130

    aSolubility in pH 4.2 phthalate buffer.bDistribution coefficient values in pH 7.4.cPartition coefficient was calculated using ACD/Labs software.

    dCorneal permeability.eAfter 2 h of corneal infusion (0.43%, 200mL) via topical well.fThe maximum concentration of GCV in aqueous humor.gThe area under the curve for 0 h to infinity in aqueous humor.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2479

  • 8/11/2019 Penetracion Ocular

    19/36

    hydrolyzed slower than ester and are metabolized

    in iris-ciliary body and choroid/retina rather than

    the cornea. However, in the case of a more

    lipophilic NSAID such as flurbiprofen, amidation

    did not change the corneal permeability

    (Tab. 13).142

    Steroids

    Prednisolone and dexamethasone derivatives areused for the treatment of ocular inflammation.

    Their acetyl and phosphate esters at the 21-

    position of prednisolone are widely used as eye

    drops. Both esters of prednisolone rapidly repro-

    duced the parent drug in the cornea (Tab. 14). The

    purpose of acetylation is to enhance the corneal

    permeability, while the purpose of phosphoryla-

    tion is to increase the aqueous solubility. The

    corneal permeability of the acetate and phosphate

    are 30-fold higher and 10-fold lower than that of

    their parents, respectively.143 Although the phos-

    phorylation greatly decreased its corneal perme-

    ability, the AUC06h values in aqueous humor

    after instillation of phosphate 0.5% solution to

    rabbits were approximately equal to that after

    instillation of acetate 0.5% suspension possibly

    due to the great increase in aqueous solubility.144

    Esterification of dexamethasone at the 21-

    position with fatty acids also increases the corneal

    permeability (Tab. 15).145 Increasing the alkylchain from acetyl to butyryl gradually enhanced

    the corneal permeability. The valeryl ester is

    slightly resistant to hydrolysis, but its flux of

    dexamethasone through the cornea is similar to

    that of butyryl ester. The palmitoyl ester did not

    permeate substantially to the cornea. Since the

    lipophilicity of palmitoyl ester is too high (log

    P12.4), it may be trapped in the cornealmembrane. The phosphate and m-sulfobenzoateprodrugs of dexamethasone have been marketed

    Table 12. Physicochemical and Pharmacokinetic Properties of Acyclovir Ester Derivatives

    Compound

    Solba

    (mM)

    logD7.4b/

    ACD logD7c

    Papp(106 cm/s)d

    C25 mine or

    Cmaxf,g (mM)

    AUCinff,h

    (mM min) Refs.

    Aliphatic fatty acid esters

    RH 11.2 1.22b/1.76c 3.7 38e 118,126

    RCOCH2CH3 5.0 0.85b/0.57c 4.3 118,126

    RCO(CH2)2CH3 4.6 0.08b/0.04c 5.1 42e 118,126

    RCOCH2CH(CH3)2 4.8 0.06b/0.22c 3.9 118,126

    RCO(CH2)3CH3 1.5 0.30b/0.49c 6.5 59e 118,126

    RCOC(CH3)3 1.6 0.37b/0.13c 118,126

    RCO(CH2)4CH3 0.7 0.93b/1.02c 8.5 79e 118,126

    a-amino acid or dipeptide esters

    RH >30 1.76c 4.2 129

    RVal >30 1.71c 12 124g 7247 122,129

    RValVal >30 1.52c 9.9 34g 2063 122,129

    RValGly >30 2.27c 12 200g 12007 122,129

    RValTyr >30 0.84c 7.2 129

    RTyrVal >30 1.33c 8.3 80g 13930 122,129

    aSolubility in pH 7.4 phosphate buffer.bDistribution coefficient values in pH 7.4.cDistribution coefficient in pH 7 was calculated using ACD/Labs Software.dCorneal permeability.eACV concentration in aqueous humor at 25 min after instillation of 50mL of a 1 mMsolution. The datawereconstructedfromthe

    graph in Ref. 126.f

    After 2 h of corneal infusion (200mL)via topical well.gThe maximum concentration of ACV in aqueous humor.hThe area under the curve for 0 h to infinity in aqueous humor.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2480 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    20/36

    as water-soluble derivatives. The corneal perme-

    ability of phosphate andm-sulfobenzoate is about1.3 and 10 times less, respectively, than that of the

    parent. The 2.5 mM solution of the phosphate

    partly penetrates into aqueous humor after

    instillation. The AUC of the phosphate was

    twofold less than that of the 2.5 mM dexametha-

    sone suspension and threefold less than that of the

    butyrate suspension. In contrast, the m-sulfo-benzoate solution was not detectable in aqueous

    humor probably due to its low corneal perme-

    ability.

    Table 13. Physicochemical and Pharmacokinetic Properties of NSAIDs

    Compound ACD log D7a Corneal permeability Papp (10

    6 cm/s) Refs.

    Nepafenac

    1.17 64 140

    Amfenac

    0.62

    Bromfenac

    0.30 3.4 140

    Diclofenac

    1.28 15 140

    Ketorolac

    0.60 2.3 140

    Flurbiprofen

    1.31 21 142

    Flurbiprofen amide

    3.06 22 142

    aDistribution coefficient in pH 7 was calculated using ACD/Labs Software.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2481

  • 8/11/2019 Penetracion Ocular

    21/36

    Table 14. Ocular Pharmacokinetic Properties of Prednisolone Derivatives143,144

    Compound Papp (106 cm/s)a Cmax (mg/mL)

    b AUC06h (mg h/mL)c

    Prednisolone

    RH

    2.7

    Prednisolone disodium phosphate

    RP(O)(ONa)2 (1% solution)

    0.26 1.5 (2.1)d 3.7 (4.5)d

    Prednisolone acetate

    RCOCH3 (1% suspension)

    83 1.6 4.2

    aCorneal permeability.bPrednisolone concentration in aqueous humor after instillation of prednisolone prodrugs.c

    The area under the curve for 06 h in aqueous humor.dTotal concentration (prodrugprednisolone).

    Table 15. Physicochemical Ocular Pharmacokinetic Properties of Dexamethasone Derivatives145

    Compound log Pa Papp (106 cm/s)b Cmax (ng/mL)

    c AUC03h (ngh/mL)d

    Dexamethasone

    RH

    2.12 5.1 75 104

    Dexamethasone

    disodium phosphate

    RP(O)ONa2

    0.54 3.9 39 54

    Dexamethasone

    sodium m-sulfobenzoate

    R

    1.65 0.51

  • 8/11/2019 Penetracion Ocular

    22/36

    Fatty acid esterification of steroids is extremely

    effective for enhancing of corneal permeability.

    Phosphorylation of steroids at the 21-position can

    provide highly water-soluble prodrugs. Although

    the phosphorylation led to a great decrease in the

    corneal permeability, its drug penetration into

    aqueous humor is similar or slightly lower thanthe parent drug due to compensation by the

    increased aqueous solubility. It is expected that

    the increasing drug concentration in formulation

    will produce large concentration gradients and

    high ocular penetration. Esterification with m-sulfobenzoic acid also resulted in an increase in

    aqueous solubility, but its ocular penetration

    would be reduced greatly because of a decrease

    in corneal permeability.

    OthersCalpain Inhibitor

    Calpains, a family of cysteine endoproteases,

    degrade lens proteins such as crystalline. There-

    fore, calpain inhibitors are studied as a potential

    anti-cataract agent.146148 SJA6017, a dipeptidyl

    aldehyde, shows very high potency against two

    major calpain isoforms.149 However, instillation

    of SJA6017 aqueous suspension showed only low

    concentration in aqueous humor (Tab. 16).150

    The low ocular penetration of SJA6017 may be

    in part due to the presence of the reactive

    aldehyde group, which may form reversible co-

    valent adducts with nucleophiles in biomolecules.

    Consequently, to investigate calpain inhibitors

    that have the superior corneal permeability,

    Nakamura et al.150 modified the structure of

    SJA6017 using two approaches. One approach

    was to introduce into a pyridine ring as a water-

    solubilizing group. Another is to convert the

    reactive aldehyde moiety with hemiacetal moiety

    that is a masked aldehyde with less reactivity.

    Both approaches increased the ocular penetra-

    tion. 3-Pyridlyacetomide analog (SNJ-1664) and

    hemiacetal analog (SNJ-1709) of SJA6017 showeda six- and eight-fold higher ocular penetration,

    respectively, following instillation of their suspen-

    sions than SJA6017. Furthermore, the conversion

    of a (4-fluorophenyl)sulfonamide moiety of SNJ-

    1709 into a phenylthiourea moiety provided a

    compound (SNJ-1715) with about 30-fold higher

    increase of AUC in aqueous humor. SNJ-1709 and

    SNJ-1715 have the similar aqueous solubility and

    log P values, but have significantly differentmelting points (about 162 and 628C, respectively).

    The superior ocular penetration of the thiourea

    derivative is possibly due to the difference in

    melting point. The low melting point may be

    capable of enhancing corneal permeability

    through an increase in the dissolution rate. A

    good positive linear correlation between disso-

    lution rate and oral bioavailability has beenreported.151 It was also reported that transdermal

    absorption of drugs correlated with their melting

    points.152 In the case of suspension formulation,

    the difference in dissolution rate greatly affects

    the ocular penetration because of the short con-

    tact time between the drugs and the corneal

    surface.153

    Moreover, Nakamura et al.154 reported the

    cyclic hemiacetal analogs (Tab. 16). A cyclic

    hemiacetal SNJ-1757 in this series showed a

    3.5-fold increase in corneal permeability in vitro

    than did the corresponding linear aldehyde(dehydroxy analog SNJ-1770). Both compounds

    were evaluated for reactivity with semicarbazide

    (H2NNHCONH2) hydrochloride, which simulates

    biomolecules containing nucleophiles such as

    the NH2 and SH groups. The cyclic hemiacetal

    SNJ-1757 did not significantly react with semi-

    carbazide, whereas the corresponding linear

    aldehyde SNJ-1770 immediately reacted and

    formed a semicarbazide adduct. These results

    suggested that the presence of aldehyde moiety is

    a limiting factor for corneal permeability.

    Conversion of the aldehyde into the hemiacetal

    provided an increase in corneal permeability,

    which may be ascribed to a reduction of the

    electrophilicity. A decline of electrophilicity by

    structure modification or formation of prodrug

    is desirable for enhancement of the corneal

    permeability.

    Quinidine

    Quinidine is not an ocular drug, but has been

    investigated for the corneal permeability as a

    model P-gp substrate. Jain et al.155 attempted

    the transporter-targeted prodrug derivatization.Esterification of the hydroxyl group of quinidine

    with L-valine or L-valyl-L-valine showed a 1.5- and

    3-fold increase in the corneal permeability com-

    pared to quinidine (Tab. 17).156 The corneal

    permeation of quinidine is most likely to be

    limited by efflux, via P-gp on the corneal epi-

    thelium. Attaching L-valine or L-valyl-L-valine to

    quinidine not only significantly decreases the

    affinity to P-gp but also may increase the affi-

    nity to oligopeptide transporters. Such prodrug

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2483

  • 8/11/2019 Penetracion Ocular

    23/36

    approaches are useful for the improvement ofcorneal permeability of P-gp substrates.

    APPROACHES FOR MOLECULAR DESIGN TOENHANCE OCULAR PENETRATION

    As mentioned above, to enhance the ocular

    penetration of drugs, aqueous solubility and

    corneal permeability can be important factors.

    Approaches for molecular design to improve these

    factors are described below. There are roughly two

    classifications of these approaches: the structuremodification of molecules themselves and the

    prodrug approach. These strategies are summar-

    ized in Table 18.

    Structure Modification Approach(Nonprodrug Approach)

    Ophthalmic drug compounds require not only

    high aqueous solubility but also adequate lipo-

    philicity to penetrate to the membranes. Although

    Table 16. Physicochemical and Ocular Pharmacokinetic Properties of Calpain Inhibitors

    Compound

    Solb

    (mg/mL)a logD7b

    Papp(106 cm/s)c

    Cmax(mg/mL)d

    AUC

    (mg h/mL)e Refs.

    SJA6017

    0.10 1.7 ND 0.037 0.051 150

    SNJ-1664

    >100 ND 0.21 0.31 150

    SNJ-1709

    1.5 0.60 ND 0.21 0.39 150

    SNJ-1715

    1.5 0.70 1.1 1.0 1.7 150

    SNJ-1757

    2.0 0.38 19 ND ND 154

    SNJ-1770

    0.91 0.54 5.4 ND ND 154

    aSolubility in pH 7 phosphate buffer.bDistribution coefficient in pH 7.cCorneal permeability.dAqueous humor concentration after instillation of 50mL of 0.5% suspension.eThe area under the curve for 03 h.

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2484 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    24/36

    an introduction of strong ionizable centers, such

    as phosphate, sulfonate, carboxylate and guani-

    dino groups increases the aqueous solubility, it

    will also simultaneously decrease the membrane

    permeability because of the increase of the

    charged form. In contrast, incorporation of lipo-

    philic moieties to enhance the corneal perme-

    ability led to a decline in the aqueous solubility.

    Moreover, the excess lipophilicity would reduce

    the unbound fraction in the target tissues and the

    pharmacological activities. It is desirable that the

    molecules for eye drops should be designed to have

    high aqueous solubility without a loss of lipophi-

    licity for absorption. These examples of molecular

    design are summarized in Table 19.

    An example of the molecular design is an

    incorporation of heterocycles including more than

    two heteroatoms, which generally show high

    solubility for both aqueous solution and organic

    solvent, and can be considered to be amphipathic

    molecules (amphiphiles). Introduction of hetero-

    cycles led to the ophthalmic drugs, such as

    dorzolamide, timolol and brimonidine, having

    excellent ocular pharmacokinetic properties.

    Another approach is to introduce a nonionic

    amphiphile like an oligoethylene glycol methyl

    ether chain, which is able to enhance aqueous

    solubility without a large loss of lipophilicity. In

    the case of oral absorption, incorporation of such

    functionality produced the highly oral bioavail-

    able compounds.157,158 Introduction of a basic

    moiety like an amino or pyridine moiety as anionizable center is also useful for increasing

    aqueous solubility. In this case, one problem is

    the pKavalue of the basic groups. The compoundsthat extensively ionize around neutral pH may

    decrease the corneal permeability. Since ampho-

    lytes like fluoroquinolones and dorzolamide would

    exist as zwitterions, which is an apparent non-

    ionic species, it is expected that the compounds

    will show high aqueous solubility and the corneal

    permeability to some extent.

    Table 17. Partition Coefficient and Corneal Permeability of Quinidine and its Val and ValVal Derivatives156

    Compound log Pa

    Papp (106 cm/s)b

    None Verapamilc GlySard

    Quinidine

    RH 19 71

    ValQuinidine

    RVal 3.52 31 35 18

    ValValQuinidine

    RValVal 4.62 52 50 35

    aPartition coefficient.bCorneal permeability.cCorneal permeability with verapamil (P-gp inhibitor).dCorneal permeability with Glycylsarcosine (oligopeptide transporter substrate).

    Table 18. Summary of Strategies for Improvement of

    Ocular Penetration

    Structure modification (nonprodrug) approaches

    Optimization of lipophilicity

    Enhancement of aqueous solubility

    Reduction of hydrogen bond donors

    Reduction of molecular weight

    Removal of highly reactive moieties

    Decline in melting point

    Prodrug approaches

    Optimization of lipophilicity

    Enhancement of aqueous solubility

    Reduction of hydrogen bond donors

    Enhancement of affinity for uptake transporters

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2485

  • 8/11/2019 Penetracion Ocular

    25/36

    In addition to lipophilicity, hydrogen-bond

    ability also involves membrane permeability. A

    decline in the number of hydrogen bond donors

    can enhance the membrane permeability.

    Removal of hydrogen bond donor is more effective

    for improvement of membrane permeability than

    introduction of additional hydrocarbon.159 The

    corneal permeability of the 15-keto-17-Ph PGF2a-

    IE is twofold higher than 17-Ph PGF2a-IE (the

    corresponding 15-alcohol analog; Tab. 9).108

    Removal of the 15-alcohol group of PGF2a also

    provided about 10-fold increase in corneal perme-

    ability.102 In the case ofb-blockers, the drugs with

    hydrogen bond donors in the substituents showed

    lower permeability than the drug without hydro-

    gen bond donors (Tab. 4).79 Additionally, it has

    been reported that P-gp, an efflux transporter,

    recognize hydrogen bond donors. An increase in

    Table 19. Examples of Structure Modification (Nonprodrug) Approaches for Improvement of Ocular Penetration

    Strategy Approach and examples

    Lipophilicity and aqueous solubility Incorporate heterocycles including more than two heteroatoms

    Preferable logD7.4 is about 23

    Incorporate nonionic amphiphiles

    Incorporate amino groups

    Avoid strong ionizable centers

    Hydrogen bonding Reduce hydrogen-bond donors

    Molecular weight Reduce molecular weight MW

  • 8/11/2019 Penetracion Ocular

    26/36

    number of hydrogen bond donors in molecules

    may raise the possibility of substrate recognition

    by efflux transporters including P-gp. Therefore,

    to enhance the corneal permeability, reduction of

    number of hydrogen bond donor is important.

    Small molecular weight tends to increase both

    aqueous solubility and membrane permeability.For instance, CNS drugs on the market, which

    need to pass BBB that is a very tight barrier, have

    smaller molecular weight than nonCNS drugs.160

    In ophthalmology, the P3 truncated calpain

    inhibitors can pass the corneal membrane more

    easily than calpain inhibitors having the P3

    moiety (Tab. 16).154 Small inhibitors will possess

    both high aqueous solubility and corneal perme-

    ability, possibly leading to a high ocular bioavail-

    ability.

    Reactive functionalities often limit the corneal

    permeability. Of course, the functionalities thatform irreversible covalent adducts should be

    avoided. Functionalities like aldehydes, which

    can react with amino or thiol groups in biomole-

    cules and produce the reversible covalent adduct,

    and possibly reduce the corneal permeability.154

    Replacement of an aldehyde moiety with a

    hemiacetal moiety, which is a masked aldehyde,

    increases the corneal permeability possibly due to

    decline of the electrophilicity in the molecule

    (Tab. 16).150,154 The lower reactivity would

    diminish the reaction between the compound

    and nucleophiles in membrane substances. Both

    replacement of the reactive moieties with less or

    nonreactive ones, and designing prodrugs with

    their reactive sites masked are favorable for

    enhancement of the corneal permeability.

    In the case of suspension formulation, not only

    aqueous solubility but also the dissolution rate

    will affect ocular penetration. The rapid dissolu-

    tion rate of the compound is preferable to

    suspension formulations. A tentative index to

    predict the dissolution rate is the meting point.

    The thiourea calpain inhibitor with lower melting

    point (MP 628C) demonstrated superior ocular

    penetration to the sulfonamide derivative (MP1628C).150

    Prodrug Approach

    Prodrug design is a useful method to improve

    physicochemical and pharmacokinetic properties.

    Prodrugs are drugs with attached functionalities

    in order to obtain favorable structural natures and

    will regenerate their active parent form by

    enzymatic or chemical reactions.161 In ophthal-

    mology, this approach is mainly used to improve

    the corneal permeability and aqueous solubi-

    lity.17,162,163 Additionally, application for the

    site-specific chemical delivery system (CDS) for

    iris-ciliary targeting has also been investigated.20

    These prodrug approaches are capable of signifi-cantly improving the physicochemical properties

    without a decreasing pharmacological activity.

    However, the functional groups that can connect

    the pro-moiety are limited to several groups, such

    as hydroxyl, carboxyl and so on. These examples of

    this approach are summarized in Table 20.

    Prodrug derivatization is very effective for

    enhancing not only the lipophilicity but also the

    corneal permeability in circumstances where

    incorporation of polar functionalities like a

    carboxylic acid and an alcohol moiety lowered

    the corneal permeability of the molecules. Thecompounds including carboxylic functionalities

    generally showed low corneal permeability due to

    their ionization in tears at a neutral pH. Ester

    derivatization of carboxylic acids can increase

    lipophilicity and lead to the significantly improved

    corneal permeability. Since the cornea has high

    esterase activity, eater prodrugs can easily

    regenerate the parent drugs.17 The major pro-

    blems of ester prodrug derivatization are that

    ester prodrugs would show decreased aqueous

    solubility and increased susceptibility for hydro-

    lysis. Considering their stability in an aqueous

    solution, the bulky esters are desirable for pro-

    moieties. For instance, all marketed ophthalmic

    PGF2a prodrugs are bulky isopropyl

    esters.104,107,108 However, more bulky tert-butylesters are inadequate for pro-moieties because

    they resist enzymatic hydrolysis.164,165 Conver-

    sion of carboxylic acids into amides is also effective

    for improving the corneal permeability, but its

    effect is less than that of esters (Tab. 9). Although

    the nonsubstituted amides are partly hydrolyzed

    by ocular tissues (Tab. 13),140 the reactions of

    mono-substituted amides are generally slower

    and di-substituted amides are not substantiallyhydrolyzed.109 Thus, mono- and di-substituted

    amides are not suitable for a pro-moiety.

    The presence of hydrogen bond donors like an

    alcohol or a phenol group causes low corneal

    permeability. The conversion of them into fatty

    acid esters increases the corneal permeability

    (Tabs. 5, 8, 11, 12, 14 and 15). Taking into

    consideration their stability in an aqueous solu-

    tion, pivaloyl esters are favorable as pro-moiety.

    The dipivaloyl ester of epinephrine is marketed as

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2487

  • 8/11/2019 Penetracion Ocular

    27/36

    the ophthalmic solution for the treatment of

    glaucoma.62 Ester prodrug derivatization is also

    used in steroid drugs. In the case of compounds

    having multiple alcohol functionalities such as

    prostaglandins, each ester prodrugs having pro-

    moiety at various positions should be compared,

    because the impact on corneal permeability

    depends on the position of esterification. The

    permeability of multiple esters of a compound

    with more than two alcohol groups is not alwayshigher than that of monoesters (Tab. 8).

    Replacement of alcohol moiety by ketone leads

    to an increase in corneal permeability through a

    decline in the number of the hydrogen bond

    donors. Since ketones are most likely to regener-

    ate alcohols by ketone reductase in the corneal

    epithelium and the iris-ciliary body,20 ketones can

    often be a pro-moiety. Furthermore, conversion of

    ketones into oximes or methyloximes provides the

    prodrug with increased stability in an aqueous

    solution and sustained release of the parent

    compound, because the oximes and methoximes

    are generally more chemically and enzymatically

    stable than the corresponding ketones. Since the

    oximes and methoximes are hydrolyzed to the

    corresponding ketones by enzymes that exist in

    the iris-ciliary body, these can be considered as

    site-specific enzyme activated delivery systems.

    The several oximes or methoximes analogs ofb-

    blocking agents showed a higher and moresustainable IOP reducing effect than the parent

    compounds (Tab. 6).20 These compounds did not

    induce the transient bradycardia, a major side

    effect ofb-blockers, due to their nonactivation in

    plasma.

    On the other hand, the introduction of a strong

    ionic moiety like a phosphate as pro-moiety easily

    enhances aqueous solubility. Phosphate and m-sulfobenzoate ester prodrug approaches are use-

    ful to improve the aqueous solubility. Although

    Table 20. Examples of Prodrug Approaches for Improvement of Ocular Penetration

    Strategy Approach and examples

    Lipophilicity Convert carboxylic acid into ester

    Convert carboxylic acid into amide (slow hydrolysis)

    Convert alcohol into ester fatty acid esters

    Aqueous solubility Convert alcohol to phosphoric or m-sulfobenzoic acid ester

    Hydrogen bonding Convert alcohol into ketone

    Transporter Convert alcohol to a-amino acid or di-peptidyl esters (transporter-mediated)

    Others Convert alcohol into oxime or methoximes via ketone (slow hydrolysis)

    JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 200 8 DOI 10.1002/jps

    2488 SHIRASAKI

  • 8/11/2019 Penetracion Ocular

    28/36

    such strong ionic moieties significantly increase

    the aqueous solubility, they decrease corneal

    permeability. Therefore, these approaches may

    be beneficial for lipophilic parent drugs like

    steroids (Tabs. 14 and 15). In this case, the

    increased solubility may decrease the corneal

    permeability. The phosphate prodrug of predni-solone shows approximately equal ocular pene-

    tration after instillation to the corresponding

    acetate ester, even though the phosphate had a

    320-fold lower corneal permeability than the

    acetate.

    Conversion of a hydroxyl group into a specific a-

    amino acid ester or a dipeptide ester with a

    specific sequence results in enhanced corneal

    permeability most likely due to transportation by

    amino acid transporters (such as LAT 1 and 2) or

    oligopeptide transporters (such as PepT1) in

    corneal epithelium. Incorporation of a free aminogroup also contributes to the increased ocular

    penetration through an increase in aqueous

    solubility. The compounds with (1) a specific a-

    amino acid ester or (2) dipeptide esters with a

    specific sequence recognized as a substrate by

    uptake transporters, both showed higher ocular

    penetration than that expected by their lipophi-

    licity (Tabs. 11 and 12). This is a very effective

    approach for enhancement of ocular penetration,

    but the use will be limited by the parent compound

    structure, its molecular weight, and physicochem-

    ical properties.

    So far, ester and amide prodrugs have been

    reported as marketed eye drops. Since the linkage

    between pro-moiety and the parent drug is most

    likely to be chemically labile, the stability of the

    formulation is often problematic.

    CONCLUSION

    As the many previous reports mentioned, a

    balance between lipophilicity and hydrophilicity

    is the most important factor for ocular penetra-

    tion. The compounds for ophthalmic solution arerequired a higher aqueous solubility than oral

    drugs. The amphipathic structure incorporating

    heterocycles or nonionic amphiphiles would be

    favorable for the enhancement of ocular penetra-

    tion due to addition of an appropriate lipophilicity

    and hydrophilicity. Prodrug approaches for car-

    boxylic acids and alcohols are also useful for the

    enhancement in corneal permeability. In this

    case, we should pay attention to the decline in

    aqueous solubility and stability in an aqueous

    solution. In the case of formulating the compounds

    as an aqueous suspension, a compound having a

    lower melting point is preferable. P-gp substrates

    should be avoided to enhance corneal permeabil-

    ity. On the other hand, the substrates of uptake

    transporters may demonstrate high permeability.

    Thus, in an ophthalmic drug, the molecular designconsidering pharmacokinetic properties can pro-

    vide more effective and less adverse drugs.

    Moreover, such physicochemical property-based

    drug design may be able to provide ocular drugs

    that can reach the posterior segment of the eye via

    topical instillation, in addition to the anterior

    segment.

    FUTURE DIRECTIONS

    Currently, the conformation and pharmacophoreof many transporters and metabolic enzymes are

    not fully understood. In future studies, the

    understanding of crystal structure and the site

    of binding for compounds will be developed and

    will result in the capability of designing molecules

    with good pharmacokinetic properties by in silicoscreening. I expect that further development in

    the field of ocular pharmacokinetics and an

    understanding of transporters and metabolic

    enzymes will produce excellent drugs in the near

    feature.

    REFERENCES

    1. Kempf DJ, Sham HL, Marsh KC, Flentge

    CA, Betebenner D, Green BE, McDonald E,

    Vasavanonda S, Saldivar A, Wideburg NE, Kati

    WM, Ruiz L, Zhao C, Fino L, Patterson J, Molla A,

    Plattner JJ, Norbeck DW. 1998. Discovery of rito-

    navir, a potent inhibitor of HIV protease with high

    oral bioavailability and clinical efficacy. J Med

    Chem 41:602617.

    2. Arrowsmith JE, Campbell SF, Cross PE, Stubbs

    JK, Burges RA, Gardiner DG, Blackburn KJ. 1986.Long-acting dihydropyridine calcium antagonists.

    1.2-Alkoxymethyl derivatives incorporating basic

    substituents. J Med Chem 29:16961702.

    3. Smith DA, Jones BC, Walker DK. 1996. Design of

    drugs involving the concepts and theories of drug

    metabolism and pharmacokinetics. Med Res Rev

    16:243266.

    4. Lin J, Sahakian DC, de Morais SM, Xu JJ, Polzer

    RJ, Winter SM. 2003. The role of absorption, dis-

    tribution, metabolism, excretion and toxicity in

    drug discovery. Curr Top Med Chem 3:11251154.

    DOI 10.1002/jps JOURNAL OF PHARMACEUTICAL SCIENCES, VOL. 97, NO. 7, JULY 2008

    DESIGN FOR ENHANCEMENT OF OCULAR PENETRATION 2489

  • 8/11/2019 Penetracion Ocular

    29/36

    5. Balani SK, Miwa GT, Gan LS, Wu JT, Lee FW.

    2005. Strategy of utilizing in vitro and in vivo

    ADME tools for lead optimization and drug candi-

    date selection. Curr Top Med Chem 5:1033

    1038.

    6. Clark RD, Wolohan PR. 2003. Molecular design

    and bioavailability. Curr Top Med Chem 3:12691288.

    7. Schwardt O, Kolb H, Ernst B. 2003. Drug discov-

    ery today. Curr Top Med Chem 3:19.

    8. Thompson TN. 2001. Optimization of metabolic

    stability as a goal of modern drug design. Med

    Res Rev 21:412449.

    9. Kumar GN, Surapaneni S. 2001. Role of drug

    metabolism in drug discovery and development.

    Med Res Rev 21:397411.

    10. Lipinski CA, Lombardo F, Dominy BW, Feenesey

    PJ. 2001. Experimental and computational

    approaches to estimate solubility and permeability

    in drug discovery and development settings. Adv

    Drug Delivery Rev 46:326.11. Gombar VK,Silver IS, Zhao Z. 2003. Role of ADME

    characteristics in drug discovery and their in silico

    evaluation: In silico screening of chemicals for

    their metabolic stability. Curr Top Med Chem

    3:12051225.

    12. Darvas F, Keseru G, Papp A, Dorman G, Urge L,

    Krajcsi P. 2002. In Silico and Ex silico ADME

    approaches for drug discovery. Curr Top Med

    Chem 2:12871304.

    13. van De Waterbeemd H, Smith DA, Beaumont K,

    Walker DK. 2001. Property-based design: Optimi-

    zation of drug absorption and pharmacokinetics.

    J Med Chem 44:13131333.14. Kola I, Landis J. 2004. Can the pharmaceutical

    industry reduce attrition rates? Nat Rev Drug

    Discov 3:711714.

    15. Blake JF. 2005. Identification and evaluation of

    molecular properties related to preclinical optimi-

    zation and clinical fate. Med Chem 1:649655.

    16. Majumdar S, Mitra AK. 2006. Chemical modifica-

    tion and formulation approaches to elevated drug

    transport across cell membranes. Expert Opin

    Drug Deliv 3:511527.

    17. Lee VHL, Li VHK. 1989. Prodrugs for improved

    ocular drug delivery. Adv Drug Deliv Rev 3:138.

    18. Sugrue MF. 1996 The preclinical pharmacology of

    dorzolamide hydrochloride, a topical carbonicanhydrase inhibitor. J Ocul Pharmacol Ther

    12:363376.

    19. Resul B, Stjernschantz J, Selen G, Bito L. 1997.

    Structure-activity relationships and receptor pro-

    files of some ocular hypotensive prostanoids. Surv

    Ophthalmol 41:S47S52.

    20. Bodor N, Buchwald P. 2005. Ophthalmic drug

    design based on the metabolic activity of the

    eye: Soft drugs and chemical delivery systems.

    AAPS J 7:E820E833.

    21. Mannermaa E, Vellonen KS, Urtti A. 2006. Drug

    transport i