throbber
Current M'('a'r'('r'mt1'Clwiirfslry, 2009, 16, 4481-4489
`
`4481
`
`The Design and Development of Fesoterodine as a Prodrug of 5-
`Hydroxymethyl Tolterodine (5-HMT), the Active Metabolite of Tolterodine
`
`B. Malhotra*‘], K. Gandelrnan], R. Sachsez, N. Wood3, M.C. Miehel4
`
`iffizer Inc’, New Yorlr, NY, USA.‘ 2SCHl/VARZ Bi'0Sei'eriCes, M0m’1ei'm, Ger'mcm_v,' 3Pfizer Global Re.rear'c’l1 and
`Development, S(II’1i$lWlCl’t, Kent, UK,‘ 4Urilversl{v of/lmsterdam, Amsterdam, The Netherlands
`
`Abstract: This review highlights the design and development of fesoterodine (Toviazo) as a prodrug of5—hydi'oxymetliy1
`tolterodine (5-11MT). which is also the active metabolite oftolterodine. for the treatment of overactive bladder (OAB).
`Tolterodine and 5-HMT are both potent antimusearinic agents. A prodrug approach was necessary for systemic bioavail-
`ability of 5—HMT after oral administration. Fesoterodine was selected amongst a series of ester analogues of 5—HMT to
`develop an advanced OAB treatment with an optimum hiopharmaeeutics profile. while maintaining a pharmacological
`link to tolterodinc.
`
`the logD value. a determinaiit of lipophilieity and
`While tolterodinc and 5-l-{MT have similar antimusearinie activity.
`permeability across biological interfaces such as the gut wall and blood-brain barrier. is eonsidembly lower for 5-llMT
`(0.74) versus tolterodinc (1.83). In contrast to the cytochrome P450 (CYP) 2D6-mediated metabolism of tolterodinc. 5-
`HMT formation from fesoterodine occurs via ubiquitous nonspecific esterases. Consequently, treatment with fesoterodine
`results in consistent. genotype-independent exposure to a singular active moiety (5-HMT): treatment with tolterodinc re-
`sults in CYPZDO genotype-dependent exposure to varying proportions of two active moieties (5-HMT and tolterodinc). At
`least partially due to the avoidance ofvariations in pliarmaeokinetie exposures observed with tolterodinc. it was possible
`to develop fesoterodine with the flexibility oftwo efficacious and well-tolerated dosage regimens of4 and 8 mg daily.
`
`Keywords: Fesoterodine. prodrug. 5-HMT. tolterodinc. metabolite. lipophilicity. CNS.
`
`INTRODUCTION
`
`Overactive bladder (OAB) is defined by the lnternational
`Continence Society as urgency, with or without urgency in-
`continence. usually accompanied by increased daytime fre-
`quency and nocturia [l]. The etiology of OAB remains un-
`clear, although it
`is generally accepted that OAB is often
`associated with detrusor overactivity. which may result from
`inappropriate levels of cholinergic activation of muscarinic
`receptors on the detrusor muscle or afferent nerves [2]. An-
`timuscarinic agents are the mainstay of pharmacologic
`treatment for OAB;
`their therapeutic effect
`is presumably
`mediated via blockade of muscarinic receptors on detrusor
`smooth muscle cells. urothelium, and/or sensory afferents,
`thereby inhibiting detrusor contractions [3, 4].
`
`tolterodinc, and
`solifcnacin,
`Darifenacin, oxybutynin,
`trospium are among the antimuscarinic drugs approved for
`the treatment of urgency urinary incontinence (UUI) and
`other symptoms related to OAB. The effectiveness of oxybu-
`tynin has been demonstrated in several clinical studies. but
`the clinical usefulness of oxybutynin is limited due to an-
`timuscarinic side effects. Dryness of the mouth is the most
`common experienced side effect which may be severe
`enough to result in poor compliance or discontinuation of
`treatment [5-7]. Tolterodine. a potent and competitive an-
`timuscarinic agent. was the first drug specifically developed
`for OAB treatment. Preclinical pharmacological data show
`that tolterodinc exhibits more potent antimusearinic receptor
`activity in vivo in the urinary bladder over the effect on the
`salivation, whereas oxybutynin exhibits the reverse selectiv-
`ity [8]. The favorable bladder selectivity of tolterodinc dem-
`onstrated in preclinical studies has been confirmed in clinical
`
`’’‘Address correspondence to this author at the Clinical Sciences. Pfizer Inc.
`(:85 3rd Avenue. New York. NY 10017. USA: Tel: 1-212-733-4723:
`Fax: 1-212-441-4490: E-mail: himal.k.ma1hotraaflpfizer.com
`
`studies [6]. Tolterodine being a tertiary amine, combined
`with other physicochemical attributes such as low lipophilie-
`ity and positive charge. has relatively low central nervous
`system (CNS) penetration in animal models and a low inci-
`dence of CNS adverse events in clinical trials [9-l 1]. Thus
`good clinical efficacy combined with low incidences of dry
`mouth and other antimuscarinic side effects makes tol-
`
`terodinc a treatment ofehoiee for OAB [12, 13]. However.
`clinicians and patients are not completely satisfied with OAB
`therapy despite the availability of tolterodinc and the newer
`antimuscarinic agents, solifenacin and darifenacin. because
`of a suboptimal efficacy/tolcrability balance or the inability
`to achieve optimal efficacy in difficult-to-treat patients.
`The clinical effects of tolterodinc are derived from the
`
`combined exposure to two active moieties, the parent drug
`(tolterodinc)
`and
`its
`equipotent
`active metabolite
`(5-
`hydroxymethyl tolterodinc; 5-HMT). which is formed via a
`major pathway involving the cytochrome P450 (CYP) 2D6
`enzyme [14]. Since CYPZD6 is genetically polymorphic
`there is considerable variability among patients in the pro-
`portion of these moieties, with higher exposures to tol-
`terodinc in CYPZD6 poor metabolizers (PMs) compared
`with extensive metabolizers (EMs) [l4.
`l5].
`ln a multiple-
`dose study of tolterodinc, the tolterodinc area under the con-
`centration-timc curve (AUC) values varied over a 130-fold
`range across CYPZD6 EMs and PMs whereas those for 5-
`HMT varied over a 5-fold range in EMs (5-HMT was not
`formed in PMs) [15]. Likely dLIe to the high variability of
`tolterodinc exposures.
`the development of tolterodinc was
`limited to only a single dosage of4 mg daily, although the
`dose may be lowered to 2 mg daily based on individual re-
`sponse and tolerability.
`
`involving
`Delivery of 5-HMT as a new drug without
`CYPZD6-mediated metabolism of tolterodinc has advantages
`compared to tolterodinc because only one active principle
`would be handled by the patients with less variability. which
`
`U929-8673/09 $55.0[l+.ll[l
`
`(0 2009 Beiitliam Science Publisliers Ltd.
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0001
`
`

`
`4482 Current M'('a'r'cr'naI CII('nn'.I‘(ry, 2009 V01. I6, N0. 33
`
`M'aHm.rra at :11.
`
`should result in a lower variation in efficacy and fewer side
`effects. The introduction of an additional hydroxyl group in
`the toltcrodine molecule, however, results in reduced lipo-
`philicity, which could produce a side effect profile with CNS
`penetration even lower than that of toltcrodine but would
`also result
`in lower absorptionfbioavailability.
`In order to
`overcome the absorption disadvantage, different prodrugs of
`5-IIMT have been synthesized and tested for their antimus-
`carinic activity, potential
`absorption through biological
`membranes. and enzymatic cleavage.
`
`Fesoterodine (Toviaza) is the isobutyric acid ester of 5-
`11MT which, based on its optimum biopharmaceutical attrib-
`utes, was selected amongst a series of carboxylic acid ester
`analogs for development as a treatment for OAB at dosages
`of 4-mg and 8-mg daily. Fesoterodine is rapidly and exten-
`sively converted to 5-HMT, such that
`the pharmacologic
`activity appears to be primarily attributable to 5-HMT [16,
`17]. The physieoehemieal, pharmacologic, and metabolic
`properties of fesoterodine,
`in relation to toltcrodine, and the
`rationale for the development of fesoterodine are the subject
`of this review.
`
`1. Antimuscarinic Pharmacology of Tolterodine and 5-
`HMT
`
`The antimuscarinic activities of fesoterodine, tolterodine
`and 5-IIMT have been evaluated in membrane preparations
`of Chinese hamster ovary cells expressing the five mus-
`carinic receptor subtypes (M1—M5);
`the functional activities
`were determined in organ-bath studies using the rat bladder
`in vifm and in eystometry studies in rats in vivo [18]. These
`studies demonstrated that 5-HMT is the main active principle
`of fesoterodine, which acts as a prodrug, and that both to]-
`tcrodine and 5-HMT are specific muscarinic receptor an-
`tagonists at the M1 through M5 subtypes [18]. The potencies
`of various antimuscarinic agents, expressed
`Ki values for
`each muscarinic receptor subtype, are summarized in Table 1
`[19—24]. Across different studies reported in the literature,
`the absolute values of the in vitm affinity estimates vary
`somewhat, however,
`the relative affinities for the receptor
`subtypes are consistent for each antimuscarinic drug. Feso—
`terodine, being a prodrug, has very low but similar affinity
`across the five musearinie receptor subtypes.
`
`2. Biopharmaceutics and Preclinical Pharmacologic As-
`sessment: Selection of Fesoterodine
`
`A comprehensive evaluation of the absorption, distribu-
`tion, metabolism, and excretion (ADME) properties in pre-
`
`clinical models and in Phase 1 clinical pharmacology studies
`forms the biopharmaceutics-related determination of the de-
`velopment potential of therapeutic drug molecules. An inte-
`gration of the knowledge of the biopharmaceutics and pre-
`clinical pharmacological properties of a drug along with
`anatomic distribution of receptors can become useful
`in a
`qualitative prediction of the beneficial and adverse effects of
`the drug.
`
`Lipophilkity and CNS Penetration af5-HMT
`
`Muscarinie receptors are not only located in the target
`organ responsible for efficacy (bladder) but also in other
`tissues and organs (gastrointestinal
`tract, salivary glands,
`heart, and brain) that can affect the tolerability and safety of
`an antimuscarinic drug. For example, antimuscarinic drugs
`with greater M3 receptor occupation may cause greater inci-
`dence of dry mouth and constipation [25]. Relevant to the
`antimuscarinic pharmacology in the CNS, physicochemical
`properties such as lipophilicity and permeability are impor-
`tant predictors of the ability of a molecule to penetrate the
`blood-brain barrier (BBB), such that those with higher lipo-
`philicity at physiologic pH are more likely to exert pharma-
`cologic adverse effects (AEs) in the CNS. The logD values at
`the physiologic pH 7.4, determined from the oetanolzwater
`distribution coefficient [26], are commonly used to compare
`the lipophilicity of drugs as an indicator of their ability to
`cross the BBB and their propensity to cause CNS AEs. A
`significant and positive correlation between logD and CNS
`AEs has been demonstrated across the class of triptan drugs
`for the treatment of migraines [27].
`
`The logD for 5-HMT [14], toltcrodine [14], and other an-
`timuscarinic agents [14, 28-30] is shown in Table 2. 5-H MT
`has demonstrated low lipophilicity at acidic pH and higher
`lipophilicity at more basic pH [31], as expected for a com-
`pound with a pKa value of 9.28. The logD values of tol-
`tcrodine and 5-HMT have been reported to be 1.83 and 0.74,
`respectively (Table 2) [14]. In addition,
`the lipophilicity of
`several antimuscarinic agents used for treatment of OAB has
`been reported previously [14, 29, 32, 33]. Tolterodine has
`low lipophilicity and low CNS penetration (brain/blood ratio,
`0.1-0.3 for radioactivity) in mice [14] with no significant
`CNS AEs observed in patients [34]. LogD values for solifen-
`acin and oxybutynin are reported to be 1.69 and >3.3, re-
`spectively, suggesting that both of these compounds are
`more lipophilic than 5-HMT (Table 2) [24, 35]. Trospium
`chloride is a quaternary ammonium compound and, as such,
`would not cross the BBB because of its low lipophilicity
`(Table 2) [32]. The small logD for 5-HMT suggests that it is
`
`Table 1.
`
`Affinity of Various Antimusearinic Drugs for Human Muscarinie Receptor Subtypes
`
`Muscarinic
`Receptor
`
`Tolterodine
`|2 1, 22, 24]
`3.0, 3.0, 2.7
`3.8, 6.4, 4.2
`3.4. 12, 4.4
`5.0, 1.9, 6.6
`3.4, 4.6, 2.5
`
` ‘).U, 8.2
`
`5-HMT
`|2l , 22]
`
`Fesoterodine
`|22]
`
`Ki. nM
`
`Trospium
`|23]
`
`Oxybutynin
`|22, 24]
`
`Solifenacin
`|24]
`
`Darifenacin
`| 1'9, 24]
`35,3]
`56, 100
`1.2, 2.0
`13,52
`
`ND: Not f)ele1‘min'.1b|e ("S1J‘?=I. bintling at
`
`I
`
`|.IM),
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0002
`
`

`
`Dm'i'gr: and Dvvdripriwrrf 0f'F:*.\'riterridirrc’
`
`C'urrw1t ."Wt’df£'fl!fl[ Clrwuiiflry, 2009 Vol. M. Na. 33
`
`4483
`
`Table 2. Lipophilieity of Various Antimusearinie Agents for Overactive Bladder
`
`Antimusearinie
`Agent
`Lngl)
`
`Truspium -I .22
`|28]
`
`5-HMT 0.74
`H4]
`
`Tulterorline L83
`H4]
`
`Lil 6
`
`@,.®
`
`O
`U 0 OH
`
`0
`
`Antimuscarinic
`Agent
`L0gD
`
`Solifenacin 1.69
`
`Darifenacin 2.7
`I30]
`
`() xyhutynin >-3.3
`I14]
`
`unlikely to cross the BBB and cause CNS effects. The low
`CNS penetration potential of 5-HMT is of particular clinical
`relevance in elderly patients. who may be taking a number of
`concomitant medications and are vulnerable to CNS Alis.
`
`A study comparing changes in quantitative-topographical
`clcctrocncephalogram (qEEG) activity following administra-
`tion of antimuscarinic agents demonstrated that oxybutynin.
`a tertiary amine, caused significant changes in four fre-
`quency bands compared with placebo and produced the
`highest number of CNS-related effects [34]. Neither
`tol-
`terodine. also a tertiary amine. or trospium chloride. a quar-
`ternary amine, produced significant changes in qEE(} activ-
`ity compared with placebo.
`
`The brain/plasma ratios of fesoterodine-related radioac-
`tivity have been reported to be approximately 0.04 and 0.07
`for maximal drug concentration (C.,,.‘,_..) and AUC, respec-
`tively. after single oral doses of MC-fesoterodine given to
`mice (data on file. Pfizer Inc). Similarly. permeability studies
`in a porcine model demonstrated that the BBB permeability
`eoeflieient of 5-H MT is 6.5x l()‘° em/s compared with per-
`meability coefficients of 23><l0'° cm/s
`for oxybutynin,
`solifenacin, and darifenacin (data on file: Pfizer Inc). Lower
`brain/plasma ratio and permeability with fesoterodine com-
`pared with tolterodine suggest an even lower risk of CNS
`AEs with fesoterodine than with tolterodine.
`In Phase 3
`
`following once-daily ad-
`trials with fesoterodine,
`clinical
`ministration of fesoterodine 4 or 8 mg or tolterodine 4 mg,
`there were no CNS AEs with an incidence rate greater than
`2% or exceeding that following placebo administration [9,
`ll), 36, 37]. Similarly,
`in a Phase 3 study of tolterodine, the
`incidence of CNS AEs ofsomnolenee and dizziness was low
`
`(3% and 2%, respectively) but slightly higher than that for
`placebo (2% and 1%, respectively) [3 8].
`
`Permeability 0f5-H1141} Improving Absorbubility by Pro-
`drug Apprmtrch
`
`While the lower lipophilicity of 5-HMT favors its CNS
`tolerability through lower CNS penetration. it also affects the
`ability of 5-IIMT to permeate other biological barriers, such
`as gut wall or skin for oral or transdermal delivery, respec-
`tively. Therefore. a prodrug approach was considered to
`structurally modify 5-llMT for enhanced permeability across
`human skin to facilitate the delivery of 5-HMT into the sys-
`temic circulation. Ester derivatives were considered as the
`
`choice for developing the prodrug of 5-HMT (Table 3), be-
`cause esterases are known to be ubiquitous and are not
`known to be subject to genetic polymorphism or drug inter-
`actions of clinical significance [39-41]. As a result of rapid
`and eflieient hydrolysis of an ester prodrug immediately af-
`ter permeability aeross the gut wall or skin. only 5-HMT
`would be available in the systemic circulation.
`
`To investigate the feasibility of nonsystemic delivery, the
`biological membrane permeability of 5-HMT along with its
`various ester analogues was assessed using flux rates across
`human skin in the flow-through cell model
`in vitro [42].
`Substitution of both the hydroxy groups of 5-l-IMT leads to
`approximately a 20-fold increase in skin permeation in rela-
`tion to 5-HMT. Surprisingly, monosubstitution of the pheno-
`lic hydroxy group resulted in even higher. about 50-fold in-
`creased, penetration rate through human skin. The in virr-0
`metabolism ofdifferent ester prodrugs to generate the active
`metabolite. 5-IIMT, by enzymatic process was evaluated in
`pooled human liver S9-preparation. The formation of the
`active metabolite depended on the substituents both at the
`benzylic and phenolic side of the respective compounds,
`with turnover rates ranging from 96% to 63%. Similar to
`permeability. monosubstitution of the phenolic hydroxy
`group resulted in highest
`turnover to form 5-IIMT [42].
`Overall, based on its optimized permeability (Table 3) and
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0003
`
`

`
`4484 Current M'('a'r't'r'r:a1' CII('nn'.I‘(ry, 2009 V01. I6, N0. 33
`
`M'aHm.rra at al.
`
`Table 3. In Vitm Permeability of Various Structural Analogs of 5-HMT [42|
`
`(RHO 6
`
`O-R2
`HY
`\“\/N\‘/
`
`5-HMT Ester Derivatives
`
`Chemical designation (name)
`
`Permeation Rate (pg/em3l24 hours)
`
`
`
`Hydroxy
`
`Hydroxy
`
`lsobutyrate
`
`Propionate
`
`Hydroxy
`
`lsobutyrate
`
`lsobutyrate
`
`Propion-ate
`
`HO-/-OH (5 -hydroxymethyl tolterodine)
`
`HO-/-(')BL1t(fesoterodine)
`
`But()-/-()But
`
`|’rop(J-/-()|’rop
`
`rapid hydrolytic conversion, the isobutyrate monoester (feso-
`terodine) was considered to be the best candidate for bio-
`pharmaceutical development. Furthermore,
`the results ob-
`tained in the receptor binding and tissue assays demonstrated
`that the anticholinergic activity of the compounds decreases
`with increased derivatization, such that the ester derivatives
`were significantly less potent
`than 5-HMT:
`therefore, an
`ester such as fesoterodine. which has optimum permeability
`and efficient hydrolysis, would act effectively as prodrug of
`5-HMT.
`
`3. Elimination Pathways of Tolterodine, Fesoterodine,
`and 5-HMT
`
`it was demonstrated
`Based on pharmacokinetie studies,
`that fesoterodine is rapidly and extensively converted by
`nonspecific, ubiquitous esterases to 5-HMT, such that feso-
`terodine is undetectable in plasma after oral dosing [16, 17].
`Furthermore, compared with 5-HMT, fesoterodine is 2 or
`more orders of magnitude less potent at muscarinie receptors
`[22]. Further biotransformation of 5-HMT results in inactive
`metabolites, making 5-llMT the principal active moiety of
`fesoterodine after oral administration [l6, l7, 22] 5-HMT is
`also an active metabolite of tolterodine. but
`in contrast to
`fesoterodine,
`the metabolism of tolterodine to 5-llMT is
`more complex; it undergoes oxidation via CYPZD6 primarily
`in the liver (Fig. 1) [14]. Both tolterodine and 5-HMT con-
`tribute to the pharmacologic activity observed following
`administration of tolterodine, but the proportion of plasma
`tolterodine/5-H MT varies according to a patient’s CYP2D6
`genotype [15, 43]. Because the CYP2Do pathway is subject
`to genetic polymorphism,
`the efficiency with which indi-
`viduals can metabolize tolterodine to 5-HMT varies across
`
`the population [44]. Overall, CYP2D6 metabolizer status can
`be broken down into EMs, intermediate metabolizers (IMs),
`or PMs [44]. Approximately 7—l0"o of the Caucasian popu-
`lation are devoid of CYP2D6 (ie, PMs) and are unable to
`metabolize tolterodine efficiently and do not form 5-HMT.
`The CYP2D6 EMs can efficiently metabolize tolterodine to
`5-HMT, resulting in approximately similar exposures to tol-
`terodine and 5-HMT. IMs are less efficient at metabolizing
`tolterodine, resulting in a smaller proportion of5-HMT. This
`
`genetic variability in metabolic status, as well as variability
`arising from drug interactions with CYP2D6. leads to highly
`variable ratios of tolterodine and 5-llMT in different patients
`(Fig.1) [15, 45, 46]. As highlighted in Fig. (1), because feso-
`terodine functions as a prodrug and due to the involvement
`of esterases in the formation of 5-llMT, all patients are ex-
`posed to a single active moiety when fesoterodine is admin-
`istered. On the contrary. tolterodine itself is also active and
`due to CYPZD6-mediated metabolism of tolterodine to 5-
`
`HMT, depending on patient’s CYP2D6 genotype, varying
`proportions of two different active moieties are involved
`when tolterodine is administered.
`
`Whether formed after tolterodine or fesoterodine admini-
`
`stration, 5-HMT is subsequently metabolized to inactive ear-
`boxy. carboxy-N-desisopropyl. and N-desisopropyl metabo-
`lites, with CYP2D6 and CYP3A4 identified as likely to con-
`tribute
`to
`the
`formation of
`the
`earboxy and
`the N-
`desisopropyl metabolites.
`respectively,
`following incuba-
`tions with heterologously expressed human CYP isoforms
`and CYP chemical
`inhibitors (Fig. 2) [20]. Thus, CYP ac-
`tivity appears to be responsible for the further metabolism of
`5-HMT after administration of fesoterodine or tolterodine.
`
`The apparent oral clearance of 5-HMT is reduced by ap-
`proximately 40% in CYPZD6 PMs compared with EMs. A
`similar reduction in the apparent oral clearance of5-HMT is
`also noted; during concomitant administration of
`feso-
`terodine and the potent CYP3A4 inhibitor ketoconazole [47,
`48]. The observation that the magnitude of the increase in 5-
`llMT exposure was similar when either of the 2 pathways
`was absent/inhibited indicates
`that both CYP2D6 and
`
`CYP3A4 may play equally important roles in the elimination
`of5-llMT [47]. In addition to metabolic inactivation via two
`major pathways involving CYP3A4 and CY P2D6 enzymes,
`5-HMT is also excreted unchanged in the urine. accounting
`tor about 16% of the orally administered dose offesoterodine.
`Because of the availability of multiple metabolic and renal
`pathways for the elimination of 5-HMT. patient
`intrinsic
`(hepatic or renal impairment) or extrinsic (drug interactions
`with potent CYP3A4 or CYPZD6 inhibitors) factors have
`only a modest effect of about 2.5—fold increase in 5-HMT
`exposures [36]. These modest increases in 5-HMT exposures
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0004
`
`

`
`Desigrr and Develapmem 0f'Fe.\'rJterrJdr'r:e
`
`C'ur'reut :'l4'eal'i::'imu' Clrerrtixtry, 2009 Vol. M. Na. 33
`
`4485
`
`Fesoterodine Conversion to 5-HMT Is Sim la and Predictable
`Esterases
`
`Fesoterodine
`
`/ 5-HMT
`Ubiquitous
`
`Tolterodine Metabolism is More Com [ex and Less Predictable
`
`Tolterodine
`
`CYP2 D6
`
`Z
`Liver, gut
`
`Tolt
`
`+
`
`Tolt
`
`"'
`@
`
`Tolt
`
`Extensive
`Melabolizers
`-78%
`
`Interm ediate
`Metabolizers
`-1 5%
`
`Poor
`Metabolizers
`-7%
`
`Fig. (1). l:'ste1‘ase-Mediated vs. CYP2D6-Mediated Forination of5-HMT; Tolt: Tolterodine; 5-HMT: 5-hydroxymethyl tolterodine.
`
`after fesoterodine administration are managed through sim-
`ple dosing recommendations in the product label that require
`fesoterodinc doses not exceeding the initial dose of4 mg/day
`under the aforesaid situations [36]. On the other hand, since
`CYP2D6 is the single predominant pathway involved in the
`elimination of tolterodine. potent inhibition of CYPZD6 ac-
`tivity by iluoxetine results in marked reduction in clearance
`(up to 93%) and increase in exposure (up to 14-fold) of tol-
`terodine. where as 5-HMT was undetectable [49]. Therefore,
`as a result of variations in patient-intrinsic and -extrinsic
`factors, relatively low inter-patient or inter-occasion variabil-
`ity in active moiety exposures is expected following feso-
`terodine administration compared with tolterodine.
`
`Fesolerodlne
`
`The antimusearinic effects of drugs used for OAB treat-
`ment [50] can potentially be attributed to the presence ofMg
`and M3 receptors (with a predominance of M3 receptors) on
`the detrusor muscle and the urothelium [2]. As shown in
`Table 4, some antimusearinic agents have an appreciable
`urinary excretion of unchanged parent drug (trospium [51,
`52] and solifenaein [53]) or an active metabolite (toltcrodine
`[10] and fcsoterodinc [36]), providing exposure to the active
`moiety locally in the bladder. The bladder exposure to the
`antimusearinic active moieties in these cases may have an
`effect on the afferent nerves located near the urothelium
`
`[54], which could impart additional efficacy in reducing
`OAB symptoms.
`
`J\‘¢°
`
`Tolterodine
`
`Tollblndlno-Spetifif mclabnlllt‘ N—dea||<ylated 5—HMT
`
`Nrdealkylaled metabolite
`
`OHY
`Hr
`
`Carboxyrmerabolire
`
`X CYFEA4
`
`Neealkylaled S—CM
`
`Fig. (2). Biotransfiarmation Pzttllwztys of Fesoterodine and Tolterodine: 5-HMT: 5-hydroxymcthyl tolterodinc
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0005
`
`

`
`4486 Current M'('a'r'cr'nat' CIrt'rm'.rrry, 2009 Vol. 16, No. 33
`
`M'aHm.rra at al.
`
`Table 4. Systemic Bioavailability & Urinary Excretion of Various Antimuscarinic Drugs for Overactive Bladder
`
`4 mg
`(once-daily)
`
`toltcrodine
`
`|7"xi. (EM)
`(15% [ PM)
`
`Sanctura®
`|51|
`ttrnspiunl)
`
`20 mg
`(twice-daily)
`
`trnspiurn
`
`Sanctura®
`XR|52|
`ttrnspiunl)
`
`60 mg
`(once-daily)
`
`trnspiurn
`
`Vesica rc®
`|53|
`(snlifcnacin)
`
`10 mg
`(once-daily)
`
`snlifcnacin
`
`Enal)|ex® I5ll|
`(darifcnacin)
`
`Ditropan XL® [35|
`(nxyhutynin)
`
`15 mg
`(once-daily)
`
`darifcnacin
`
`20 mg
`(once-daily)
`
`nx_vhut_vnin
`
`dcscth_vl-
`mryhutynin
`
`60% of urinary
`L‘.\L‘1‘L‘tit)1t
`
`60% of urinary
`
`L‘.\L‘1‘L‘tit)1t
`
`Trade Name
`(generic
`name)
`
`Dose
`(regimen)
`
`Toviaz® I36|
`
`Detrol L/\® [1ll|
`
`(fesoterodine)
`
`(toltcrodine)
`
`8 mg
`(once-daily)
`
`Active Mnicty
`
`5-HMT
`
`Systemic
`Bioavailability
`ol'Active
`Mnicty
`
`Dose Excretecl
`as Active
`Moicty in
`Urine
`
`Active Moiety
`li.\c1'ctcd in
`Urine. mg**
`
`*Based on absolute bioavailability of'Sanctura® and the plasma area under the curve of tmspium following of Sanctura® XR vs. Sanctura®.
`*“L‘o1nputet.i based on highest dose and ‘A. ol't.iose e,~.ereted in urine (dose. hioavailability and ‘A. of urinary excretion l'o1'Sanctu1'a®).
`NR=not reported.
`
`4. Clinical Pharmacokinetic Studies of Fesoterodine
`Versus Tolterodine
`
`From an overall safety, tolerability and efficacy perspec-
`tive, following administration of therapeutic doses ofa drug,
`it becomes desirable to achieve a narrower and more predict-
`able range of systemic concentrations of the drug and/or its
`active mctabolite(s) across patients. This allows the physi-
`eian to use the drug in different patient types with a reason-
`able expectation of the effects in individual patients based on
`the results of controlled clinical trials.
`
`Findings from studies assessing the pharmacokinetics
`(PK) of toltcrodine and fesoterodine suggest
`that
`the PK
`variability observed after dosing with toltcrodine appears to
`be attributable to the toltcrodine molecule rather than 5-
`
`llMT. In an open-label, multiple-dose study, the PK of tol-
`tcrodine extended release (ER) were investigated in healthy
`volunteers [15]. Because CYPZD6 is the predominant elimi-
`nation pathway for toltcrodine and results in the formation of
`5-HMT. both toltcrodine and 5-HMT are active. Therefore
`the sum of both active moieties is relevant to the effects of
`
`toltcrodine. Fig. (3) demonstrates the serum concentration
`versus time profiles of the 2 active moieties separately and
`also combined following oral administration of toltcrodine
`ER, with each plot line representing individual subjects in a
`heterogeneous population of CYPZD6 EMs and PMs.
`In
`patients with normal CYP2D6 activity (EMs). roughly equal
`concentrations of toltcrodine and 5-HMT are seen. However,
`in patients with CYPZD6 activity absent
`(PMs),
`the tol-
`tcrodine concentrations are several-fold higher than those in
`EMs (Panel A) whilst 5-HMT is not quantifiable (Panel B).
`As a consequence, when toltcrodine was administered to a
`group of CYP2D6 EMs and PMs, the active moiety concen-
`trations varied over a wide range (almost 2 orders of magni-
`tude) as apparent in Panel C [15].
`
`When we separate the active moiety concentrations into
`individual contributions from toltcrodine and 5-HMT.
`it
`is
`
`apparent that unchanged toltcrodine is the primary source of
`variability. whereas 5-HMT exposures are maintained within
`a relatively narrow range (Fig. 3). For instance, examination
`of peak exposures indicates that the lowest and highest val-
`ues are 6-fold apart for toltcrodine but only 4-fold apart for
`5-HMT. The C.,,,,,, of toltcrodine varies between 1-60 nM
`[15], whereas the Cum of 5-HMT only varies between 3-13
`nM [55]. This is primarily related to CYP2D6 activity. as
`PMs have the highest toltcrodine concentrations. Since tol-
`tcrodine and 5—HMT are similar antimusearinie agents,
`the
`less variable pharmacokinetics of 5-HMT makes it a more
`desirable therapeutic molecule.
`
`Based on these data, it would be ideal to deliver 5-HMT
`without reliance on toltcrodine or CYP2D6, which is pre-
`cisely what has been accomplished by developing feso-
`terodine as a prodrug of 5-HMT. Fesoterodine itself is inac-
`tive and is converted rapidly and extensively by esterases to
`form 5-HMT,
`the singular active moiety in case of feso-
`terodine. In a separate open-label. single-dose study. the PK
`and dose proportionality investigation of fesoterodine sus-
`tained release (SR) were investigated in healthy volunteers
`[55]. In this study of fesoterodine in CYP2D6 PMs and EMS.
`each subject- whether EM or PM- has exposure to 5-llMT
`because its formation does not depend on CYPZD6. After
`fesoterodine administration. the inherent lower variability of
`5-llMT is maintained
`was seen with toltcrodine, without
`the high variability associated with toltcrodine.
`
`Although these data were obtained from 2 separate stud-
`ies in which toltcrodine or fesoterodine were administered to
`
`a heterogeneous group of EMs and PMs, comparisons of
`active moieties are of interest and valid, given the objective
`nature of PK measurements and the inclusion of subjects
`genotyped as EMs and PMs in both studies [15, 55, 56].
`While the fesoterodine study was single-dose, these data are
`representative of steady state because fesoterodine does not
`accumulate and the PK are time invariant. The individual
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0006
`
`

`
`Desigrr and Drwralrlpnirarn‘ 0fFe.mt‘errJdiur3
`
`Current MediciIraICI:eI:1i.v1ry, 2009 V01. 16. Na. 33
`
`4487
`
`A
`
`100
`
`Toltorodlne ER 4 mg
`
`B
`
`100
`
`Tollerodine ER 4 mg
`
`u‘:
`
`E3:
`
`5 3
`
`oDEE
`
`10
`
`«E
`::
`
`1
`
`10
`
`1
`
`3 E‘
`
`.5’
`
`8 E E
`
`EO
`
`i-
`
`
`
`Tolterodine+5—HMTSerumCone(ng!mL)‘-7
`
`Time (hours)
`
`Time (hours)
`
`Tolterodlne EH 4 mg
`
`_. O0
`
`‘I0
`
`DI
`
`
`
`12
`Tlme (hours)
`
`IE
`
`24
`
`Fig. (3). Serum Concentrations of Active Moictics following Administration of Toltcrodinc 4 mg: (A) Toltcrodinc; (B) 5—HMT; (C) Sum of
`Active Moieties [15]. Solid curves represent CYP2D6 PMs and dashed curves represent CYP2D6 EMs; EMs: Extensive Metabolizers; PMs;
`Poor Mctabolizers.
`
`profiles in the graph for fesoterodine (Fig. 4) fall within a
`much narrower range than the profiles for tolterodine ER.
`This indicates that the range of exposures to active moiety
`(i.e., PK variability) across subjects is considerably less vari-
`able for fesoterodine than it is for tolterodine ER. When the
`
`dose of fesoterodine is increased to 8 mg, the resulting expo-
`sure range from fesoterodine remains similarly narrow. At
`higher exposures, efficacy generally reaches a ceiling due to
`receptor saturation but the incidence of some adverse events
`may increase. it is because of the lower PK variability that
`more patients are likely to fall within the therapeutic range
`after either dose of fesoterodine. Therefore,
`it may be con-
`cluded that fesoterodine delivers markedly reduced variabil-
`ity in total active moieties relative to toltcrodinc,
`in part al-
`lowing for the development of a higher dose of fesoterodine.
`Higher drug exposures with fesoterodine 8 mg combined
`with lower variability may improve consistency and predict-
`ability of symptom response, allowing individualization of
`therapy in patients treated for OAB.
`
`In various Phase 1 studies in healthy volunteers, feso-
`terodine doses from 4 to 28 mg exhibited dose-proportional
`increase in 5—HMT exposures, without significant changes in
`
`time to maximal concentration or half life across doses [16,
`55, 56]. Following fesoterodine administration, both C...“
`and AUC of 5—HMT showed low coefficient of variation,
`suggesting reproducibility.
`in two Phase 2 studies,
`feso-
`terodine has been administered to OAB patients without sig-
`nificant safety concerns up to the supratherapeutic dose of 12
`mg;
`the efficacy analyses showed that the l2-mg dose of
`fesoterodine provided no further appreciable incremental
`benefit compared with the 8—mg dose [57, 58].
`
`5. Clinical Efficacy of Fcsotcrodinc
`
`it was demon-
`In a pooled analysis of two phase 3 trials,
`strated that fesoterodine has the ability to significantly re-
`duce OAB symptoms, including urgency and UUI, in a dose-
`dependent fashion [59]. The 8-mg dose provides significant
`additional benefit in improving most bladder diary variables
`compared with the lower dose and allows for dose escalation
`and individualization in subjects with OAB. This allows sub-
`jects to achieve a better balance between treatment efficacy
`and tolerability.
`In a Phase 3 trial, fesoterodine 4 mg pro-
`vided numerically greater symptom relief in nearly all effi-
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2074 - 0007
`
`

`
`4488 Current fl/Iea'r'cr'uaI C'Irt:rm'.u'!ry, 2009 VIII. 16, N0. 33
`
`A
`
`100
`
`Fesoterodine 4 mg
`
`U3
`
`Fesoterodine 8 mg
`
`Mkrflmtra at al.
`
`10
`
`r
`
`
`
`Tlme (hours)
`
`
`
`
`
`5-HMTPlasmaCan

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