throbber
E)_(p_ert
`Opinion
`
`1. Overview of the market
`
`.”"F'E“!"
`
`Introduction to the compound
`
`Chemistry
`
`Pharmacodynamics
`Pharmacokinetics
`and metabolism
`
`Clinical efficacy
`
`5-°.°°."'.°" Conclusions
`
`Safety and tolera bility
`
`Regulatory affairs
`
`10. Expert opinion
`
`Drug Evaluation
`
`Fesoterodine: a novel muscarinic
`
`receptor antagonist for
`the treatment of overactive
`
`bladder syndrome
`
`Martin C Michel
`
`Urziversity ofA msrerdam. Department afPImrmrzml'ogy zmd Pbarmmxotlaempy,
`Academic Medical Center, Meibergdreqff 5, I I 05 .A.ZAm5reralam, The Netherlands
`
`Background: Fesoterodine is a newly approved drug for the treatment
`of overactive bladder syndrome. Objective: The aim of this study was
`to review the preclinical and clinical data on fesoterodine. Methods:
`The study involved a search of the Medline database and the proceedings
`volumes
`of
`urological
`congresses. Results/conclusions:
`Fesoterodine
`functions as an orally active prodrug that
`is converted to the active
`metabolite
`5-hydroxymethyltolterodine
`by
`non-specific
`esterases.
`5-Hydroxymethyltolterodine is a muscarinic receptor antagonist. Fesoterodine
`is primarily eliminated as inactive metabolites along with significant renal
`excretion as the unchanged active metabolite 5—hydroxymethyltolterodine.
`Fesoterodine is indicated for use at doses of 4 and 8 mg once daily.
`In
`clinical studies both doses of fesoterodine were consistently superior to
`placebo in improving the symptoms of overactive bladder syndrome, with
`8 mglday having significantly greater effects than 4 mglday.
`
`Keywords: blood~brain barrier, Cytochrome P450 ZDG, fesoterodine,
`5-hydroxymetliyltolterodine, muscarinic receptor antagonist, overactive bladder syndrome
`
`Expert Ofiifi. P/mrrmlrat/Jar: (2008) 9U 0).'I787-I796
`
`1. Overview of the market
`
`Overactive bladder syndrome (OAB) is present in ~ 16% of the adult population
`and muscarinic receptor antagonists are the standard of care in its treatment |l].
`More recently they have also been considered for
`the treatment of male
`lower urinary tract
`symptoms, which historically have been attributed to
`benign prostatic hyperplasia [2]. Several representatives of this class are available,
`including darifenacin, oxybutynin, propiverine.
`solifenacin,
`tolterodine and
`trospiurn, with tolterodine being the global market leader. While generally well
`tolerated, their efficacy relative to placebo is only moderate [1,5]. While several of
`these drugs have multiple registered doses, close dependency is only poorly
`established. Moreover, many of these drugs have usage restrictions in special
`patient populations. Other drug classes have not been approved for use in OAB,
`but botulinum toxin and fiyadrcnoceptor agonists are currently in advanced
`clinical development.
`
`2. Introduction to the compound
`
`informa
`healthcare
`
`Fesoterodine functions as an orally active prodrug that is converted to its active
`metabolite 5-hydroxymethyltolterodine
`(5-HMT)
`by non-specific
`esterases.
`5—Hydro)-tymethyltolterodine is a competitive muscarinic receptor antagonist.
`
`10.1 51 7‘.’146S5560802l66944 © 2008 Informa UK Ltd ISSN 1465-6566
`
`1787
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0001
`
`

`
`Fesote rodine
`
`HO
`
`Fesoterodine
`
`Nonspecific
`esterases
`
`l
`
`OH YN
`H
`
`Tolterodine
`
`iCYP3A4
`
`T»
`CYP2D6
`
`Ho
`
`°” Y
`H
`
`I
`
`5-HMT
`
`J CYP3A4
`
`C
`
`OH
`
`H
`
`H
`
`N
`
`.
`
`O
`
`OH
`
`H
`
`T.
`CYF'2D6
`
`H0
`
`H
`
`N
`
`'
`
`T»
`CYP2D6
`
`Ho
`
`°” Y
`H Y
`
`5-CM
`
`X CYP3A4
`
`OH
`
`0 0 N
`T
`., Y
`
`HO
`
`Z.
`
`CYP2D6
`
`N-dealkylated metabolite
`(Tolterodine-specific)
`
`N-dealkylated 5-HMT
`
`N-dealkylated 5—CM
`
`Figure 1. Schematic depiction of the metabolism of tolterodine and fesoterodine.
`Courtesy of Pfizer Inc.
`S-CM: 5-Carboxy metabolite of 5-HMT.
`
`3. Chemistry
`
`The structures of fesoterodine and 5-HMT are shown in
`
`Figure 1. S-Hydroxymethyltolterodine is chemically identical
`to an active metabolite of tolterodine, which has also been
`
`mentioned in previous reports under the code names DD01,
`I’NU—200577 or SPM 7605.
`
`4. Pharmacodynamics
`
`rapidly and
`Following oral administration fesoterodine is
`completely metabolised to 5-HMT and the parent compound
`fesoterodine is not detected in the peripheral blood (see
`below). Therefore, most of the preclinical and clinical
`pharmacology of fesoterodine is
`relevant and discussed
`in relation to the active metabolite 5-HMT rather than
`
`The pharmacological properties of 5-HMT have been
`investigated in radioligand binding studies with cloned
`human muscarinic receptor subtypes and with endogenously
`expressed receptors in the urinary bladder, salivary glands
`and other tissues. An early study reported 5-HMT affinities
`values) of 2.3, 2.0, 2.5, 2.8 and 2.9 nM at cloned M1,
`M2, M3, M4
`and M5
`receptors,
`respectively, whereas
`tolterodine exhibited slightly but consistently lower affinities
`of 3.0, 3.8, 3.4, 5.0 and 3.4 nM, respectively [4]. A later
`study largely confirmed these findings and additionally
`reported that
`the parent drug fesoterodine had much
`lower affinities at all muscarinic receptor subtypes (631,
`501, > 1000, 158 and > 1000 nM,
`respectively)
`[5]. The
`affinities of 5-HMT, as determined in studies in the urinary
`bladder, salivary glands and other tissues of guinea-pigs [4],
`mice [6,7] and humans [8], are consistent with the data
`
`the parent compound fesoterodine
`
`from the cloned human receptor subtypes (Table 1). Thus,
`
`1 788
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0002
`
`

`
`Table 1. Affinities of tolterodine, fesoterodine and 5-HMT in radioligand binding studies and functional
`experiments.
`
`Tolterodine
`
`Fesoterodine
`
`5-HMT
`
`Ref.
`
`Michel
`
`Binding to cloned human muscarinic receptors (-log Ki)
`
`M2
`
`M2
`
`M3
`
`M3
`
`8.4
`
`8.2
`
`8.5
`
`7.9
`
`Binding to muscarinic receptors in tissues (-log Ki)
`
`Guinea pig bladder
`Mouse bladder
`
`Mouse bladder
`
`Human bladder (detrusor)
`
`Human bladder (urothelium)
`
`Guinea pig salivary gland
`
`Mouse salivary gland
`
`Guinea pig heart
`Mouse heart
`
`8.6
`8.9
`
`8.9
`
`ND
`
`ND
`
`8.3
`
`8.9
`
`8.8
`8.7
`
`Functional in vitro antagonism in the bladder (pKB)
`Rat
`ND
`
`Guinea pig
`Human
`
`ND: Not determined.
`
`8.5
`9.0
`
`ND
`
`6.3
`
`ND
`
`< 6.0
`
`ND
`ND
`
`ND
`
`8.6
`
`8.6
`
`ND
`
`ND
`
`ND
`ND
`
`8.7
`
`ND
`ND
`
`8.7
`
`8.8
`
`8.6
`
`8.2
`
`8.5
`9.1
`
`9.1
`
`ND
`
`ND
`
`8.3
`
`8.9
`
`9.0
`9.1
`
`8.8
`
`9.1
`9.0
`
`[4]
`
`[5]
`
`[4]
`
`[5]
`
`[4]
`[5]
`
`[7]
`
`[5]
`
`[8]
`
`[4]
`
`[5]
`
`[4]
`[5]
`
`[5]
`
`[11]
`[12]
`
`based on pharmacological characterisation combined with
`undetectable systemic exposure of fesoterodine after its oral
`administration to humans,
`the parent drug fesoterodine at
`its therapeutic doses is considered pharmacologically inactive
`and its clinical effects appear
`to be fully mediated by
`5—HMT [9]. Furthermore, 5-HMT lacks relevant affinity for
`a range of more than 50 other molecular targets [I0]. These
`data demonstrate that 5-HMT, as the active metabolite of
`
`fesoterodine, has a high selectivity for muscarinic receptors
`and recognises all of their subtypes with similar affinity.
`Nevertheless,
`a
`slightly higher
`affinity for muscarinic
`receptors in the bladder as compared to those in the salivary
`glands has been reported consistently across several studies
`(Table 1): this tissue selectivity can most likely be explained
`by differential tissue penetration [6].
`Functional in virro studies on the effects of 5-HMT have
`
`been reported from isolated bladder strips of rats [5], guinea-
`pigs
`[11]
`and humans
`[12]. 5-Hydroxymethyltolterodine
`caused a concentration-dependent parallel right shift of the
`concentration—response curve of the muscarinic receptor
`agonist carbachol
`in rats without affecting the maximal
`carbachol response: based upon this competitive antagonism
`an affinity (KB value) of 1.6 nM was calculated [S], which
`is
`in good agreement with the radioligand binding data.
`
`the
`fesoterodine under
`A similar apparent potency of
`same experimental conditions indicated rapid conversion
`of
`fesoterodine
`to
`5-HMT by
`the
`bladder
`strips.
`5-Hydroxymethyltolterodine
`also
`inhibited
`contraction
`of the rat bladder upon electrical field stimulation,
`that is
`due to the release of endogenous agonist, in a concentration-
`dependent manner
`and a
`concentration of 0.]
`].1M
`was
`sufficient
`for maximal
`inhibition [5]. Competitive
`antagonism with a similarly high potency was demonstrated
`for 5-HMT in bladder strips from guinea—pigs
`[4] and
`humans (Table 1)
`[12].
`In the latter study 5-HMT also
`inhibited the contraction induced by field stimulation but not
`the receptor-independent contraction evoked by KCI, confirming
`the specificity of this drug for muscarinic receptors.
`In cystometric in vivo studies in female rats intravenous
`5-HMT at a dose of 0.01 mglkg significantly increased
`bladder capacity by 10% and contraction intervals by 11%
`and reduced micturition pressure by 63% [5]: similar findings
`were also obtained with intravenous administration of
`fesoterodine.
`In similar
`studies with anaesthetised cats
`
`5-HMT inhibited bladder contraction with an lD50 of
`15 nmollkg, whereas a 50% inhibition of salivary secretion
`required a close of 40 nmollkg (p < 0.05 versus the bladder),
`while in parallel experiments the potency of tolterodine
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`1789
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0003
`
`

`
`Fesote rodine
`
`(ID50 values 101
`
`for both effects
`was consistently lower
`and 257 nmolfkg, respectively) [41,11].
`Oral fesoterodine or 5-1-IMT at doses of 30 mg/kg did
`not increase the gastrointestinal transit time in mice, whereas
`atropine (20 mgfkg), darifenacin (10 and 30 mg/kg) and
`solifenacin (10 and 30 mg/kg) inhibited the transit
`time
`by 33, 18 — 25 and 20 — 29%, respectively [13].
`Dedicated clinical studies on the possible adverse effects
`of 5-HMT on the CNS have not been reported yet.
`However, the CNS penetration of a drug depends largely on
`its lipophilicity [14].
`In this regard 5-HMT has a much
`lower
`lipophilicity than tolterodine
`(the octanol:water
`coefficient expressed as log D values was 0.74 versus 1.83)
`or other clinically used muscarinic receptor antagonists
`except for trospium [15,16]. Following oral administration of
`radiolabelled tolterodine only a
`small
`fraction of the
`radioactivity reaches the CNS and a CNS:blood ratio of
`radioactivity of 0.1 — 0.3 was
`reported in mice [15].
`Consistent with the lower lipophilicity of S-HMT relative
`to tolterodine,
`the CNS penetration of 5-HMT (assessed
`following oral administration of radiolabellcd fesoterodine)
`was even lower and a CNS:blood ratio of only 0.04 — 0.07
`was reported in mice [19]. These data indicate that, due to
`its
`low lipophilicity, 5-HMT exhibits a very low CNS
`penetration. This
`is
`consistent with data from mice,
`where > 80% of circulating active drug following tolterodine
`dosing was S-HMT and even considerably supratherapeutic
`doses did not affect memory [17]. Whether this translates
`into a clinically relevant advantage cannot be decided
`definitively
`based
`upon
`the
`presently available
`data,
`as
`tolterodine in doses up to 0.3 mg/kg also did not
`affect memory formation in rats under conditions where
`the same dose of oxybutynin or scopolamine significantly
`impaired it [18]. In non-selected groups of patients, including
`specific studies in the elderly [19], CNS-related adverse
`effects have typically not been reported with the use of
`tolterodine, but case reports have indicated that tolterodine
`may induce memory impairment or hallucinations in some
`individuals
`[20-23]. A recently reported dedicated study
`in VUlU.I1[CCl'S
`dC[f.‘C[Cd HO 3dVCfSC CFFCCKS Of
`[Ul[Cl'Od1Ilf.‘
`
`in a subgroup analysis
`on cognitive function [24]. However,
`of this
`study the phenotype for drug metabolism by
`CYPZDG [25] appeared to play a relevant role in adverse
`effects on rapid eye movement
`sleep. Thus, volunteers
`with the more frequently expressed ‘extensive metaboliser’
`(EM) phenotype, who can effectively convert tolterodine to
`S-HMT, exhibited no impairment of their REM sleep. In
`contrast, volunteers with the ‘intermediate metaboliser' (IM)
`or ‘poor metaboliser’
`(PM) phenotype, who produce little
`S-HMT from tolterodine [26], showed impaired REM sleep
`upon tolterodine treatment, that is the relative contribution
`of REM sleep declined significantly from 21.8 to 17.4% [24].
`These data indicate that
`the risk of CNS adverse events
`
`could be even lower with fesoterodine than with tolterodine,
`
`particularly in patients with the IM or PM phenotype with
`
`regard to CYPZDG: however, this remains to be confirmed
`in dedicated clinical studies.
`
`the available preclinical pharmacology
`Taken together,
`data demonstrate that S-HMT is a high-affinity, competitive
`antagonist of muscarinic receptors in the urinary bladder of
`several species, including humans and its potency is in good
`agreement with data from radioligand binding studies. Starting
`at
`low doses S-HMT affects bladder function in viva in
`
`experimental animals, as is to be expected from a muscarinic
`receptor antagonist. In direct comparative studies 5—HMT
`has an antagonist potency that is similar to or slightly higher
`than tolterodine. Consistent with
`its
`physicochemical
`difference to tolterodine,
`that
`is hydroxylation, 5-HMT
`exhibits less CNS penetration and, in patients with the IM
`or PM phenotype,
`this could translate into an even lower
`risk of CNS adverse events with fesoterodine.
`
`5. Pharmacokinetics and metabolism
`
`Following oral administration fesoterodine is not detected in
`the peripheral blood, thereby indicating a rapid and complete
`conversion to 5-1-IMT by non—specific esterases [9]. Although
`5-HMT is also a metabolite of tolterodine,
`there is an
`
`tolterodine
`fesoterodine,
`unlike
`distinction:
`important
`metabolism to 5-HMT is mediated by CYPZDG (Figure 1)
`and is subject
`to the CYPZDG phenotype of the patient.
`Accordingly, subjects with the EM phenotype of CYPZDG
`exhibit roughly similar serum concentrations of tolterodine
`and 5-I-IMT [11,27]. However,
`in subjects with the PM
`phenotype their plasma concentrations of tolterodine are
`considerably higher, with low or even undetectable levels of
`5-HMT [27]. Accordingly, the mean Cm” value and AUC of
`tolterodine can be five and 10 times higher,
`respectively,
`in subjects with the PM phenotype as compared to the
`EM phenotype with regard to CYPZDG [28]. Individual Cm“
`and AUC values span a range of over 100-fold across
`the genotypes [28]. In contrast, average plasma concentrations
`of 5-I-IMT following oral administration of fesoterodine
`differ by less than a factor of 2 in subjects with the PM
`and EM phenotypes, for example the values are 1.89 versus
`3.45 ng/ml, respectively, following a single dose of 4 mg
`of fesoterodine [29].
`
`A significant proportion of 5—HMT is excreted renally
`without additional metabolism and the renal clearance of
`5—I-IMT is ~ 250 ml/min, with > 15% of the administered
`
`fesoterodine dose excreted as unchanged S-HMT [30]. This
`raises the possibility that 5-HMT also could work from the
`luminal side of the bladder, an effect that may be clinically
`relevant based upon current discussions around the role of
`muscarinic receptors in the urothelium [31,32].
`pathways
`In
`addition
`to
`renal
`excretion, multiple
`exist for the metabolism of 5—HMT to inactive metabolites.
`
`are
`regard CYP2D6 and CYP3A4
`this
`In
`alternative and equally prominent pathways
`its carboxy and N-desisopropyl metabolites,
`
`two
`the
`form
`that
`respectively:
`
`1790
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0004
`
`

`
`each
`
`in
`
`turn
`
`is
`
`further metabolised
`
`to
`
`the
`
`final
`
`these
`[11]. Each of
`carboxy-N-desisopropyl metabolite
`three secondary metabolites is pharmacologically inactive.
`In
`addition to S-HMT,
`the
`inactive metabolites
`are
`also common between fesoterodine and tolterodine. The
`
`metabolic pathways of tolterodine and fesoterodine are
`shown schematically in Figure 1
`(the enzymes
`involved
`in the formation of the secondary metabolites have not
`been experimentally confirmed in all cases).
`The prodrug fesoterodine is absorbed almost completely
`from the gut, but non-specific esterases rnetabolise it rapidly
`and completely so that only S-HMT is detectable in the
`peripheral blood after oral administration of fesoterodine [9].
`This is also consistent with undetectable fesoterodine not
`
`only in the blood but also an absence of any appreciable
`fesoterodine being excreted in the urine or faeces. Although
`fesoterodine is completely converted to form 5—IcIMT,
`some of the converted 5-HMT is metabolised presystemically:
`nevertheless,
`the absolute bioavailability of 5-HMT from
`orally administered fesoterodine remains high, averaging
`~ 52% [33]. The pharmacokinetics of 5-I-IMT were found
`to be dose linear with the
`administered fesoterodine
`
`dose [29]. The appearance of 5-HMT in the blood is food
`independent
`[34], which allows
`fesoterodine to be taken
`with or without a meal. Specific Phase I studies have not
`demonstrated any evidence for clinically relevant pharmaco-
`kinetic differences between the genders or age groups [35]
`or between ethnic groups [56].
`Fesoterodine has been developed using a sustained release
`formulation for which the maximal plasma concentrations
`of S-HMT are reached after ~ 5 h with a terminal half-life
`
`of ~ 7 — 9 h across multiple studies in different populations
`with single and repeated dosing [9,29.3o,34.39]. In line with the
`half-life, no evidence of accumulation was
`seen upon
`fesoterodine treatment for multiple days in those studies.
`Taken fogffthef PIICSC ph3fInaCOl(lnC[lC data Suppflft Ofl.CC'd.3.lly
`administration of fesoterodine.
`
`With regard to special patient populations apart from
`gender, age [35] and ethnicity [36] the standard studies have
`been performed in patients with impaired kidney or liver
`function. In this regard one study compared subjects with
`normal
`renal
`function (glomerular
`filtration rate (GFR)
`> 80 ml/min) and those with a GFR of 50 — 80,
`
`30 — 50 and < 30 mlfmin [39]. Following log-transformation
`the
`5—HMT peak plasma concentrations
`in the renal
`impairment groups relative to those in healthy subjects were
`1.35, 1.48 and 2.03-fold, respectively: similar findings were
`obtained for
`the AUC values. These findings
`suggest
`that
`the starting dose of 4 mg/day of fesoterodine can
`also be used in patients with impaired renal
`function
`and can be titrated, with caution, to 8 mg/day in subjects
`with mild—to—moderate renal
`impairment.
`In contrast,
`the
`package inserts of tolterodine and tolterodine extended
`release (ER) require dose adjustment, at
`least for patients
`with severe renal impairment (GFR < 30 ml/min).
`
`Michel
`
`In another study the pharmacokinetics of fesoterodine
`were compared between healthy subjects and patients with
`moderate impairment of liver function (Child—Pugh B)
`[33].
`In the liver-impaired patients the Cm“ and AUC values were
`approximately twice the values of the healthy controls,
`whereas the terminal half—life was not significantly affected.
`These data indicate that the Starting dose of 4 mgfday can
`also be used in patients with mild-to-moderate hepatic
`impairment and can be titrated, with caution, to 8 mg/day
`in subjects with mild hepatic impairment. No data are
`currently available for patients with severe impairments of
`liver function and, thus, fesoterodine use is not recommended
`
`for
`the package inserts
`In contrast,
`in these patients.
`tolterodine and tolterodine ER require dose adjustment in
`all patients with impaired liver function.
`Finally, an interaction study with the potent CYP3A4
`inhibitor ketoconazol (200 mg) was performed,
`in which
`subjects with the EM and PM phenotypes with regard to
`CYPZD6 were analysed separately [37]. The maximal plasma
`concentrations of 5-HMT during ketoconazol administration
`in the EM and PM phenotype subjects were 2.2 and
`1.5
`times
`as high as
`in the absence of ketoconazol,
`respectively:
`the AUC value was affected similarly. These
`data suggest that the standard fesoterodine dose of 4 mg.’day
`can also be used in patients concomitantly receiving
`potent CYP3A4 inhibitors,
`regardless of their CYPZDG
`phenotype. In contrast,
`the package inserts for tolterodine
`and tolterodine ER do not
`recommend their use upon
`concomitant treatment with potent CYP3A4 inhibitors.
`Formulations allowing once-daily dosing are available for
`most of the muscarinic receptor antagonists used clinically
`for OAB treatment. In some cases this is achieved by specific
`formulations (e.g. oxybutynin, propiverine and tolterodine),
`while in other cases this is due to the intrinsic phatmacokinetic
`properties of the drug (darifenacin and solifenacin)
`[40].
`In this regard the very long elimination half-life of ~ 60 h
`for solifenacin deserves Consideration, as it may theoretically
`lead to accumulation [41].
`
`While some muscarinic receptor antagonists are primarily
`excreted Via the kidneys (e.g. trospium), others are intensively
`metabolised by the liver
`(e.g. darifenacin, oxybutynin,
`propiverine, solifenacin and tolterodine)
`[42]. Based upon
`these excretion pathways, dose adjustments are required in
`patients with impaired kidney or liver function as described
`in the respective package inserts. In contrast,
`the standard
`starting fesoterodine dose can be used in patients with
`impaired renal or hepatic function due to the combination
`of renal excretion and hepatic metabolism of 5~HMT.
`It
`is
`an
`intriguing observation from the
`available
`pharmacokinetic studies with fesoterodine administration
`that
`the interindividual variability in drug exposure is
`relatively small as compared to that reported, for example,
`for
`tolterodine (Figure 2). Obviously,
`such comparisons
`involve various processes, such as absorption, which is highly
`variable with trospium, metabolism and excretion [40]. In
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`1791
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0005
`
`

`
`doses but lasted 12 weeks and involved 173 — 186 patients
`per group [43],
`that
`is
`it had a design and size similar
`to many previous Phase III studies in this indication. This
`study also demonstrated significant
`improvements of both
`primary efficacy variables relative to placebo. Based on these
`data on efficacy and tolerability, fesoterodine doses of 4 and
`8 mg/day were selected for the pivotal Phase III studies.
`As the OAB definition by the International Continence
`Society does not
`require urodynamic proof of detrusor
`overactivity [46], one of the Phase II studies had also involved
`pressure—flow investigations
`[45]. Their
`analysis
`showed
`similar clinical improvements in patients with and without
`detrusor overactivity at each fesoterodine dose tested.
`The design of the two Phase III studies was in line
`with the regulatory recommendations by the EMEA and
`FDA,
`that
`is
`these were placebo-controlled,
`randomised,
`double-blind multicentre studies of 12 weeks duration
`
`with > 250 patients per treatment group. The inclusion and
`exclusion criteria were similar to those of studies with other
`
`muscarinic receptor antagonists that have been submitted
`for
`registration in recent years. One of the two studies
`(performed in the US) had three arms and compared
`fesoterodine doses of 4 and 8 mg/day with placebo [47].
`The other study (largely performed in Europe) had a very
`similar design but additionally included a fourth group of
`patients who received 4 mg/day of tolterodine ER as active
`control
`[48]. Secondary analyses of the pooled data from
`both Phase III studies have also been reported [49,50].
`The patients in both Phase III studies had rather similar
`baseline characteristics and also exhibited rather similar
`
`[47.48]. Therefore,
`symptom improvements upon treatment
`they are described together. Both doses of fesoterodine were
`significantly more effective than placebo for
`the three
`co-primary efficacy variables of urinary frequency, urgency
`incontinence
`episodes
`and the percentage of patients
`reporting a treatment
`response (Figure 3), as well as for
`many secondary efficacy variables including the number of
`urgency episodes. The extent of symptom improvement was
`in a range similar to that reported with other muscarinic
`receptor antagonists
`in studies of a similar design [1].
`Only the symptom of nocturia was not consistently improved
`by fesoterodine. This is not surprising based upon the
`low baseline
`level of nocturia
`in these
`studies,
`the
`causes of nocturia and the observations
`multifactorial
`
`that other muscarinic receptor antagonists also did not
`yield consistent nocturia improvements in OAB patients [5]].
`An efficacy parameter, which has
`rarely been used in
`other studies but which may have direct
`relevance for
`patients, was
`the extrapolated number of incontinence-
`free days per week:
`this was consistently improved by
`both fesoterodine doses in both studies.
`
`Although several doses are available for most muscarinic
`receptor antagonists
`for OAB treatment,
`a significantly
`greater benefit with higher doses has typically not been
`reported [1]. In contrast, the fesoterodine dose of 8 mg/day
`
`Fesote rodine
`
`l
`
`_. C)O
`
`
`
`Coefficientofvariation(°/o)
`
`Figure 2. Variability (expressed as coefficients of variation)
`in maximal plasma concentrations upon oral treatment
`with fesoterodine (assessed as 5-HMT)
`in comparison
`with published data on tolterodine. The figure is based
`upon published data [9,25,27,29,34—35,3a—4o,55]. Note that data
`from both genders and different age and ethnic groups were
`included, whereas data from diseased subjects (other than OAB)
`were excluded.
`
`the case of fesoterodine the CYPZDG-independent generation
`of the active drug moiety appears to be a key explanation
`for
`the relatively small variability in 5-HMT exposure.
`Irrespective of such mechanistic considerations, a relatively
`small variability in exposure may be therapeutically beneficial,
`as it may lead to a greater predictability in clinical response.
`However, whether
`this indeed translates into a clinically
`relevant benefit cannot be determined from the presently
`available data.
`
`5. Clinical efficacy
`
`The clinical efficacy and tolerability of fesoterodine has been
`explored in 20 Phase I (partly described above), three Phase
`II and two Phase III studies. This includes a thorough study
`on the possible effects on QT intervals (see the section on
`clinical
`tolerability). Fesoterodine doses of 2, 4,
`8 and
`I2 mgfday were tested in placebo-controlled, randomised,
`double—blind multicentre
`trials
`during
`the Phase
`II
`studies [43.44|. One of these studies also involved urodynamic
`pressute—flow investigations
`[45]. The primary efficacy
`parameter of the Phase II studies was the number of daily
`micturition and urgency incontinence episodes. In the study
`performed in the US, which involved an 3-week treatment
`of only 38 — 47 patients per group, significant improvements
`relative to placebo were observed for both efficacy parameters,
`despite the small numbers of subjects, at fesoterodine doses
`of 4, 8 and 12 mg/day [44]. The second trial, performed
`in Europe,
`Israel and South-Africa,
`involved the same
`
`1792
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0006
`
`

`
`Michel
`
`was significantly more effective for most efficacy parameters
`than 4 mgfday in the pooled analysis of both Phase III
`studies [30]. Given the adverse effects of OAB on quality of
`life it also appears
`important
`that significant symptom
`l.IiIl.pl'OVCII1C1'lt5 HIE delfflctablff
`35 Carly 35 2 WeEk5 after
`initiation of fesoterodine treatment, that is the earliest time
`
`point of assessment after randomisation [50]. In this context
`it is also relevant that several rating scales assessing patient
`reported
`outcomes
`and
`quality of
`life
`have
`shown
`dose—dependent
`and significantly greater
`improvements
`with fesoterodine than with placebo [52].
`overall
`the
`in
`With regard
`to
`clinical
`end-points
`OAB population, comparisons among data sets obtained
`from different
`studies are problematic and only direct
`comparative studies
`allow reliable conclusions
`[1,3]. At
`present, one direct comparative study between fesoterodine
`and another muscarinic receptor antagonist exists, which is
`the four—armed Phase III study also including a patient
`group receiving tolterodine ER at 4 mg/day [48].
`In that
`study 8 mg/day of fesoterodine was numerically superior to
`both 4 mg/day of fesoterodine and 4 mg/day of tolterodine
`ER with regard to efficacy, but also had more adverse events
`(primarily dry mouth). A subsequent post /90: analysis of
`this study, comparing maximum registered doses of both
`agents,
`reported that fesoterodine at 8 mg/day was also
`statistically superior to tolterodine ER at 4 mg/day for the
`primary end-point
`(the number of urgency incontinence
`episodes) as well as for several secondary end-points [53].
`In contrast,
`the incidence of adverse events was almost
`
`identical with 4 mglday of fesoterodine and tolterodine ER,
`allowing for a direct comparison of their efficacy. Fesoterodine
`at 4 mg/day performed numerically better than tolterodine
`ER at 4 mg/day across all efficacy parameters, but a
`statistical analysis of this comparison has not been reported.
`Therefore,
`the present data only allow the conclusion
`that, at a similar adverse event
`incidence,
`the efficacy
`of 4 mg of fesoterodine is at
`least as good as that of
`tolterodine ER.
`
`7. Safety and tolerability
`
`In the Phase III trials, adverse events were noted in 55% of
`
`in the US study,
`the patients during placebo treatment
`whereas the corresponding incidences were 61 and 69%
`with 4 and 8 mglday of fesoterodine, respectively [47]. In the
`mainly European study these incidences were 38% with
`placebo and 50 and 58°43 with 4 and 8 mg/day of
`fesoterodine, respectively [43]. As expected for a muscarinic
`receptor antagonist, dry mouth was the most
`frequently
`reported adverse event but was rated as mild to moderate
`in most cases. In one Phase III study it was seen in 7, 16
`and 36% of patients receiving placebo, 4 and 8 mgfday
`of
`fesoterodine,
`respectively [47], whereas
`in the other
`Phase III study it was seen in 7.1, 21.7 and 33.8% in
`the same groups (16.9% for 4 mg/day of tolterodine) [43].
`
`20
`
`ski
`
`Micturitions
`
`it
`
`¢,v°
`\9°
`<2
`
`569
`
`<9
`us‘?
`0%
`o
`K\-
`(<56:
`{<69
`«o
`Urgency urinary incontinence
`
`”
`
`029°
`Q\@-
`
`40*
`
`bxég
`
`QQCQO
`Treatment response
`
`b-dig
`
`‘bioq
`Q00
`
`15
`
`E
`.9
`‘G
`.3 10
`
`E3
`
`9
`
`5
`
`0
`
`100
`
`so
`
`r:
`u3
`-3 so
`
`E 4
`B2
`
`0
`
`20
`
`0
`
`100
`
`so
`
`60
`
`40
`
`20
`
`0
`
`E1::
`I:
`.In
`E
`39
`
`on
`
`o°°°
`Q\%
`
`b-‘iii
`45‘
`
`D-($5
`
`«Q-3°
`
`QFQ
`(<9ro°
`
`Figure 3. Efficacy of 4 and 8 mglday of fesoterodine in
`comparison to that of placebo and 4 mglday of tolterodine
`ER in the European Phase III study for the three co-primary
`end-points: number of micturitions (baseline 11.5 — 12.0I24 h
`across all groups), number of urgency incontinence episodes
`(baseline 3.7 — 3.8/24 h across all groups) and treatment
`responders (defined as ‘greatly improved’ or 'improved').
`Adapted from [48].
`
`Expert Opin. Pharmacother. (2008) 9(10)
`
`1793
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2073 - 0007
`
`

`
`Fesote rodine
`
`reported adverse events during fesoterodine
`The other
`treatment were also qualitatively and quantitatively in the
`range of what has been reported with other modern
`muscarinic receptor antagonists [I].
`Based upon recent concerns regarding the cardiovascular
`safety of drugs in general, a thorough study of the possible
`effects on QT intervals was performed [33]. This included
`parallel groups of 64 — 68 subjects each, who were treated
`for 3 days with 4 mg/day of fesoterodine,
`the highly
`supratherapeutic dose of 28 mgfday of fesoterodine,
`the
`active control moxifloxacin at 400 mgfday or placebo. Both
`the standard dose of 4 mg.’day and the highly supratherapeutic
`dose of 28 mg/day did not provide any evidence of QT
`prolongation (e.g. QTC for 28 mg/day from 404.5 i 16.7 to
`400.] i 14.0 ms. with delta: -5.0 i 7.9 ms). In comparison
`a similar study with tolterodine showed no effect on QTC at
`the recommended doses (2 X 2 mg/day) whereas a modest
`QTC prolongation (5.6 i 2.1 ms) was
`seen at
`the
`supratherapeutic dose of 2 X 4 mg/day: while the authors
`considered that
`to be clinically irrelevant
`[27], others have
`questioned this interpretation [54].
`
`8. Regulatory affairs
`
`Based upon the accumulated preclinical and clinical data
`the EMEA granted marketing authorisation approval
`for
`fesoterodine in April 2007. A registration procedure with
`the US authorities is under way.
`
`9. Conclusions
`
`Taken together, the available Phase II and III data show that
`fesoterodine at 4 and 8 mg/day is effective in the treatment
`of OAB symptoms. The overall efficacy and tolerability are
`in the range of what has been reported with other muscarinic
`receptor antagonists. A major difference as compared to
`other drugs is a significant dose dependency of its efficacy.
`This could be related to the smaller variability in exposure
`to active drug (see above), which facilitates the detection of
`
`dose differences due to a smaller data scatter. Moreover,
`
`based upon its pharmacokinetic and metabolic profile, only
`small restrictions

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