throbber
Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0001
`
`

`
`E. Callegari et al.
`
`Introduction
`
`Overactive bladder (OAB) is a syndrome defined by the
`International Continence Society as urgency, with or
`without urgency incontinence, usually with increased
`daytime frequency and nocturia [1]. OAB affects at least
`10% of the adult population [2] and the prevalence
`increases with age [3]. Postulated aetiologies for this con-
`dition include increased afferent activity, decreased inhi-
`bitory control and increased sensitivity of the detrusor
`muscle to efferent stimulations [4, 5]. Muscarinic receptors
`are thought to mediate the detrusor contractions of
`normal voiding, but in OAB the muscarinic receptors are
`associated with bladder contraction leading to urinary fre-
`quency, urgency and urgency incontinence. Antimuscar—
`inic agents, such as darifenacin, oxybutynin, solifenacin,
`trospium, tolterodine and fesoterodine have consistently
`demonstrated significant efflcacy for the treatment of OAB
`symptoms [6, 7]. However, treatment is associated with
`typical anticholinergic side effects such as dry mouth,con—
`stipation, somnolence and blurred vision. The adverse
`effects (AE5) of antimuscarinic drugs may occur because
`muscarinic receptors are located throughout the body [8]
`where inhibition of specific receptor subtypes is associated
`with side effects. For example, in the central nervous
`system (CNS), muscarinic receptors, particularly the M.
`subtype, are thought to play an important role in the con-
`solidation of long-tenn memory. Cognitive impairment is
`associated with anticholinergic therapy in the elderly [9]
`and the side-effect profiles seen with some antimuscarinic
`OAB drugs are consistent with inhibition of muscarinic
`receptors in the CNS [10]. Although the incidence of CNS
`AEs of antimuscarinic agents is generally much lower than
`that of dry mouth [6, 7], CNS AEs can be of great concern,
`particularly in the elderly [1 1]. The incidence of CNS AEs
`among the available antimuscarinic agents for OAB seems
`to differ. For instance, while darifenacin has been shown to
`
`have no significant effects on memory vs. placebo, oxybu-
`tynin ER caused significant memory deterioration which
`was deemed comparable with brain ageing of 10 years
`[12]. The ability of certain antimuscarinic OAB drugs to
`exert CNS-related pharmacological effects at therapeutic
`doses for OAB treatment depends on their ability to pen-
`etrate the CNS as well as relative affinity for relevant mus-
`carinic receptor subtypes in the CNS, particularly M.
`[13—1S]. CNS penetration of drugs depends on the perme-
`ability properties of the blood—brain barrier (BBB) [16, 17]
`and the influence of active efflux transporters present in
`brain tissue, such as P-glycoprotein (P-gp) [18, 19].There-
`fore, the relative permeability and affinity of OAB agents
`for P-gp is an important consideration in understanding
`their potential to exert AEs manifested in the CNS.
`The purpose of the present paper was to present a
`comprehensive set of non—clinica|
`in vitro and animal
`studies that investigated in parallel the CNS penetration
`potential of antimuscarinic OAB drugs. The following
`
`236 I 72:2 / Br] Clin Pharmacol
`
`studies were conducted: (i) physicochemical characteriza-
`tion, including lipophilicity; (ii) in vitro RRCK cell passive
`permeability assessment; (iii) in vitro P-gp mediated efflux
`in MDCK—MDR1 transcellular flux assay; and (iv) in vivo
`brain, plasma and CSF concentrations following a single
`subcutaneous dose in rats. Strategies for assessment of
`brain penetration of compounds have focused on deter-
`mination of unbound brain : unbound plasma concentra-
`tion ratios (Kp,free), and consideration of the involvement
`of transporter proteins at the BBB, in particular P-gp [19].
`Therefore, to understand further CNS disposition, the
`unbound brain : unbound plasma concentration ratios
`were estimated using brain and plasma binding experi-
`ments in vitro [19, 20].The overall aim of this package of
`data was to enable an understanding of the brain penetra-
`tion potential of antimuscarinic OAB drugs in relation to
`their physicochemical and permeability properties. The
`antimuscarinic agents included in these studies were
`5—hydroxymethyl tolterodine (5—HMT, the active metabo-
`lite of tolterodine as well as fesoterodine), darifenacin,
`
`oxybutynin, solifenacin, tolterodine and trospium. Fes-
`oterodine is a pro—drug that is rapidly and extensively con-
`verted to 5-HMT by esterases in vivo, and is not detectable
`after oral administration [21, 22]. Therefore, 5-HMT was
`
`evaluated as the relevant active moiety of fesoterodine in
`this study.
`
`Methods
`
`Materials
`
`Oxybutynin, N-methylscopolamine HBr and atropine
`(internal standard) were purchased from Sigma-Aldrich.
`5-hydroxymethyl tolterodine (5-HMT), solifenacin and tro-
`spium chloride were purchased from Toronto Research
`Chemicals Inc. (Ontario, Canada).
`
`Fesoterodine and scopolamine were obtained from
`Pfizer Global Research and Development central com-
`pound stores (Milwaukee, WI, USA).To|terodine was pur-
`chased from Sequoia Research Products (Pangbourne,UK).
`Darifenacin was purchased from Toronto Research Chemi-
`cals Inc. (Ontario, Canada) and Sequoia Research Products
`(Pangbourne, UK). Hanks's balanced salt solution (HBSS),
`cell culture media and supplements were purchased
`from lnvitrogen (Paisley, UK). All other reagents were
`obtained from Sigma-Aldrich (Poole, UK) or J.T. Baker
`(Phillipsburg, NJ. USA).
`
`Assessment of physical properties
`of compounds
`The definitions of physical properties of compounds were:
`log D, logarithm of the distribution coefficient between
`octanol and buffer at pH 7.4;c|og P, logarithm of the calcu-
`lated partition coefficient between octanol and water,
`polar surface area (PSA), the area of molecular surface
`belonging to polar atoms (units: A’) [17], rotatable bond
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0002
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0002
`
`

`
`count, count of all non—terrnina| single bonds and hydro-
`gen bond acceptor and donor counts, counts of all atoms
`in a molecule that are potentially involved in a hydrogen
`bond as acceptor or donor atoms, respectively. All calcu-
`lated parameters were determined within the proprietary
`Pfizer database (RGate), where clog P was determined
`using the BioByte program clog P, version 4.3 and PSA
`using a published method [17]. Log D values were taken
`from published sources [23].
`
`Monolayer efflux studies in MDCK
`(Madin-Darby canine kidney) and
`RRCK cell lines
`
`MDCK-MDR1 expressing P—gp were originally obtained
`from Netherlands Cancer Institute (Amsterdam, the Neth-
`
`erlands). RRCK cells were generated in house (Pfizer lnc.,
`Groton, MA, USA) as a subclone of MDCK wi|d—type (MDCK-
`WT) cells that displayed low expression of endogenous
`P—gp (approximately 1—2% of MDCK—WT cells, based on
`mRNA level). The rank order of permeability values for
`compounds whose transcellular flux was predominantly
`by passive diffusion were similar for RRCK and MDCK-WT
`(data not shown). Monolayer efflux studies were con-
`ducted as previously described in the literature for MDCK-
`MDR1 cells [24]. Cells were cultured in minimal essential
`
`medium on with supplements and passaged when 70-80%
`confluent. Cell monolayer flux studies were conducted 5
`days after seeding in 24-well transwell inserts [MDCK-
`MDR1 in 0.4-pm pore size (Corning Costar) at 1.8 x 105
`cells cm"; RRCK in 1-pm pore size (Becton Dickinson,
`Cowley, UK) at 4.2 x 10‘ cells cm"]. Donor and acceptor
`solutions were prepared from HBSS, containing HEPES at
`20 mM, pH 7.4. Stock solutions of test compounds were
`prepared at 10 mM in dimethyl sulphoxide (DMSO) and
`used to prepare donor solutions of 2 pM compound in
`0.05% (v/v) DMSO and also containing 2 pM nadolol used
`as monolayer integrity marker. Apparent permeability
`(Paw) of compounds was detennined in apical to basolat-
`eral (A—>B) and basolateral to apical (B—)A) directions in
`triplicate by incubation with compound for 2 h at 37°C.
`Samples of medium (20 pl) from both donor and acceptor
`chambers were analysed by tandem liquid chromatogra-
`phy and mass spectrometry (LC/MS-MS). The LC/MS-MS
`system consisted of a 2.1 x 15 mm C18 optilynx column
`(Optimize Technologies lnc., Oregon city, OR, USA) in line
`with Onyx monolithic C18 column 50 x 4.6 mm (Phenom-
`enex, Torrance, CA, USA) operating at a flow rate of
`3 ml min". The aqueous mobile phase consisted of 90%
`2 mM ammonium acetate, 10% methanol, 0.1 % formic acid.
`
`The organic mobile phase was 10% 2mM ammonium
`acetate, 90% methanol, 0.1 % formic acid. Mass spectrom-
`etry was performed on a SCIEX API 4000 triple quadrupole
`mass spectrometer (Applied Biosystems, Ontario, Canada),
`with turbo ion spray source. Data were acquired in positive
`ion mode with an ion spray probe voltage of 5.5 kV. The
`following selected reaction monitoring transitions, given
`
`CNS penetration potential of OAB agents
`
`as mass : charge ratio (m/z) were used to measure
`the compounds: tolterodine m/z 310—>201 and nadolol
`m/z 310—>254 at collision energy of 25 eV; darifenacin
`m/z 427—>147, oxybutynin m/z 358—>124, 5—HMT m/z
`342—>223, fesoterodine m/2 412—>223 and nadolol m/z
`
`310—>201 at a collision energy of 40 eV; solifenacin
`m/z 364—>110, trospium m/z 392—>182 and nadolol m/z
`310—)56 at a collision energy of 55 eV.
`P,,,,, values were calculated according to the equation
`P,,,,, = (Q/t) x 1/Co x 1/A, where Q is the sampled concen-
`tration in the acceptor compartment, t incubation time; Co
`is the initial concentration in the donor compartment and
`A is the area of the filter of the transwell plate. Monolayers
`with nadolol Paw values of less than 1 x 105cm s" were
`deemed intact.
`
`In vivo brain penetration study
`All procedures performed on animals were in accordance
`with US federal regulations and established NIH guidelines
`and were reviewed and approved by the Institutional
`Animal Care and Use Committee. Male Sprague-Dawley
`rats weighing approximately 250 g (n = 3) received a single
`dose of compound subcutaneously (0.3 or 1mg kg").
`Dosing solutions were prepared fresh in saline on the
`day of the study and were administered at 1 ml kg“. At
`1 h postdose, animals were euthanized using CO2 and
`approximately 3 ml of blood removed by cardiac puncture
`and plasma prepared by centrifugation. Cerebrospinal
`fluid (CSF) was drawn from the cistema magna using a 25
`gauge needle attached to polyethylene tubing (2 mm) and
`a syringe. Brains were removed, rinsed with saline and
`weighed. All samples were immediately frozen on dry ice
`after processing and stored at —20°C until analysis. Brain
`samples were homogenized in four volumes of phosphate
`buffered saline (1 in 5 tissue dilution). Plasma, CSF and
`
`brain samples were analysed using a LC/MS-MS method.
`Calibration standard and quality control
`(QC) samples
`were prepared in untreated rat plasma or brain homoge-
`nate. The lower limit of quantitation (LLOQ) for all com-
`pounds was
`0.1 ng ml“ while the upper
`limit of
`quantitation (ULOQ)
`ranged from 25 to 100 ng mf‘,
`depending on the dose received. Aliquots of plasma or
`brain (50 pl) were basified using 50 mM K2HPO4, pH 8.0
`(200 pl) and mixed with internal standard (atropine, 10 pl
`of 50 ng ml“) before loading onto a 96-well solid phase
`extraction plate (Waters Oasis HLB, 10 mg). Following
`several washes (400-pl water, followed by 400-pl metha-
`nol :water 10:90), compounds were eluted using methanol
`(400 pl) and eluant dried under nitrogen heated to 40°C.
`Samples were reconstituted in 50-pl 5mM ammonium
`acetate buffer containing 0.1% fonnic acid (v/v) before
`analysis by LC/MS-MS. Extracted samples (10 pl) were
`injected onto a Phenomenex Luna C18 analytical column
`(5 pM, 2.1 x 50 mm). The liquid chromatography system
`consisted of a gradient mixture of acetonitrile and 5 mM
`ammonium acetate pH adjusted with formic acid (0.1%,
`
`Br] Clin Pharrnacol
`
`/ 72:2 / 237
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0003
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0003
`
`

`
`E. Callegari et al.
`
`v/v) maintained at a flow rate of 0.3 ml min"‘. Samples con-
`taining concentrations above the ULOQ were diluted with
`control matrix and re—ana|ysed. CSF samples (20 pl) were
`mixed with internal standard and ammonium acetate
`
`buffer [5 mM with 0.1% formic acid (v/v)] and were quanti-
`fied using a direct—inject method onto the LC/MS—MS
`system described above with standard curves prepared in
`untreated rat plasma ultrafiltrate. Data were acquired in
`positive ion mode with an ion spray probe voltage of
`5.5 kV using the same reaction monitoring transitions as in
`the transcellular flux experiments, with the internal stan-
`dard atropine monitored at m/z 290—>124.
`
`Plasma and brain binding
`Binding of compounds to rat plasma or rat brain was deter-
`mined by a previously described equilibrium dialysis
`method [20]. Briefly, untreated plasma or brain homoge-
`nate (5x diluted with PBS) wasfortified with test compound
`to yield a final concentration of 1 pM. Samples were placed
`in a 96-well equilibrium dialysis block (HTDialysis, Gales
`Ferry,CT, USA) fitted with Spectra—Por 2 membranes (Spec-
`trum Laboratories, Rancho Dominguez, CA, USA) and incu-
`bated at a temperature of 37°C, humidity of 95% and CO;
`concentration of 5%. After 6 h,a|iquots (10 pl) of buffer and
`matrix were removed and added directly to a 96-well
`polypropylene block containing internal standard in aceto-
`nitrile (atropine, 50 ng mF‘).An equal volume of the oppo-
`site matrix was added to each sample to yield unifonn
`sample composition. All samples were analysed using
`LC/MS—MS with conditions similar to those described
`
`above.The unbound fraction in plasma (f..,,) was calculated
`using the ratio of drug concentrations in buffer to matrix.
`Unbound fraction in brain (f...,) was calculated in a similar
`fashion but tissue dilution factor (D) was taken into account
`
`according to the correction described by Kalvass & Maurer
`[20], whereby f..., = 1/D/[(1/f..2) — 1] + I/D, where f..; =
`unbound fraction measured in diluted tissue homogenate.
`
`Analysis of in vivo brain penetration data
`The treatment of data obtained from in vivo brain penetra-
`tion studies has been discussed extensively in the litera-
`ture and has underlined the value of applying knowledge
`
`Table 1
`
`Physicochemical properties of various antimuscarinic agents
`
`of tissue binding in comparing tissue concentrations, and
`hence understanding brain penetration [19, 20, 25]. Fur-
`thermore, comparsion of results obtained using unbound
`brain tissue homogenate concentrations with in vivo
`microdialysis has supported the use of free concentrations
`in brain and CSF as surrogates for concentrations in brain
`interstitial fluid [26]. Therefore, the tissue concentration,
`
`plasma protein binding and brain binding data were used
`to calculate the following parameters:
`
`Brain : plasma ratio (8 : P):
`total concentration in brain (ng g"):
`total concentration in plasma (ng mF')
`
`Unbound concentration in plasma =
`total concentration in plasmaxfup
`
`Unbound concentration in brain:
`
`total concentration in brainxf,,.,
`
`Ratio of unbound fraction fractions
`
`in brain and plasma: f,,, :f.,,,
`
`Kp,free = unbound concentration in brain:
`unbound concentration in plasma
`
`CSF : free plasma ratio:
`concentration in CSF: unbound concentration in plasma
`
`Results
`
`Physicochemical property assessment of
`OAB agents
`The physicochemical properties that describe the lipophi-
`licity, hydrogen bonding potential, polarity and flexibility
`of the range of antimuscarinics studied are summarized in
`Table 1.The compounds used for this investigation possess
`relatively low molecular weight (range 325-426 Da),
`typical of small drug molecules and, with the exception of
`trospium, can be classed as lipophilic based on their log D
`and calculated log P values (ranges log D 0.74—>3.3 and
`
`Antimustarinlc agent
`
`Clog P
`
`Log D
`
`Hydrogen bond acceptors
`
`Hydrogen bond donors
`
`
`
`Rotatable bonds
`
`Molecular weight (Dal
`
`PSA (A2)
`
`For definition of parameters in the table, refer to Methods.
`
`238 / 72:2 / Br|C|in Pharrnacol
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0004
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0004
`
`

`
`CNS penetration potential of OAB agents
`
`I
`
`mill:
`
`§6
`
`
`
`Pappx|0"cms" 33
`.
`
`Trosplum 5-I-IMT DarIfen- Solifen- ToItero- Oxybut-
`acln
`acin
`dine
`ynln
`
`I 3.7
`
`4.0
`
`I .3
`
`I .4
`
`0.6
`
`B-*A:A-B
`efflux ratio 4.3
`
`III9
`
`AO
`
`PappxI0'‘cms" —NWOO9
`
`0 T
`
`rvosplum 5-HMT Darlfe- SoIifen- ToIteno- Oxybut-
`nacin
`acin
`dine
`ynln
`
`Figure 1
`Passive transport of antimuscarinic agents in RRCK cells. Fluxes of corn-
`pounds aaoss RRCK cell monolayers in the apical to basolateral (A—)B)
`direction are shown. lncubations were performed in triplicate and error
`bars represent SDs
`
`clog P 3.6-4.9). The rank order of lipophilicity determined
`by log D [23] was oxybutynin > darifenacin > tolterodine =
`solifenacin z fesoterodine > SHMT > trospium.Trospium
`(log D -1 .22,c|og P -1.2) is a much more hydrophilic com-
`pound in comparison due to the presence of a quaternary
`amine group that is ionized at physiological pH. Hydrogen
`bonding potential evaluated as the sum of hydrogen
`bond donors and acceptors was broadly similar between
`compounds, with rank order darifenacin = 5-HMT (5) >
`oxybutynin = trospium (4) > tolterodine = solifenacin =
`fesoterodine (3). All the compounds possessed relatively
`low PSA values ranging from 23.5A’ for tolterodine to 55.6
`A’ for darifenacin.The number of rotatable bonds in a mol-
`
`ecule is a measure of the flexibility of the compound. The
`range in the number of rotatable bonds in this series of
`compounds is relatively large with solifenacin having the
`lowest value (4) and 5-HMT and darifenacin having the
`highest value (8).
`
`Transcellular flux across RRCK and MDR 1 -
`MDCK cell monolayers
`In vitro cell membrane permeability and the influence of
`P-gp were assessed in cell lines designed to measure tran-
`scellular flux in the presence and absence of P-gp. A stable
`population of MDCK cells was selected by flow cytometry
`to have little or no functional P-gp that would contribute
`to efflux of substrates of this protein. In this cell line, des-
`ignated RRCK, compounds displayed a range of flux
`values in the apical
`to basolateral direction (A—>B)
`(Figure 1), and compounds could be classed as possessing
`low (<5 x 10" cm 5''), moderate (5-1 5 x 10* cm s") or high
`(>15 x 10* cm s") transcellular flux Thus, trospium (P.,,,, =
`0.63 x 10" cm s") possessed low flux, 5-HMT moderate
`and solifenacin, tolterodine, darifenacin and oxybutynin
`high flux values.
`The ability of compounds to act as P-gp substrates was
`assessed in MDCK cells transfected with the human mdri
`
`gene that expresses P-gp (MDCK-MDR—1). The ratio of
`
`Figure 2
`Transcellular flux of antimuscarinic agents in MDCK-MDR1 cells. Fluxes of
`compounds across MDCK-MDR1 cell monolayers were measured in apical
`to basolateral (A—>B) and basolateral to apical (B—)A) directions.The ratio
`of A—)B/B~>A fluxes (efflux ratio) is shown under each compound. Incu-
`bations were perfonned in triplicate and error bars represent SDs.A—>B
`flux (Z ); B—>A (-)
`
`fluxes
`to apical
`to basolateral : basolateral
`apical
`(A—>B:B—>A efflux ratio) was used as an index of P—gp-
`mediated
`efflux
`across MDCK—MDR—1 monolayers
`(Figure 2). The efflux ratios shown by the range of com-
`pounds varied between 0.6 and 13.7. Using an efflux ratio
`of >2 to indicate significant P-gp—mediated efflux [27], it
`was found that trospium, 5-HMT and darifenacin were
`likely to be substrates of the P-gp transporter, whereas
`solifenacin, tolterodine and oxybutynin were unlikely to
`act as substrates.
`
`In vivo CNS penetration of OAB agents in rats
`Detennination of plasma, brain and CSF drug concentra-
`tions by specific LC/MS-MS afforded the required sensitiv-
`ity and selectivity for accurate detennination of drug
`concentration in tissues and ensured that metabolites did
`
`not contribute to apparent drug concentrations (the tissue
`concentration data used to determine brain penetration
`are shown in Table 2). CNS penetration was initially
`assessed by calculating the total brain : plasma concentra-
`tion ratios (B : P) and CSF :free plasma concentration ratios
`(CSF :free plasma). During collection of the brain in these
`experiments, blood remains in the tissue and following
`homogenization and LC/MS-MS analysis the residual
`blood can contribute an observed B:P of up to 0.04,
`reflecting presence of drug in the vasculature of the brain
`rather than true brain penetration [28]. Hence, values of
`B: P below 0.04 and CSF :free plasma approaching 0 are
`consistent with no significant brain penetration and no
`equilibrium between CSF and free plasma. The results of
`CNS penetration in vivo are summarized in Tab|e3 and
`displayed graphically in increasing order of their CNS pen-
`etration in Figure 3.
`In parallel experiments, scopolamine (0.3 mg kg") and
`N-methylscopolamine (0.3 mg kg") were administered as
`
`Br] Clin Pharmacol
`
`/ 72:2 / 239
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0005
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0005
`
`

`
`E. Callegari et al.
`
`Table 2
`
`Mean concentrations and unbound fractions of antimuscarinic agents in rat tissues following subcutaneous dosing
`
`.
`.
`
`Compound
`
`Tiospiuln
`S-HMT
`Darifenacin
`solifenacin
`Tolterodine
`Oxybutynin
`N-Inethylsoopolamine
`Scopolamine
`
`Plasma
`concentration
`(ng ml-‘)
`
`106 1 85
`27.3 1 1.3
`39.0 1 5.8
`59.3 1 4.6
`20.8 1 2.7
`22.5 1 5.2
`45.9 1 6.3
`29.8 1 2.0
`
`Brain
`concentration
`(ng g")
`2.68 1 1.31
`4.38 1 0.58
`3.04 1 0.10
`180 1 55
`61.3 1 6.7
`141 1 27
`2.33 1 0.92
`102 1 6
`
`0.019 1 0
`
`CSF
`concentration
`(ng ml")
`0.33 1 0.34
`0.74 1 0.07
`0.17 1 0.04
`10.9 1 1.7
`1.07 1 0.17
`0.71 1 0.26
`0.38 1 0.25
`27.9 1 NA
`
`0.72 1 0.02
`0.72 1 0.03
`0.074 1 0.006
`0.13 1 0.02
`0.32 1 0.02
`0.019 1 0.001
`0.96 1 0.06
`0.83 1 0.04
`
`0.19 1 0.02
`0.16 1 0.01
`
`0.012 1 0.001
`0.025 1 0.005
`0.01 1 0
`0.40 1 0.06
`0.46 1 0.02
`
`Values in the table are mean 1 SD for three separate determinations. fm, unbound fraction in brain; fup, unbound fraction in plasma.
`
`Table 3
`In vivo CNS penetration of various antimuscarinic agents in rats following subcutaneous administration
`
`:
`
`Brain : plasma concentration
`ratio
`
`CSF : free plasma
`
`concentration ratio
`
`Compound
`
`‘lrospiutn
`5-HMT
`Darifenadn
`solifenacin
`Tolterodine
`Oxybutynin
`N-Inethylscopolamine
`Scopolamine
`
`Definition of terms: fm, unbound fraction in brain; fup, unbound fraction in plasma; Kp_free_ ratio of unbound concentrations in brain : plasma.
`
`comparator compounds that display significant (positive
`control) or marginal (negative control) CNS effects, respec-
`tively, at similar doses in mice [29] and in humans [30].
`Scopolamine, oxybutynin and solifenacin displayed B:P
`and CSF : free plasma concentration ratios greater than 1,
`consistent with significant CNS penetration and equilib-
`rium between CSF and free p|asma.Tolterodine had a B : P
`>1 (2.95) and CSF :free plasma of 0.1 6, consistent with sig-
`nificant CNS penetration but with CSF not in equilibrium
`with free plasma under these experimental conditions.
`5-HMT, darifenacin and trospium showed results that were
`similar to N-methy|scop|amine,with B : P in the range 0.03-
`0.16 and CSF:free plasma of 0.004—0.06, consistent with
`no significant CNS penetration.
`
`In vitro plasma and brain binding data and
`estimation of Kp, free
`The binding of compounds in rat brain tissue homogenate
`and plasma were determined in vitro and data expressed
`as unbound fraction in brain (f...,) and plasma (f.,,,).The ratio
`of f.,,, : f.,., was calculated in order to estimate the distribu-
`tion of compounds between brain and plasma assuming
`CNS penetration is due to passive diffusion [31]. 5—HMT,
`darifenacin, oxybutynin and trospium displayed slightly
`
`240 / 72:2 / Br]C|in Phannacol
`
`higher unbound fractions in plasma than in brain (f.,,,:f.,.,
`ratios
`1.9-4.5), whereas
`tolterodine and solifenacin
`
`showed even higher f.,,,:f,,;, ratios of 13 and 11, respec-
`tively. Scopolamine and N-methylscopolamine were found
`to have similar f.,,,: fur, ratios of 1.8 and 2.4, respectively.
`Assuming CNS distribution is due to passive permeability,
`the in vitro binding results would predict that all the anti-
`muscarinic agents have significant CNS penetration (f.,,, : f...,
`>1).
`
`The in vitro brain binding data and observed B : P ratio
`were used to calculate the unbound brain : unbound
`
`plasma ratio (Kp,free). Kp,free ratios were compared with
`the observed CSF :free plasma ratios to assess whether
`free brain and CSF concentrations were in equilibrium with
`free peripheral concentrations in the plasma (Table 3).
`5-HMT, trospium and darifenacin could be categorized as
`having low Kp,free with values ranging from 0.01 to 0.04
`and low CSF :free plasma with values between 0.004 and
`0.06. The low ratios were similar to Kp,free and CSF :free
`plasma of N-methylscopolamine, 0.02 and 0.01, respec-
`tively. Oxybutynin and scopolamine afforded a high
`Kp,free and CSF :free plasma approaching unity. To|terod-
`ine provided intermediate Kp,free and CSF :free plasma
`(0.23 and 0.16) while solifenacin had a similar Kp,free (0.28)
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0006
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0006
`
`

`
`CNS penetration potential of OAB agents
`
`
`
`Tissueconcentration(ngml"orngg")
`
`f,,
`
`f“'
`
`
`
`Trospium 5-HMT
`
`0.72
`
`0. I 9
`
`0.12
`
`0. I 6
`
`Darlfen-
`acln
`0.074
`
`SoIifen-
`acln
`0. I 3
`
`ToItero- Oxybut- N-MS
`dine
`ynln
`0.32
`0.0 I 9
`
`0.96
`
`ScopoIa-
`mine
`0.83
`
`0.0 I 9
`
`0.0 I 2
`
`0.025
`
`0.0 I
`
`0.40
`
`0.46
`
`Figure 3
`Tissue concentrations of antimuscarinic agents following subcutaneous administration in rats. Mean concentrations of compounds in plasma, brain and CSF
`determined following subcutaneous closing to three animals are shown,where error bars represent SDs. Unbound fractions in plasma (f.,,,) and brain (f...) are
`shown underneath each compound. plasma (ng ml") E); brain (ng g“) (-);CSF (ng ml") (2)
`
`but higher CSF:free plasma of approximately 1.41. The
`results are consistent with low CNS penetration of S-HMT.
`darifenacin, N-methylscopolamine and trospium, and
`significant CNS penetration of oxybutynin, scopolamine.
`solifenacin and tolterodine. However, Kp,free values
`for solifenacin and tolterodine were significantly lower
`than unity, suggestive of some possible restriction in CNS
`penetration.
`
`Discussion
`
`The main objective of the present study was to detennine
`the CNS penetration of antimuscarinic agents used for the
`treatment of OAB. CNS penetration of drugs may be limited
`by the passive penneability properties and efflux trans-
`porters, such as P—gp, in the endothelial cells of the cere-
`brovascular capillaries that comprise the BBB. Hence, a
`secondary objective was to provide a mechanistic expla-
`nation of any apparent differences in the ability of OAB
`drugs to cross the BBB. The drugs were assessed in a rat
`model that determined their CNS penetration in vivo by
`measuring tissue concentration and binding of com-
`pounds.The in vivo results were then put into context with
`physical property assessment and results from permeabil-
`ity and P-gp substrate assays to provide a mechanistic
`interpretation of the CNS penetration results.
`
`CNS penetration of OAB agents in vivo
`In the present study, rats were administered subcutaneous
`doses of compounds and brain, plasma and CSF were
`
`sampled 1 h after dosing. As a result of theoretical pharma-
`cokinetic considerations combined with results of in vivo
`
`microdialysis experiments, it has been suggested that
`equilibration of drugs between brain and blood occurs
`very rapidly [32]. Other work that examined the time to
`reach brain equilibrium in rats dosed subcutaneously with
`a range of compounds found that compounds with a com-
`bination of low brain binding and high blood brain barrier
`permeability reached equilibrium (B: P at plateau) within
`10 min to 2 h [33]. However, tissue sampling at a single
`time point remains a potential limitation in experimental
`design because it is possible that compounds may not
`have attained steady-state distribution between brain and
`plasma within that time.The most rigorous protocol would
`employ tissue sampling over a time course,a||owing tissue
`concentration ratios to be detennined at a time point
`shown to represent steady state, and potentially over a
`range of doses. However, such a protocol is extremely time
`consuming and considerably increases the number of
`animals used to generate the data. The single dose and
`time point design employed in our study aimed to mini-
`mize animal use and provide plasma and tissue concentra-
`tions high enough to pennit their accurate measurement
`by specific LC/MS-MS assay, while avoiding saturation of
`transporter proteins that could affect tissue distribution.
`The ratio of total concentration in brain and plasma
`(B: P) is most commonly used to assess CNS penetration.
`However,
`if CNS penetration is solely detennined by
`passive penneability, steady-state B : P is dependent on the
`relative non—specific binding to plasma proteins (f..,,) and
`brain tissue (f.,.,) suggesting that the binding ratio can be a
`
`Br] Clin Pharmacol
`
`/ 72:2 / 241
`
`Patent Owner, UCB Pharma GmbH — Exhibit 2005 - 0007
`
`
`
`Patent Owner, UCB Pharma GmbH – Exhibit 2005 - 0007
`
`

`
`E. Callegari et al.
`
`good surrogate for in vivo steady state B : P [20, 31]. The
`f..,, : f..;, values obtained for the OAB agents were found to
`be in the range of 1.9 to 13.Therefore, if these compounds
`have high passive permeability and are not substrates for
`efflux transporters, these results taken alone would predict
`that all OAB agents will have significant brain penetration,
`as suggested by their free fractions in plasma relative to
`brain. However, based on tissue concentration data in rats,
`
`the OAB agents may be broadly categorized into two
`groups: one consisting of trospium, 5—HMT and darifenacin
`with no significant CNS penetration and another consist-
`ing of oxybutynin, solifenacin and tolterodine with signifi-
`cant CNS penetration. Within each group, the positive
`and the negative
`controls,
`scopolamine
`and N-
`methylscopolamine helped confirm the categorization
`as CNS—penetrant and non—penetrant, respectively. The
`observed B : P ratios for solifenacin,to|terodine and oxybu-
`tynin were >1 and similar to scopolamine, consistent with
`significant CNS penetration. For these compounds the in
`vitro f.,,, : f.., was consistent with the in vivo results since
`predictions were within ~fourfold of observed. Tissue
`binding results were also used to calculate unbound tissue
`concentrations and enable comparison of free concentra-
`tions in plasma, brain and CSF, to indicate whether equilib-
`rium between the compartments was likely to exist. As the
`concentration of protein in CSF is very low. it was assumed
`that free concentrations of drug are measured in CSF. The
`Kp,free and CSF :free plasma ratios for oxybutynin and
`scopolamine afforded ratios of approximately 1, indicating
`that equilibrium between free concentrations in the brain,
`CSF and plasma had been achieved. However, for solifena-
`cin the CSF :free plasma was approximately 1, consistent
`with equilibrium between the CSF and free plasma com-
`partment, but Kp,free was 0.28 indicating that free brain
`and free plasma concentrations were not at equilibrium.
`Tolterodine also demonstrated significant CNS penetra-
`tion, yet Kp,free (0.23) and CSF :free plasma (0.16) did not
`reach unity indicating that equilibrium between CNS
`compartments had not been achieved. These results with
`solifenacin and tolterodine indicate some limitation in CNS
`
`penetration or may reflect slow attainment of equilibrium
`across the BBB relative to the other OAB agents. However, it
`cannot be ruled out that they act as substrates for a trans-
`porter protein other than P-gp that limits free brain and
`CSF concentrations. It is also possible that differences in
`the physiology of the CSF/plasma barrier and BBB play a
`role in the difference in Kp,free and CSF :free plasma ratio
`for solifenacin. CSF is produced at the choroid plexus, a
`fenestrated endothelial cell layer lacking the tight junc-
`tions of the BBB. Additionally, there have been reports of
`different expression and localization of transporter pro-
`teins in the choroid plexus relative to the BBB.These differ-
`ences in permeability and transporter expression may lead
`to different brain tissue and CSF free concentrations and
`
`different rates in reaching equilibrium across the OAB
`class [34].
`
`242 I 72:2 / Br] Clin Pharmacol
`
`The B: P, Kp,free and CSF :free plasma ratios for tro-
`spium, 5—HMT and darifenacin were similar to those of
`N—methy|scopo|amine, which is consistent with no signifi-
`cant CNS penetration. For these compounds, the P : B ratios
`of unbound fractions (f..,,: f...,) significantly over—predicted
`thei

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