throbber
The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`Themed Issue: NIDA/AAPS Symposium on Drugs of Abuse: Mechanisms of Toxicity, Toxicokinetics and Medical Consequences,
`November 4-5, 2005
`Guest Editors - Rao S. Rapaka and Jagitsing H. Khalsa
` Vesicular Monoamine Transporter 2: Role as a Novel Target for
`Drug Development
` Submitted: June 30 , 2006 ; Accepted: July 18 , 2006; Published: November 10, 2006
` Guangrong Zheng , 1 Linda P. Dwoskin , 1 and Peter A. Crooks 1
` 1 College of Pharmacy, University of Kentucky, Department of Pharmaceutical Sciences, Lexington, KY 40536-0082
`
` A BSTRACT
` In the central nervous system, vesicular monoamine trans-
`porter 2 (VMAT2) is the only transporter that moves cyto-
`plasmic dopamine (DA) into synaptic vesicles for storage
`and subsequent exocytotic release. Pharmacologically
`enhancing DA sequestration by VMAT2, and thus prevent-
`ing the oxidation of DA in the cytoplasm, may be a strategy
`for treating diseases such as Parkinson ’ s disease. VMAT2
`may also be a novel target for the development of treat-
`ments for psychostimulant abuse. This review summarizes
`the possible role of VMAT2 as a therapeutic target, VMAT2
`ligands reported in the literature, and the structure-activity
`relationship of these ligands, including tetrabenazine ana-
`logs, ketanserin analogs, lobeline analogs, and 3-amine-2-
`phenylpropene analogs. The molecular structure of VMAT2
`and its relevance to ligand binding are briefl y discussed.
`
`intestinal tract. 5-7 VMAT2 is also expressed in at least 2
`endocrine cell populations and in neurons. 6 Both VMAT1
`and VMAT2 are more widely expressed during embryonic
`development. 8 Substrate recognition and inhibitor sensitivi-
`ties for differences between VMAT1 and VMAT2 have been
`studied using membrane vesicles prepared from stable trans-
`formed cell lines from Chinese hamster ovaries (CHO) that
`express the respective proteins. 9 VMAT2 has a consistently
`higher affi nity for all of the monoamine substrates tested,
`particularly histamine, and has a greater sensitivity than
`VMAT1 to the inhibitor tetrabenazine (TBZ).
` The natural alkaloid reserpine and TBZ are considered 2 clas-
`sical VMAT inhibitors. 10 Reserpine inhibits the transport of
`amines into chromaffi n granules and synaptic storage vesi-
`cles 11 , 12 by binding with high affi nity to VMAT, presumably at
`the amine recognition site. It has been suggested that TBZ, on
`the other hand, binds to a site on VMAT that is different from
`the substrate binding site at which reserpine interacts. 12-14
`
` K EYWORDS: vesicular monoamine transporter 2 , Parkinson ’ s
`disease , psychostimulant abuse , tetrabenazine , ketanserin ,
` lobeline
`
` INTRODUCTION
` The vesicular monoamine transporter (VMAT), a member of
`the vesicular neurotransmitter transporter family, is respon-
`sible for the translocation of monoamines (serotonin, dopa-
`mine, norepinephrine, and histamine) from the cytoplasm
`into synaptic vesicles via a proton electrochemical gradient
`generated by the vacuolar type H + -adenosine triphospha-
`tase. 1 Two pharmacologically distinct VMAT isoforms,
`VMAT1 and VMAT2, have been cloned and described. 2-4
`Adult human and rodent monoaminergic neurons of the cen-
`tral nervous system (CNS) and sympathetic postganglionic
`neurons express only VMAT2, 5-7 while VMAT1 is predomi-
`nantly expressed in neuroendocrine cells such as chromaffi n
`cells of the adrenal medulla and enterochromaffi n cells of the
`
` Corresponding Author: Peter A. Crooks, College of
`Pharmacy, University of Kentucky, 907 Rose Street, Room
`501B, Lexington, KY 40503 . Tel: (859) 257-1718 ; Fax:
` (859) 257-7585 ; E-mail: pcrooks@email.uky.edu
`
` VMAT2 AND NEUROPROTECTION
` Oxidative deamination of monoamines by monoamine oxi-
`dase is accompanied by the reduction of molecular oxygen
`to a toxic product, hydrogen peroxide. 15 Therefore, mainte-
`nance of low cytoplasmic concentrations of neurotransmit-
`ters by their reuptake into synaptic vesicles for storage is
`important to minimize their inherent toxicity. 16 Further-
`more, storage of neurotransmitters in synaptic vesicles pre-
`cludes their metabolism in the cytoplasmic compartment
`and reduces the synthetic demands on the cell. 16 In the cen-
`tral nervous system, VMAT2 is the only transporter that
`moves cytoplasmic dopamine (DA) into synaptic vesicles
`for storage and subsequent exocytotic release. 1
` Parkinson ’ s disease is a degenerative, progressive disorder
`that dramatically affects neurons of the substantia nigra and
`the basal ganglia. The etiology of Parkinson ’ s disease has
`not been elucidated, but exposure to endogenous or envi-
`ronmental toxins may contribute to the development of the
`disease. 17-21 In this regard, DA may play a role as an endog-
`enous toxin, since the normal metabolism of DA produces
`hydrogen peroxide as a byproduct, and the formation of
`DA-associated reactive oxygen species may contribute to
`E682
`
`Apotex Ex. 1003
`
`Apotex v. Auspex
`IPR2021-01507
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`the loss of nigrostriatal DA neurons. 22 Accordingly, phar-
`neurotransmitter transporters that regulate synaptic DA con-
`macologically enhancing DA sequestration by VMAT2, and
`centrations. 43-45 Recent studies have demonstrated that psy-
`thus preventing the oxidation of DA in the cytoplasm, may
`chostimulants alter VMAT2 function. 46 , 47 Cocaine inhibits
`be a strategy for treatment of Parkinson ’ s disease.
`DA transporter function, induces a rapid and reversible
`increase in vesicular DA uptake and dihydrotetrabenazine
` Exposure to the neurotoxin N -methyl-4-phenyltetrahydropyri-
`(DTBZ) binding, and causes a shift in the ratio of cytoplas-
`dine (MPTP) results in clinical symptoms closely approximat-
`mic to vesicular DA, all of which suggests that VMAT2
` N -Methyl-4-phenylpyridinium
`ing Parkinson ’ s disease. 17
`may be a novel target for the development of treatments for
`(MPP + ), the active toxic metabolite of MPTP, is a substrate for
`cocaine abuse. 48 Amphetamine and its analogs, such as
`VMAT2. 23-27 VMAT2 sequesters MPP + in synaptic vesicles
`methamphetamine, decrease vesicular DA sequestration by
`and thereby protects catecholamine-containing neurons from
`inhibiting vesicular uptake and promoting release from the
`MPP + -induced toxicity and degeneration. 3 , 28-32 CHO cells,
`vesicles. 49 , 50 Amphetamine diffuses across the vesicular
`which are normally sensitive to MPP + toxicity, because they
`membrane, decreasing the pH gradient, which results in the
`lack a plasma membrane amine transporter, can be made rela-
`loss of free energy needed for monoamine sequestration. 49-52
`tively insensitive to MPP + toxicity by transfection with VMAT
`Also, amphetamine that accumulates in the vesicles com petes
`complementary DNA. 3 In addition, when the transfected CHO
`with monoamines for protons, resulting in an increase in the
`cells are treated with reserpine, which inhibits VMAT2 func-
`diffusion of uncharged monoamines out of the vesicle. 52
`tion, the cells then become sensitive to MPP + toxicity. 3 Other
`High-dose methamphetamine treatment decreases vesicular
`studies using heterozygous VMAT2 knockout mice show that
`DA uptake and DTBZ binding, suggesting that there is a
`the knockouts are more susceptible to the neurotoxic effects of
`signifi cant alteration in VMAT2 function and localization at
`MPTP compared with the wild-type mice. 28 , 30 , 33 Furthermore,
`the vesicular membrane. 53 VMAT2 heterologous knockout
`heterozygous VMAT2 knockout mice are more sensitive to
`mice exhibit reduced amphetamine-conditioned place pref-
`methamphetamine-induced neurotoxicity and are more vul-
`erence (reward) and enhanced sensitivity to the locomotor
`nerable to the toxic effects of L-3,4-dihydroxyphenylalanine
`effects of apomorphine, ethanol, cocaine, and amphet-
`(L-DOPA, a DA precursor used to treat Parkinson ’ s disease)
`amine. 28 , 54 VMAT2 knockout studies also indicate that
`compared with wild-type mice. 34 , 35 The latter results suggest
`VMAT2 plays an important role in mediating the behavioral
`that reduction in VMAT2 activity might attenuate the effi cacy
`effects of psychostimulants. Taken together, these results
`of L-DOPA therapy in Parkinson ’ s patients. Finally, increased
`support the idea that VMAT2 should be considered as a valid
`sequestration of DA in synaptic vesicles by VMAT2 has been
`target for the development of pharmacotherapies to treat
`suggested to be protective in Parkinson ’ s disease. 36
`psychostimulant abuse. Other evidence supporting the role
` Recently, studies have suggested that pharmacological agents
`of VMAT2 in psychostimulant pharmacology is the fi nding
`that increase VMAT2 activity are neuroprotective. For exam-
`that benzoquinolizine derivatives, such as TBZ, which have
`ple, methylphenidate increases vesicular DA uptake in rats
`high affi nity for VMAT2, decrease locomotor activity and agg-
`and prevents persistent dopaminergic defi cits induced by high-
`ressiveness in monkeys 55 and decrease methamphetamine-
`dose methamphetamine administration. 37 , 38 Pramipexole, a
`induced hyperactivity in rodent animal models. 55
`DA D2/D3 agonist used as a therapy for Parkinson ’ s disease,
`increases vesicular DA uptake and protects against the loss of
`nigrostriatal DA neurons in methamphetamine-, 3-acetylpyri-
`dine-, and ischemia-induced neurotoxicity. 39-41 Additionally,
`apomorphine, a DA D2/D3 agonist used in Europe as a treat-
`ment for Parkinson ’ s disease and for impotence, increases
`vesicular DA uptake, and this mechanism has been suggested
`to be important for its associated neuroprotection. 42
` Taken together, the results of the above studies indicate that
`VMAT2 expression and function are important in counter-
`acting the neurotoxicity of MPP + and perhaps of other envi-
`ronmental and endogenous neurotoxins that play an etiologic
`role in neurodegenerative disease. 21
`
` VMAT2 LIGANDS
` TBZ and Its Analogs
` TBZ ( 1 , Figure 1 ), a benzoquinolizine compound, has been
`shown to deplete cerebral monoamines in rat brain by revers-
`ibly inhibiting VMAT2. 56 First introduced in 1956 as an anti-
`psychotic drug, 57 TBZ is currently used to treat hyperkinetic
`movement disorders, such as chorea associated with
`Huntington ’ s disease, tics in Tourette ’ s syndrome, and
`movement stereotypes in tardive dyskinesia. 58-60 The side
`effects associated with TBZ include sedation, depression,
`akathisia, and parkinsonism. 58 TBZ inhibits catecholamine
`uptake by VMAT2 with a K i of 3 nM 14 and acts as an inhibitor
`of both presynaptic and postsynaptic DA receptors in rat
` VMAT2 AND PSYCHOSTIMULANT ABUSE
`brain. 61 [ 11 C]TBZ (label on the 9- O -methyl group) has been
` Psychostimulant-induced behavioral activation and rein-
`synthesized 62 and used as an in vivo radioligand for positron
`forcement are mediated, at least in part, via interaction with
`emission tomography (PET) imaging of VMAT2. 63-66
`E683
`
`Apotex Ex. 1003
`
`

`

` Figure 1. Structures of tetrabenazine and its analogs ( 1-4 ).
`
`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`phy (HPLC). The (+)-isomer (2R,3R,11bR, 3a ) 74 shows
`high affi nity in vitro ( K i = 0.97 nM) for rat VMAT2, whereas
`the ( – )-isomer shows very low affi nity for VMAT2 ( K i = 2.2
`μM). Thus the binding of a -DTBZ to VMAT2 is enantiose-
`lective, with the (+)-isomer having higher affi nity. 75 , 76
` Another 4 possible DTBZ isomers (2S,3S,11bR, 3e ; 2R,3R,11bS,
` 3f ; 2R,3S,11bR, 3g ; 2S,3R,11bS, 3h , Figure 3 ) have been synthe-
`sized and tested for inhibition of VMAT2 binding using rat vesicu-
`lar membranes. Isomer 3g showed the highest affi nity ( K i = 28
`nM) in the [ 3 H]DTBZ binding assay. 77 , 78
` Methoxytetrabenazine (MTBZ) ( 4 , Figure 1 ) is another
`TBZ analog with high affi nity ( K d = 3.9 nM) for VMAT2. 79
`Similar to DTBZ, [ 3 H] and [ 11 C]MTBZ have also been syn-
`thesized 73 and used in in vitro and in vivo studies. 79-81
` Nucleophilic addition of organometallic reagents to the C-2
`keto group of TBZ generated a series of 2-alkylated DTBZ
`analogs, such as the 2-Me, 2-Et, 2-Pr, 2-iso-Pr, and 2-iso-Bu
`derivatives (all racemic mixtures, Figure 4 ). 82-85 These com-
`pounds have been evaluated for inhibition of [ 3 H]MTBZ
`binding to VMAT2 in rat striatum. 85 The b -methyl com-
`pound 5a showed the highest affi nity ( K i = 2.6 nM) in this
`series, with a nearly 5-fold higher affi nity than its diastereo-
`mer 5b ( K i = 12 nM), which is consistent with the fi nding
`that a -DTBZ exhibits higher affi nity for VMAT2 than does
` b -DTBZ. 73 Compound 5b and compounds 6 to 9 all contain
`a b -hydroxyl group and showed a general decrease in bind-
`ing affi nity upon either lengthening or branching of the
`alkyl group at C-2. 85 These results indicate that analogs
`containing considerable steric bulk at position 2 can be tol-
`erated. Thus, compound 10 ( Figure 4 ), in which an 125 I atom
`has been introduced for autoradiographic studies of VMAT2,
`has been synthesized. 86 ( ± )-Compound 10 can be separated
`by chiral HPLC into its optical isomers, and the fi rst eluted
`enantiomer binds to VMAT2 with a K d of 0.22 nM. 87
` Structure-activity relationship (SAR) studies involving TBZ
`analogs have shown that quaternization of the amine nitro-
`gen at position 5, aromatization of ring C, and elimination
`of the carbonyl group afforded compounds that were devoid
`
` TBZ analogs have been synthesized with different alkyl
`groups at the C-3 position in the molecule, such as com-
`pound Ro 4-1632 ( 2 , Figure 1 ). These analogs retain good
`amine-depleting activity. 55
` In vivo, TBZ is rapidly and extensively metabolized to its
`reduced form, DTBZ ( 3 , Figure 1 ). 67 [ 3 H]DTBZ (label on
`the C-2 hydrogen) has been used as a selective radioligand
`in in vitro brain homogenate binding studies and in autora-
`diographic studies, and is reported to have a K d value of 3.0
`nM. 13 , 14 , 68-70 [ 11 C]DTBZ (label on the 9- O -methyl group)
`has also been synthesized 71 and used for in vivo PET imag-
`ing of VMAT2. 66 , 72
` TBZ contains 2 chiral carbon centers at C-3 and C-11b;
`thus, theoretically, TBZ can exist as 4 possible stereoiso-
`mers (3R,11bR; 3S,11bS; 3R,11bS; and 3S,11bR). TBZ
`usually refers to the racemic compound, that is, a 1:1 mix-
`ture of the 3R,11bR and 3S,11bS isomers. Synthetic DTBZ,
`the product of hydride reduction of the 2-keto group of TBZ,
`can exist in 2 a -DTBZ forms (2R,3R,11bR, 3a ; and
`2S,3S,11bS, 3b , Figure 2 ) and 2 b -DTBZ forms (2S,3R,11bR,
` 3c ; and 2R,3S,11bS, 3d , Figure 2 ). a -DTBZ and b -DTBZ
`can be separated by column chromatography, and the
` a -DTBZ isomer ( K i = 6 nM) shows slightly higher binding
`affi nity in vitro for rat brain VMAT2 than does b -DTBZ ( K i =
`20 nM). 73 The 2 enantiomers of a -DTBZ have been sepa-
`rated using chiral High Performance Liquid Chromatogra-
`
` Figure 2. Stereoisomers of dihydrotetrabenazine ( 3a-d ).
`
` Figure 3. Stereoisomers of dihydrotetrabenazine ( 3e-h ).
`E684
`
`Apotex Ex. 1003
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`
` Figure 4. Structures of tetrabenazine analogs ( 5a-b and 6-10 ).
`
`of monoamine-depleting activity. 55 , 88 Thus, a basic amine
`nitrogen at position 5 is a prerequisite for TBZ-like activ-
`ity. 89 Also, methoxy groups at positions 9 and 10 appear to
`be essential for TBZ-like activity; the methylenedioxy com-
`pound 11 ( Figure 5 ) was 3 orders of magnitude less potent
`than Ro 4-1284 ( 6 ). 90
` Replacing the carbonyl oxygen in TBZ with a bis -methylthio
`group (compound 12 , Figure 5 ) affords a compound with
`similar activity to TBZ. 91 Olefi nation of the carbonyl group
`to afford compound 13 ( Figure 5 ) (EC 50 = 14 nM) resulted
`in potent inhibition of [ 3 H]DTBZ binding. 92
` Based upon a limited number of TBZ analogs ( 14 - 17 , Fig-
`ure 6 ), a correlation between the lipophilicity of the analogs
`and their affi nity for the DTBZ binding site has been estab-
`lished. 93 Compounds shown to have higher partition coeffi -
`cients (octanol/buffer) generally exhibited a greater ability
`to inhibit the specifi c binding of [ 3 H]DTBZ (IC 50 = 6 nM
`for 14 , 47 nM for 17 , 110 nM for 16 , and 2500 nM for 15 )
`to VMAT2. 93 Accordingly, compound 20 ( Figure 6 ), an
`iodinated and photosensitive derivative of TBZ, has been
`synthesized and exhibited an IC 50 of 428 nM to inhibit
`[ 3 H]DTBZ binding. 92 However, both its precursor (com-
`pound 18 , IC 50 = 8.1 nM) and the non-iodinated analog ( 19 ,
`IC 50 = 53 nM) of compound 20 showed higher affi nity at
`VMAT2 than did compound 20 . 92
` Several derivatives of compound 16 (ie, compounds 21 - 24 ,
` Figure 7 ) have been synthesized; of these, the amino com-
`pounds 21 and 22 retained affi nity for VMAT2 ( K i = 7.6 nM
`and 72.2 nM, respectively, in the [ 125 I]iodovinyl-TBZ bind-
`ing assay), whereas the amido compounds 23 and 24 exhib-
`ited diminished affi nity for VMAT2 ( K i = 730 nM and >10
`000 nM, respectively, in the [ 125 I]iodovinyl-TBZ binding
`assay). 94
`
` Ketanserin and Its Analogs
` Ketanserin ( 25 , Figure 8 ), a well-known serotonin 5-HT2
`receptor antagonist, 95 also binds to VMAT on chromaffi n
`
` Figure 6. Structures of tetrabenazine analogs ( 14-20 ).
`
`granules and synaptic vesicles. 96-98 In the studies by Darchen
`et al, 96 Henry et al, 97 and Leysen et al, 98 ketanserin competi-
`tively inhibited the binding of [ 3 H]DTBZ to VMAT2, and
`[ 3 H]ketanserin binding.
`conversely, TBZ displaced
`[ 3 H]Ketanserin binds to the TBZ binding site with a K d of
`45 nM at 30ºC and a K d of 6 nM at 0ºC. 96
` A ketanserin derivative, 7-azidoketanserin ( 26 , Figure 8 ),
`also binds to the TBZ binding site of bovine chromaffi n
`granule membranes with a K i of 23 nM (inhibition of
`[ 3 H]DTBZ binding). 99 An iodinated azido derivative of ket-
`anserin, 7-azido-8-iodoketanserin ( 27 , Figure 8 ), binds to
`the same specifi c TBZ binding site as ketanserin with a K d
`of 5.5 nM at 0ºC 99 ; 7-azido-8-[ 125 I]iodoketanserin has been
`successfully used for photoaffi nity labeling of TBZ binding
`sites of different tissues, including rat striatum, rabbit
` platelets, human pheochromocytoma, and human adrenal
`medulla. 99
` Lengthening the distance between the piperidine and the
`benzoyleneurea moieties of the ketanserin molecule by
`addition of 2 methylene groups results in a compound ( 28 ,
` Figure 9 ) that exhibits a 20-fold decrease in affi nity ( K i =
`950 nM) for the [ 3 H]DTBZ binding site. 96 Reducing the
`keto group of ketanserin (compound 29 , Figure 9 ) also
`decreases affi nity ( K i = 350 nM) for this site. Additionally,
`replacing the benzoyleneurea moiety with other heterocy-
`cles (eg, compounds 30-32 , Figure 9 ) also decreases affi nity
`( K i = 950, 814, and 3600 nM, respectively) for the [ 3 H]DTBZ
`binding site. However, minor structural changes to the
`
` Figure 5. Structures of tetrabenazine analogs ( 11-13 ).
`
` Figure 7. Structures of tetrabenazine analogs ( 21-24 ).
`E685
`
`Apotex Ex. 1003
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`
` Figure 10. Structure of lobeline ( 35 ).
`
` Figure 8. Structures of ketanserin and its analogs ( 25-27 ).
`
`neurochemical studies, suggesting that they do not act via a
`common mechanism. Nevertheless, lobeline has often been
`considered to be a nicotinic receptor agonist. Conversely,
`we and others have established that lobeline acts as a potent,
`but nonselective, nicotinic receptor antagonist. 104 , 114-117
`Lobeline inhibits nicotine-evoked [ 3 H]DA overfl ow from
`rat striatal slices with an IC 50 of 1 μM, suggesting that lobe-
`line acts as an antagonist at nicotinic receptors mediating
`nicotine-evoked DA release (ie, a 6 b 2 b 3* subtype). 116 Lobeline
`also inhibits nicotine-evoked 86 Rb + effl ux from rat thalamic
`synaptosomes with an IC 50 of 0.7 μM, indicating that lobe-
`line is also an antagonist at a 4 b 2* nicotinic receptors. 116
`Moreover, lobeline also inhibits [ 3 H]methyllycaconitine
`binding to rat brain membranes with a K i of 6.26 μM, indi-
`cating that there is an interaction with the a 7* nicotinic
`receptor subtype. 117 Lobeline has also been reported to
`be an antagonist (IC 50 of 8.5 μM) at human a 7* nicotinic
`receptors expressed in Xenopus oocytes. 118
` In addition to interacting with nicotinic acetylcholine recep-
`tors (nAChRs), lobeline inhibits [ 3 H]DTBZ binding to
`VMAT2 with an IC 50 of 0.90 μM and inhibits [ 3 H]DA
`uptake into rat striatal vesicle preparations with an IC 50 of
`0.88 μM. 119 , 120 Therefore, lobeline is a nonselective nAChR
`antagonist that also inhibits VMAT2 function. Importantly,
`lobeline has been shown to inhibit both the neurochemical
`and the behavioral effects of amphetamine in rodents. 111 , 121-123
`The mechanism underlying the lobeline-induced inhibition
`of these effects has been suggested to be noncompetitive
`inhibition of VMAT2 function. 114 The observation that lobeline
`is not self-administered is consistent with fi ndings that lobe-
`line does not evoke DA release. 111 , 114 , 119 Furthermore, the
`observation that lobeline inhibits methamphetamine-evoked
`DA release from superfused rat striatal slices 116 is consis-
`tent with its ability to decrease methamphetamine self-
`administration in rats. 123 These studies clearly implicate
`VMAT2 as a potential target for the development of
`agents to treat methamphetamine abuse. Regardless,
`lobeline is a novel prototypical molecule from which
`subtype-selective nAChR ligands and selective VMAT2
`inhibitors may be developed following appropriate struc-
`tural modifi cation.
` Systematic structural modifi cation of the lobeline molecule
`provided 2 non-oxygen-containing
`lobeline analogs:
` N -methyl-2,6-di-( cis -phenylethenyl)piperidine ( meso -trans-
`diene [MTD], 36a , Figure 11 ) and N -methyl-2,6-di-( cis -
`phenylethyl)piperidine (lobelane, 37a , Figure 11 ). The latter
`E686
`
`benzoyleneurea moiety, such as introducing a hydroxyl
`group into the ring (compound 33 , Figure 9 ) or replacing 1 of
`the oxygen atoms with a sulfur atom (compound 34 , Figure
`9 ), retains the affi nity ( K i = 14 and 40 nM, respectively). 96
`
` Lobeline and Its Analogs
` A lipophilic alkaloid from Lobelia infl ata , ( – )-lobeline
`(lobeline, 2R,6S,10S-, 35 , Figure 10 ), displaces [ 3 H]nicotine
`binding from native nicotinic receptors in the CNS with
`high affi nity ( K i = 4-30 nM). 100-104 Although lobeline has
`no structural resemblance to nicotine, and SARs do not sug-
`gest a common pharmacophore, 105 it has many nicotinelike
`effects, such as tachycardia and hypertension, 106 brady-
`cardia and hypotension in anesthetized rats, 107 anxiolytic
`activity, 108 and improvement of learning and memory. 109 In
`contrast to nicotine, lobeline only marginally supports self-
`administration in mice 110 and does not support self-admin-
`istration in rats. 111 Additionally, chronic lobeline treatment
`does not increase locomotor activity in rats and does not
`produce conditioned place preference. 112 , 113 Thus, lobe-
`line and nicotine have different effects in behavioral and
`
` Figure 9. Structures of ketanserin analogs ( 28-34 ).
`
`Apotex Ex. 1003
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`
` Figure 11. Structures of lobeline analogs ( 36a-c and 37a-c ).
`
`2 analogs showed affi nity for VMAT2 at the TBZ binding
`site ( K i of 9.88 μM for 36a , and 0.97 μM for 37a ), with
`negligible affi nity for the ligand binding sites on a 4 b 2* and
` a 7* nAChRs. 117 , 124 Compounds 36b and 36c ( Figure 11 )
`are 2 stereoisomers of 36a ; 36c was equipotent with its
` meso -isomer, MTD, but 36b was slightly less potent (2- to
`3-fold) than MTD at VMAT2. Within the lobelane series of
`compounds (ie, compounds 37a , 37b , and 37c , Figure 11 ), a
`change in C2, C6 stereochemistry from cis to trans afforded
`a modest reduction (5- to 6-fold) in affi nity at VMAT2. The
` trans enantiomers 37b and 37c exhibited comparable affi ni-
`ties at VMAT2. These data indicate that the VMAT2 bind-
`ing site is not sensitive to major stereochemical changes to
`the MTD and lobelane molecules at the C2 and C6 piperid-
`ino ring carbons.
` Interestingly, 2 conformationally fl exible, ring-opened com-
`pounds, 38a and 38b ( Figure 12 ) ( K i = 5.21 and 3.96 μM,
`respectively) and 2 acyclic compounds, 39 and 40 ( Figure
`12 ) ( K i = 2.37 and 3.07 μM, respectively), exhibited lower,
`but comparable, affi nity for VMAT2 compared with lobel-
`ane. Thus, ring opening or complete removal of the piperi-
`dine ring results in only a modest reduction in affi nity at
`VMAT2 compared with lobelane ( 37a ). The presence of a
`basic amine functionality is likely a prerequisite for VMAT2
`recognition, as is evidenced by the fact that quaternized
`compounds 41 ( K i > 100 μM) and 42 ( K i = 16.5 μM) ( Fig-
`ure 12 ) show signifi cant loss in affi nity for VMAT2. 124
` The entire lobelane structure appears to be critical for high-
`affi nity binding at VMAT2, since fragments of lobelane or
`MTD, such as compounds 43 and 44 ( Figure 13 ) (both K i >
`100 μM), exhibited no affi nity for VMAT2. 125
`
` Figure 13. Structures of lobeline analogs ( 43 and 44 ).
`
` Isomerized lobelane analogs, such as compound 45 ( Figure
`14 ) ( K i = 1.36 m M), retained affi nity for VMAT2, indicating
`that the position of the piperidine N atom relative to the C2
`and C6 side chains does not appear to be critical for VMAT2
`interaction, and that the VMAT2 binding site can tolerate
`changes in distance between the piperidine nitrogen and the
`2 phenyl rings. 125 In the lobelane structure, changes in the
` N -substituent can also be tolerated. Nor -lobelane ( 46 , K i =
`2.31 m M), nor - N -ethyl lobelane ( 47 , K i = 3.41 m M), and
` nor - N -n-propyl lobelane ( 48 , K i = 1.87 m M) ( Figure 14 )
`exhibit only a slight decrease in affi nity for VMAT2 com-
`pared with lobelane. 125 Replacing the phenyl rings of lobel-
`ane with naphthalene rings (compound 49 , K i = 0.63 m M) or
`introducing substituents into the phenyl rings (eg, in com-
`pounds 50 , K i = 0.57 m M; 51 , K i = 0.43 m M; and 52 , K i =
`0.52 m M) ( Figure 14 ) retains or somewhat improves affi nity
`at VMAT2.
` To increase the rigidity of the lobelane molecule, analogs
`were prepared in which the piperidine ring has been replaced
`with a tropene ring. The resulting compounds ( 53 , K i = 1.30
` m M; 54 , K i = 1.38 m M; and 55 , K i = 4.80 m M) ( Figure 15 )
`exhibited affi nity at VMAT2 comparable with lobelane. 126
`
` 3-Amino-2-Phenylpropene Derivatives
` Recently, a series of 3-amino-2-phenylpropene derivatives
`( Figure 16 ) have been reported as novel competitive inhibi-
`tors of the bovine chromaffi n granule membrane mono-
`amine transporter (bVMAT2). 127 With a K i of 40.3 m M,
`3-amino-2-phenylpropene (APP, 56 ) inhibits DA uptake
`into bVMAT2. Introduction of a hydroxyl group into the 3 or 4
`
` Figure 12. Structures of lobeline analogs ( 38a-b and 39-42 ).
`
` Figure 14. Structures of lobeline analogs ( 45-52 ).
`E687
`
`Apotex Ex. 1003
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`to form an ion pair and appear to provide a structural frame-
`work for substrate recognition. 129 Experiments employing a
`chimera of VMAT1 and VMAT2 indicate that 2 domains,
`TMD5 through TMD8 and TMD9 through TMD12, cooper-
`ate to confer the high-affi nity interaction of VMAT2 with
`TBZ and histamine. 130 In addition, the domain encompass-
`ing TMD3 and TMD4 infl uences serotonin affi nity but not
`histamine affi nity or TBZ sensitivity. 130 The domain encom-
`passing TMD5 through TMD7 of VMAT2 in the context of
`N-terminal VMAT2 sequences reduces the apparent affi nity
`for serotonin but not histamine or the sensitivity to TBZ. 130
`Tyrosine 434 and aspartate 461 in TMD9 through TMD12
`are identifi ed as being responsible for the high-affi nity inter-
`action of TBZ, histamine, and serotonin, but not for DA. 131
`Photoaffi nity labeling of purifi ed rat VMAT2 indicates that
`TMD1 and TMD10/11 are possibly juxtaposed and may
`interact in a functionally signifi cant manner. 132 Cysteine
`mutagenesis and derivatization of human VMAT2 revealed
`that cysteines 439, 476, and/or 497, and possibly cysteines
`126 and/or 333, are important for [ 3 H]TBZOH binding, and
`cysteines 176, 207, and 439 together are important for
`[ 3 H]serotonin transport. 133 Furthermore, a disulfi de bond
`between lumenal cysteine 126 in loop 1/2 and cysteine 333
`in loop 7/8 has been identifi ed. 134
`
` Figure 15. Structures of lobeline analogs ( 53-55 ).
`
`position of APP affords compounds 57 ( K i = 16.7 m M) and
` 58 ( K i = 15.5 m M), respectively, equally improved potency
`for bVMAT2. However, compound 59 ( K i = 103 m M), which
`has a methoxyl group at the 4 position of the phenyl ring of
`APP, led to a decrease in potency. However, compound 60 ,
`in which a methoxyl group is at the 3 position of the phenyl
`ring, led to a slight improvement in inhibitory potency with
`respect to APP. Methyl ( 61 , K i = 55.9 m M) or fl uoro ( 62 , K i =
`42.3 m M) substitution at the 4 position of the phenyl ring
`had no effect on the inhibitory potency, while chloro ( 63 ),
`bromo ( 64 ), and iodo ( 65 ) substitution led to a modest
`increase in inhibitory potency ( K i = 18.0, 17.7, and 12.9
` m M, respectively).
`
` VMAT2 STRUCTURE AND MOLECULAR BASIS
`FOR BINDING
` Predictions regarding the molecular structure of VMAT2
`from its protein sequence are that it comprises 12 putative
`transmembrane domains (TMDs) with both N- and C- ter-
`mini in the cytoplasm and a large, hydrophobic, N -glycosyl-
`ated loop between TMDs 1 and 2 facing the vesicle lumen. 1
`Structural biology studies have identifi ed important resi-
`dues that may contribute to ligand binding and monoamine
`transport. Mutagenesis studies indicate that aspartate 33,
`which contains a negative charge, in TMD1 and serines 180
`to 182 in TMD3 of VMAT2 play a critical role in substrate
`recognition, presumably by interacting with the protonated
`amino group of the ligand and hydroxyl groups on the cate-
`chol or indole ring, respectively. 128 In addition, lysine 139
`in TMD2 and aspartate 427 in TMD11 of VMAT2 interact
`
` CONCLUSION
` Signifi cant progress has been made over the last 20 years in
`elucidating the role of VMAT2 in monoamine transport and
`its potential as a therapeutic target. VMAT2 sequesters cyto-
`plasmic DA and thus prevents the oxidation of DA in the
`cytoplasm; VMAT2 also sequesters neurotoxins within ves-
`icles. These data indicate that VMAT2 may play a role in
`neuroprotection and that molecules that interact with
`VMAT2 may have value as treatments for diseases such as
`Parkinson ’ s disease. VMAT2 may also be a novel target for
`the development of treatments for psychostimulant abuse,
`and the discovery of molecules that modulate VMAT2 func-
`tion may afford useful tools for examining the pivotal role
`of this transporter in the neurochemical and behavioral
`effects of psychostimulant drugs, thus providing potential
`pharmacotherapies.
`
` ACKNOWLEDGMENTS
` This study was supported by National Institutes of Health
`grant DA 13519.
`
` Figure 16. Structures of 3-amino-2-phenylpropene and its
`analogs ( 56-65 ).
`
` REFERENCES
` 1 . Yelin R , Schuldiner S . Vesicular neurotransmitter transporters:
`pharmacology, biochemistry, and molecular analysis. In: Reith MEA, ed.
` Neurotransmitter Transporters; Structure, Function, and Regulation.
`
` 2nd. Totowa, NJ : Humana Press ; 2002 :313- 354.
`E688
`
`Apotex Ex. 1003
`
`

`

`The AAPS Journal 2006; 8 (4) Article 78 (http://www.aapsj.org).
`
` 2 . Erickson JD , Eiden LE , Hoffman BJ . Expression cl

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