throbber
USOO8524733B2
`
`(12) United States Patent
`Gant et al.
`
`(10) Patent No.:
`(45) Date of Patent:
`
`US 8,524,733 B2
`Sep. 3, 2013
`
`(54) BENZOQUINOLINE INHIBITORS OF
`VESCULARMONOAMINE TRANSPORTER 2
`
`W
`WO
`
`38E. A:
`2011 106248 A3
`
`658.
`9, 2011
`
`(75) Inventors: Thomas G. Gant, Carlsbad, CA (US);
`Manoucherhr M. Shahbaz, Escondido,
`CA (US)
`(73) Assignee: Auspex Pharmaceuticals, La Jolla, CA
`(US)
`Subject to any disclaimer, the term of this
`patent is extended or adjusted under 35
`U.S.C. 154(b) by 555 days.
`(21) Appl. No.: 12/562,621
`(22) Filed:
`Sep. 18, 2009
`
`(*) Notice:
`
`(65)
`
`Prior Publication Data
`US 2010/O 130480 A1
`May 27, 2010
`
`Related U.S. Application Data
`(60) Provisional application No. 61/097.896, filed on Sep.
`18, 2008.
`
`(2006.01)
`(2006.01)
`
`(51) Int. Cl.
`CO7D 22/06
`A6 IK3I/4353
`(52) U.S. Cl.
`USPC .............................. 514/294; 546/79:514/290
`(58) Field of Classification Search
`USPC ............................................ 546/79; 514/290
`See application file for complete search history.
`
`(56)
`
`References Cited
`
`U.S. PATENT DOCUMENTS
`2,830,993 A
`4, 1958 Brossi
`3,045,021 A
`7, 1962 Brossi
`4,316,897 A
`2f1982 Lotz
`6,221,335 B1
`4/2001 Foster
`6,440,710 B1
`8/2002 Keinan et al.
`6,603,008 B1
`8, 2003 Ando et al.
`7,517,990 B2
`4/2009 Ito et al.
`2002fOO13372 A1
`1/2002 Ekins
`8/2007 Potyen et al.
`2007/O197695 A1
`2/2008 Tung
`2008, OO33011 A1
`2010.0113496 A1
`5, 2010 Gant et al.
`2010, O189698 A1
`7, 2010 Willis
`2010/0204258 A1
`8, 2010 Harris et al.
`2012,0003330 A1
`1/2012 Gant et al.
`
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`WO
`
`FOREIGN PATENT DOCUMENTS
`9526325 A2 10, 1995
`2005077946 A1
`8, 2005
`2006053067 A2
`5, 2006
`2006078846 A1
`T 2006
`2007 1303.65 A2 11/2007
`2008058261 A1
`5, 2008
`2008064274 A1
`5, 2008
`2008112278 A2
`9, 2008
`2009003226 A1
`1, 2009
`2009 124357 A1 10, 2009
`2010018408 A2
`2, 2010
`2010044981 A2
`4/2010
`2010044981 A3
`6, 2010
`
`OTHER PUBLICATIONS
`Kushner DJ et al. Pharmaceutical uses and perspectives of heavy
`water and deuterated compounds, 1999.*
`Kenney C and Jankovik J. Tetrabenazine in the treatment of
`hyperkinetic movement disorders, 2006.*
`Alan Foster, Deuterium isotop effects in Studies of drug metabolism.
`1984.
`Helfenbein et al. Isotopic effect study of Propofol Deuteration on
`metabolism, Activity and Toxicity of the anesthetic. 2002.*
`Bauer, LA et. al., Olnfluence of long-term infusions on lidocaine
`kinetics, Clin. Pharmacol. Ther. 1982, 433-7.
`Borgstrom, Letal.; Comparative Pharmacokinetics of Unlabeled and
`Deuterium-Labeled Terbutaline: Demonstration of a Small Isotope
`Effect, J Pharm Sci, 1988, 77(11),952-4.
`Browne, T.R.; Chapter 2. Isotope Effect: Implications for Pharma
`ceutical Investigations, Pharm Lib 1997 13.
`Browne, T.R. et al.; Pharmacokinetic Equivalence of Stable-Isotope
`Labeled and Unlabeled Drugs. Phenobarbital in Man, J. Clin
`Pharmacol, 1982, 22, 309-315.
`Burm, AGL et al., Pharmacokinetics of Lidocaine and bupivacaine
`and stable isotope-labeled analogs: a study in healthy volunteers,
`Biopharmaceutics and Drug Disposition, 1988, 9, 85-95.
`Elison, C et al.:Effect of Deuteration of N-CHS (3)S Group on
`Potency and Enzymatic N-Demethylation of Morphine, Science,
`1961, 134(3485), 1078-9.
`Farmer, PB, et al.:Synthesis, Metabolism, and Antitumor Activity of
`Deuterated Analogues of 1-(2-Choloroethyl)-3-cyclohexyl-1-
`nitrosourea, Journal of Medicinal Chemistry, 1978, vol. 21, No. 6,
`514-20.
`Fisher, MB, et al.:The complexities inherent in attempts to decrease
`drug clearance by blocking sites of CYP-mediated metabolism, Curr
`Opin Drug Discov Develop; 2006, 9(1), 101-9.
`(Continued)
`Primary Examiner — Rita Desai
`(74) Attorney, Agent, or Firm — Dennis A. Bennett; Mike
`Sertic
`
`ABSTRACT
`(57)
`The present invention relates to new benzoquinoline inhibi
`tors of vesicular monoamine transporter 2 (VMAT2), phar
`maceutical compositions thereof, and methods of use thereof.
`
`
`
`3 Claims, No Drawings
`
`Apotex Ex. 1001
`
`Apotex v. Auspex
`IPR2021-01507
`
`

`

`US 8,524.733 B2
`Page 2
`
`(56)
`
`References Cited
`
`OTHER PUBLICATIONS
`Foster, AB; Deuterium Isotope Effects in Studies of Drug Metabo
`lism, Trends in Pharmacological Sciences, Dec. 1984. 524-7.
`Helfenbein, Jet al.; Isotopic Effect Study of Propofol Deuteration on
`the Metabolism, Activity, and Toxicity of the Anesthetic, J. Med.
`Chem. 2002, 45,5806-5808.
`Kushner, DJ et al., Pharmacological uses and perspectives of heavy
`water and deuterated compounds, CanJPhys Pharm 1999, 77, 79-88.
`Lee, Hetal.; Deuterium Magic Angle Spinning Studies of Substrates
`Bound to Cytochrome P450, Biochemistry 1999, 38, 10808-10813.
`Mamada, Ket al., Pharmacokinetic Equivalence of Deuterium-La
`beled and Unlabeled Phenytoin, Drug Metabolism and Disposition,
`1986, 14(4), 509-11.
`Nelson, SD et al.:The Use of Deuterium Isotope Effect to Probe the
`Active Site Properties, Mechanism of Cytochrome P450-catalyzed
`Reactions, and Mechanisms of Metabolically Dependent Toxicity,
`Drug Metabolism and Disposition 31: 1481-1498, 2003.
`Nelson, SD et al.; Primary and B-Secondary Deuterium Isotope
`Effects in N-Deethylation Reactions, Journal of Medicinal Chemis
`try, 1975, vol. 18, No. 11.
`Pohl. LRet al.; Determination of toxic Pathways of Metabolism by
`Deuterium Substitution, Drug Metabolism Rev 1985 1335.
`Rampe, D et al., Deuterated Analogs of Verapamil and nifedipine.
`Synthesis and biological activity, Eur J Med Chem (1993) 28,259
`263.
`Toronto Research Chemicals, Inc.; Tetrabenazine-d7. http://www.
`trc-canada.comidetails.php?CatNumber=T284.002.
`DaSilva, JN et al.; Synthesis of 11CITetrabenazine, a Vesicular
`Monoamine Uptake Inhibitor, for PET Imaging Studies, Appl.
`Radiat, Isot, vol. 44, No. 4, pp. 673-676, 1993.
`Kilbourn, MR et al.; Absolute Configuration of (+)-a-
`Dihydrotetrabenazine, an Active Metabolite of Tetrabenazine,
`Chirality 9:59-62 (1997).
`Mehvar, R. et al...Pharmacokinetics of Tetrabenazine and Its Major
`Metabolite in Man and Rat Bloavailability and Dose Dependency
`Studies, Drug Met Disp. 1987, 15(2), 250-255.
`Paleacu, D et al.; Tetrabenazine Treatment in Movement Disorders,
`Clin Neuropharmacol 2004:27:230-233.
`Popp, FD et al.; Synthesis of potential antineoplastic agents XXVI:
`1.3.4.6.7.11b-hexahydro-9,10-dimethoxy-2H-benzo
`a 2-quinolizinone derivatives, Journal of Pharmaceutical Sciences,
`1978, 67(6), 871-873.
`Roberts, MS et al.; The Pharmacokinetics of Tetrabenazine and its
`Hydroxy Metabolite in Patients Treated for Involuntary Movement
`Disorders, Eur J. Clin Pharmacol (1986) 29: 703-708.
`Schwartz, DE et al.; Metabolic studies of tetrabenazine, a
`psychotropic drug in animals and manBiochemical Pharmacology,
`1966, 15, 645-655.
`Zheng, Get al.; Vesicular Monoamine Transporter 2: Role as a Novel
`Target for Drug Development, The AAPS Journal 2006; 8(4) Article
`78.
`
`Baillie, Thomas; The Use of Stable Isotopes in Pharmaceutical
`Research, Pharmacological Reviews, 1981, 33(2), 81-132.
`Browne, Thomas; Stable Isotope Techniques in Early Drug Devel
`opment: An Economic Evaluation, J. Clin. Pharmacol., 1998, 38.
`213-220.
`Cherrah et al.; Study of Deuterium Isotope Effects on Protein Binding
`by Gas Chromatography/Mass Spectrometry. Caffeine and Deuter
`ated Isomers, Biomedical and Environmental Mass Spectrometry,
`1987, 14,653-657.
`Dyck et al.; Effects of Deuterium Substitution on the Catabolism of
`Beta-Phenethylamine: An in Vivo Study . J. Neurochem., 1986,
`46(2), 399-404.
`Gouyette, Alain: Use of Deuterium-Labelled Elliptinium and Its Use
`in Metabolic Studies, Biomedical and Environmental Mass Spec
`trometry, 1988, 15, 243-247.
`Haskins, N.J.; The Application of Stable Isotopes in Biomedical
`Research, Biomedical Mass Spectrometry, 1982, 9(7), 269-277.
`Wolen et al.; The Application of Stable Isotopes to Studies of Drug
`Bioavailibility and Bioecuivalence, J. Clin. Pharmacol., 1986, 26.
`419-424.
`Tonn et al.; Simultaneous Analysis of Diphenylhydramine and a
`Stable Isotope Analog (2H10) Diphenylhydramine Using Capillary
`Gas Chromatography With Mass Selective Detection in Biological
`Fluids From Chronically Instrumented Pregnant Ewes, Biomedical
`Mass Spectrometry, 1993, 22, 633-642.
`Honma et al.; The Metabolism of Roxatidine Acetate Hydrochloride,
`Drug Metabolism and Disposition, 1987, 15(4), 551-559.
`Pieiaszek et al.; Moricizine Bioavailability Via Simultaneous, Dual,
`Stable Isotope Administration: Bioecuivalence Implications, J. Clin.
`Pharmacol., 1999, 39, 817-825.
`Paleacu et al...Tetrabenazine Treatment in Movement Disorders, Clin.
`Neuropharmacol., 2004, 27(5), 230-233.
`Gant et al., Benzoquinoline Inhibitors of Vesicular Monoamine
`Transporter 2, Auspex Pharmaceuticals, Inc., WO2010044981 Inter
`national Preliminary Report on Patentability, Publication Date: Apr.
`22, 2010.
`Gant et al., Benzoquinoline Inhibitors of VMAT2, Auspex Pharma
`ceuticals, Inc., WO 2011 106248 International Preliminary Report on
`Patentability, Publication Date: Sep. 1, 2011.
`Foster, A.B., Deuterium Isotope Effects in the Metabolism of Drugs
`and Xenobiotics: Implications for Drug Design, Adv. Drug Res.,
`Academic Press, London, GB, vol. 14, 1 (1985), pp. 1-40.
`Gant et al., Benzoquinoline Inhibitors of Vesicular Monoamine
`Transporter 2, Auspex Pharmaceuticals, Inc., EP2009820972—Pros
`ecution History, Downloaded Oct. 3, 2012.
`Gant et al., Benzoquinoline Inhibitors of Vesicular Monoamine
`Transporter 2, Auspex Pharmaceuticals, Inc., NZ591615—Office
`Action, Publication Date: Jul. 21, 2011.
`Gant et al., Benzoquinoline Inhibitors of Vesicular Monoamine
`Transporter 2, Auspex Pharmaceuticals, Inc., NZ591615—Notice of
`Acceptance, Publication Date: Jun. 27, 2012.
`* cited by examiner
`
`Apotex Ex. 1001
`
`

`

`1.
`BENZOQUINOLINE INHIBITORS OF
`VESICULARMONOAMINE TRANSPORTER 2
`
`US 8,524,733 B2
`
`This application claims the benefit of priority of U.S. pro
`visional application No. 61/097.896, filed Sep. 18, 2008, the 5
`disclosure of which is hereby incorporated by reference as if
`written herein in its entirety.
`Disclosed herein are new benzoquinoline compounds,
`pharmaceutical compositions made thereof, and methods to
`inhibit vesicular monoamine transporter 2 (VMAT2) activity 10
`in a subject are also provided for, for the treatment of chronic
`hyperkinetic movement disorders.
`Tetrabenazine (Nitoman, Xenazine, Ro 1-9569), 1,3,4,6,7,
`11b-Hexahydro-9,10-dimethoxy-3-(2-methylpropyl)-2H
`benzoaquinoline, is a vesicular monoamine transporter 2 15
`(VMAT2) inhibitor. Tetrabenazine is commonly prescribed
`for the treatment of Huntington's disease (Savani et al., Neu
`rology 2007, 68(10), 797; and Kenney et al., Expert Review of
`Neurotherapeutics 2006, 6(1), 7-17).
`
`2O
`
`O
`
`N
`
`Tetrabenazine
`
`O
`1.
`
`O
`
`25
`
`30
`
`In vivo, tetrabenazine is rapidly and extensively metabo
`lized to its reduced form, 3-isobutyl-9,10-dimethoxy-1,3,4,6, 35
`7.11b-hexahydro-2H-pyrido2,1-aisoquinolin-2-ol, which
`then binds specifically to VMAT2 (Zhang et al., AAPSJour
`nal, 2006, 8(4), E682-692). Additional metabolic pathways
`involve O-demethylation of the methoxy groups, as well as
`hydroxylation of the isobutyl group (Schwartz et al., Bio- 40
`chem. Pharmacol., 1966, 15, 645-655). Adverse effects asso
`ciated with the administration of tetrabenazine include neu
`roleptic
`malignant syndrome, drowsiness,
`fatigue,
`nervousness, anxiety, insomnia, agitation, confusion, orthos
`tatic hypotension, nausea, dizziness, depression, and Parkin- 45
`Sonism.
`Deuterium Kinetic Isotope Effect
`In order to eliminate foreign Substances such as therapeutic
`agents, the animal body expresses various enzymes, such as
`the cytochrome Paso enzymes (CYPs), esterases, proteases, 50
`reductases, dehydrogenases, and monoamine oxidases, to
`react with and convert these foreign Substances to more polar
`intermediates or metabolites for renal excretion. Such meta
`bolic reactions frequently involve the oxidation of a carbon
`hydrogen (C H) bond to either a carbon-oxygen (C–O) or 55
`a carbon-carbon (C C) L-bond. The resultant metabolites
`may be stable or unstable under physiological conditions, and
`can have Substantially different pharmacokinetic, pharmaco
`dynamic, and acute and long-term toxicity profiles relative to
`the parent compounds. For most drugs, such oxidations are 60
`generally rapid and ultimately lead to administration of mul
`tiple or high daily doses.
`The relationship between the activation energy and the rate
`of reaction may be quantified by the Arrhenius equation,
`k=Ae'. The Arrhenius equation states that, at a given 65
`temperature, the rate of a chemical reaction depends expo
`nentially on the activation energy (E).
`
`2
`The transition state in a reaction is a short lived state along
`the reaction pathway during which the original bonds have
`stretched to their limit. By definition, the activation energy
`E. for a reaction is the energy required to reach the transition
`state of that reaction. Once the transition state is reached, the
`molecules can either revert to the original reactants, or form
`new bonds giving rise to reaction products. A catalyst facili
`tates a reaction process by lowering the activation energy
`leading to a transition State. Enzymes are examples of bio
`logical catalysts.
`Carbon-hydrogen bond strength is directly proportional to
`the absolute value of the ground-state vibrational energy of
`the bond. This vibrational energy depends on the mass of the
`atoms that form the bond, and increases as the mass of one or
`both of the atoms making the bond increases. Since deuterium
`(D) has twice the mass of protium (H), a C-D bond is stron
`ger than the corresponding C–H bond. If a C-H bond is
`broken during a rate-determining step in a chemical reaction
`(i.e. the step with the highest transition State energy), then
`Substituting a deuterium for that protium will cause a
`decrease in the reaction rate. This phenomenon is known as
`the Deuterium Kinetic Isotope Effect (DKIE). The magnitude
`of the DKIE can be expressed as the ratio between the rates of
`a given reaction in which a C H bond is broken, and the
`same reaction where deuterium is substituted for protium.
`The DKIE can range from about 1 (no isotope effect) to very
`large numbers, such as 50 or more. Substitution of tritium for
`hydrogen results in yet a stronger bond than deuterium and
`gives numerically larger isotope effects
`Deuterium (H or D) is a stable and non-radioactive isotope
`of hydrogen which has approximately twice the mass of pro
`tium ("H), the most common isotope of hydrogen. Deuterium
`oxide (DO or “heavy water) looks and tastes like H2O, but
`has different physical properties.
`When pure DO is given to rodents, it is readily absorbed.
`The quantity of deuterium required to induce toxicity is
`extremely high. When about 0-15% of the body water has
`been replaced by DO, animals are healthy but are unable to
`gain weight as fast as the control (untreated) group. When
`about 15-20% of the body water has been replaced with D.O.
`the animals become excitable. When about 20-25% of the
`body water has been replaced with DO, the animals become
`so excitable that they go into frequent convulsions when
`stimulated. Skin lesions, ulcers on the paws and muzzles, and
`necrosis of the tails appear. The animals also become very
`aggressive. When about 30% of the body water has been
`replaced with DO, the animals refuse to eat and become
`comatose. Their body weight drops sharply and their meta
`bolic rates drop far below normal, with death occurring at
`about 30 to about 35% replacement with D.O.The effects are
`reversible unless more than thirty percent of the previous
`body weight has been lost due to D.O. Studies have also
`shown that the use of DO can delay the growth of cancer cells
`and enhance the cytotoxicity of certain antineoplastic agents.
`Deuteration of pharmaceuticals to improve pharmacoki
`netics (PK), pharmacodynamics (PD), and toxicity profiles
`has been demonstrated previously with some classes of drugs.
`For example, the DKIE was used to decrease the hepatotox
`icity of halothane, presumably by limiting the production of
`reactive species such as trifluoroacetyl chloride. However,
`this method may not be applicable to all drug classes. For
`example, deuterium incorporation can lead to metabolic
`Switching. Metabolic Switching occurs when Xenogens,
`sequestered by Phase I enzymes, bind transiently and re-bind
`in a variety of conformations prior to the chemical reaction
`(e.g., oxidation). Metabolic switching is enabled by the rela
`tively vast size of binding pockets in many Phase I enzymes
`and the promiscuous nature of many metabolic reactions.
`Metabolic switching can lead to different proportions of
`known metabolites as well as altogether new metabolites.
`
`Apotex Ex. 1001
`
`

`

`US 8,524,733 B2
`
`10
`
`15
`
`25
`
`30
`
`3
`This new metabolic profile may impart more or less toxicity.
`Such pitfalls are non-obvious and are not predictable a priori
`for any drug class.
`Tetrabenazine is a VMAT2 inhibitor. The carbon-hydrogen
`bonds of tetrabenazine contain a naturally occurring distribu- 5
`tion of hydrogen isotopes, namely "H or protium (about
`99.984.4%), H or deuterium (about 0.0156%), and H or
`tritium (in the range between about 0.5 and 67 tritium atoms
`per 10'protium atoms). Increased levels of deuterium incor
`poration may produce a detectable Deuterium Kinetic Isotope
`Effect (DKIE) that could affect the pharmacokinetic, phar
`macologic and/or toxicologic profiles of tetrabenazine in
`comparison with tetrabenazine having naturally occurring
`levels of deuterium.
`Based on discoveries made in our laboratory, as well as
`considering the literature, tetrabenazine is metabolized in
`humans at the isobutyl and methoxy groups. The current
`approach has the potential to prevent metabolism at these
`sites. Other sites on the molecule may also undergo transfor
`mations leading to metabolites with as-yet-unknown pharma
`cology/toxicology. Limiting the production of these metabo
`lites has the potential to decrease the danger of the
`administration of Such drugs and may even allow increased
`dosage and/or increased efficacy. All of these transformations
`can occur through polymorphically-expressed enzymes,
`exacerbating interpatient variability. Further, some disorders
`are best treated when the subject is medicated around the
`clock or for an extended period of time. For all of the forego
`ing reasons, a medicine with a longer half-life may result in
`greater efficacy and cost savings. Various deuteration patterns
`can be used to (a) reduce or eliminate unwanted metabolites,
`(b) increase the half-life of the parent drug, (c) decrease the
`number of doses needed to achieve a desired effect, (d)
`decrease the amount of a dose needed to achieve a desired
`effect, (e) increase the formation of active metabolites, if any
`are formed, (f) decrease the production of deleterious
`metabolites in specific tissues, and/or (g) create a more effec
`tive drug and/or a safer drug for polypharmacy, whether the
`40
`polypharmacy be intentional or not. The deuteration
`approach has the strong potential to slow the metabolism of
`tetrabenazine and attenuate interpatient variability.
`Novel compounds and pharmaceutical compositions, cer
`tain of which have been found to inhibit VMAT2 have been
`discovered, together with methods of synthesizing and using
`the compounds, including methods for the treatment of
`VMAT2-mediated disorders in a patient by administering the
`compounds as disclosed herein.
`In certain embodiments of the present invention, com
`pounds have structural Formula I:
`50
`
`4
`or a salt, Solvate, or prodrug thereof, wherein:
`R-R, are independently selected from the group consist
`ing of hydrogen and deuterium; and
`at least one of R-R-7 is deuterium.
`In certain embodiments, Formula I can include a single
`enantiomer, a mixture of the (+)-enantiomer and the (-)-
`enantiomer, a mixture of about 90% or more by weight of the
`(-)-enantiomer and about 10% or less by weight of the (+)-
`enantiomer, a mixture of about 90% or more by weight of the
`(+)-enantiomer and about 10% or less by weight of the (-)-
`enantiomer, an individual diastereomer, or a mixture of dias
`tereomers thereof.
`Certain compounds disclosed herein may possess useful
`VMAT2 inhibiting activity, and may be used in the treatment
`or prophylaxis of a disorder in which VMAT2 plays an active
`role. Thus, certain embodiments also provide pharmaceutical
`compositions comprising one or more compounds disclosed
`herein together with a pharmaceutically acceptable carrier, as
`well as methods of making and using the compounds and
`compositions. Certain embodiments provide methods for
`inhibiting VMAT2. Other embodiments provide methods for
`treating a VMAT2-mediated disorder in a patient in need of
`Such treatment, comprising administering to said patient a
`therapeutically effective amount of a compound or composi
`tion according to the present invention. Also provided is the
`use of certain compounds disclosed herein for use in the
`manufacture of a medicament for the prevention or treatment
`of a disorder ameliorated by the inhibition of VMAT2.
`The compounds as disclosed herein may also contain less
`prevalent isotopes for other elements, including, but not lim
`ited to, C or C for carbon, S, S, or S for sulfur, 'N
`for nitrogen, and 'O or "O for oxygen.
`In certain embodiments, the compound disclosed herein
`may expose apatient to a maximum of about 0.000005% DO
`or about 0.00001% DHO, assuming that all of the C-D bonds
`in the compound as disclosed herein are metabolized and
`released as DO or DHO. In certain embodiments, the levels
`of DO shown to cause toxicity in animals is much greater
`than even the maximum limit of exposure caused by admin
`istration of the deuterium enriched compound as disclosed
`herein. Thus, in certain embodiments, the deuterium-en
`riched compound disclosed herein should not cause any addi
`tional toxicity due to the formation of DO or DHO upon drug
`metabolism.
`In certain embodiments, the deuterated compounds dis
`closed herein maintain the beneficial aspects of the corre
`sponding non-isotopically enriched molecules while Substan
`tially increasing the maximum tolerated dose, decreasing
`toxicity, increasing the half-life (T), lowering the maxi
`mum plasma concentration (C) of the minimum effica
`cious dose (MED), lowering the efficacious dose and thus
`decreasing the non-mechanism-related toxicity, and/or low
`ering the probability of drug-drug interactions.
`All publications and references cited herein are expressly
`incorporated herein by reference in their entirety. However,
`with respect to any similar or identical terms found in both the
`incorporated publications or references and those explicitly
`put forth or defined in this document, then those terms defi
`nitions or meanings explicitly put forth in this document shall
`control in all respects.
`As used herein, the terms below have the meanings indi
`cated.
`The singular forms “a,” “an and “the may refer to plural
`articles unless specifically stated otherwise.
`The term “about as used herein, is intended to qualify the
`numerical values which it modifies, denoting Such a value as
`variable within a margin of error. When no particular margin
`of error, such as a standard deviation to a mean value given in
`
`35
`
`5
`
`
`
`(I)
`
`55
`
`60
`
`65
`
`Apotex Ex. 1001
`
`

`

`US 8,524,733 B2
`
`10
`
`15
`
`25
`
`30
`
`5
`a chart or table of data, is recited, the term “about should be
`understood to mean that range which would encompass the
`recited value and the range which would be included by
`rounding up or downto that figure as well, taking into account
`significant figures.
`When ranges of values are disclosed, and the notation
`“from n ... to n' or “n-n' is used, where n and n are the
`numbers, then unless otherwise specified, this notation is
`intended to include the numbers themselves and the range
`between them. This range may be integral or continuous
`between and including the end values.
`The term “deuterium enrichment” refers to the percentage
`of incorporation of deuterium at a given position in a mol
`ecule in the place of hydrogen. For example, deuterium
`enrichment of 1% at a given position means that 1% of mol
`ecules in a given sample contain deuterium at the specified
`position. Because the naturally occurring distribution of deu
`terium is about 0.0156%, deuterium enrichment at any posi
`tion in a compound synthesized using non-enriched starting
`materials is about 0.0156%. The deuterium enrichment can
`be determined using conventional analytical methods known
`to one of ordinary skill in the art, including mass spectrometry
`and nuclear magnetic resonance spectroscopy.
`The term “is/are deuterium, when used to describe a given
`position in a molecule such as R-R-7 or the symbol “D’.
`when used to represent a given position in a drawing of a
`molecular structure, means that the specified position is
`enriched with deuterium above the naturally occurring distri
`bution of deuterium. In one embodiment deuterium enrich
`ment is no less than about 1%, in another no less than about
`5%, in another no less than about 10%, in another no less than
`about 20%, in another no less than about 50%, in another no
`less than about 70%, in another no less than about 80%, in
`another no less than about 90%, or in another no less than
`about 98% of deuterium at the specified position.
`35
`The term “isotopic enrichment” refers to the percentage of
`incorporation of a less prevalent isotope of an element at a
`given position in a molecule in the place of the more prevalent
`isotope of the element.
`The term “non-isotopically enriched refers to a molecule
`in which the percentages of the various isotopes are Substan
`tially the same as the naturally occurring percentages.
`Asymmetric centers exist in the compounds disclosed
`herein. These centers are designated by the symbols “R” or
`“S” depending on the configuration of Substituents around
`the chiral carbonatom. It should be understood that the inven
`tion encompasses all Stereochemical isomeric forms, includ
`ing diastereomeric, enantiomeric, and epimeric forms, as
`well as D-isomers and L-isomers, and mixtures thereof. Indi
`vidual stereoisomers of compounds can be prepared syntheti
`cally from commercially available starting materials which
`contain chiral centers or by preparation of mixtures of enan
`tiomeric products followed by separation Such as conversion
`to a mixture of diastereomers followed by separation or
`recrystallization, chromatographic techniques, direct separa
`tion of enantiomers on chiral chromatographic columns, or
`any other appropriate method known in the art. Starting com
`pounds of particular Stereochemistry are either commercially
`available or can be made and resolved by techniques known in
`the art. Additionally, the compounds disclosed herein may
`exist as geometric isomers. The present invention includes all
`cis, trans, syn, anti, entgegen (E), and Zusammen (Z) isomers
`as well as the appropriate mixtures thereof. Additionally,
`compounds may exist as tautomers; all tautomeric isomers
`are provided by this invention. Additionally, the compounds
`disclosed herein can exist in unsolvated as well as Solvated
`forms with pharmaceutically acceptable solvents such as
`
`55
`
`6
`water, ethanol, and the like. In general, the Solvated forms are
`considered equivalent to the unsolvated forms.
`The term “bond refers to a covalent linkage between two
`atoms, or two moieties when the atoms joined by the bond are
`considered to be part of larger substructure. A bond may be
`single, double, or triple unless otherwise specified. A dashed
`line between two atoms in a drawing of a molecule indicates
`that an additional bond may be present or absent at that
`position.
`The term “disorder as used herein is intended to be gen
`erally synonymous, and is used interchangeably with, the
`terms “disease”, “syndrome', and “condition' (as in medical
`condition), in that all reflect an abnormal condition of the
`human or animal body or of one of its parts that impairs
`normal functioning, is typically manifested by distinguishing
`signs and Symptoms.
`The terms “treat,” “treating.” and “treatment are meant to
`include alleviating or abrogating a disorder or one or more of
`the symptoms associated with a disorder, or alleviating or
`eradicating the cause(s) of the disorder itself. As used herein,
`reference to “treatment of a disorder is intended to include
`prevention. The terms “prevent,” “preventing, and “preven
`tion” refer to a method of delaying or precluding the onset of
`a disorder, and/or its attendant symptoms, barring a subject
`from acquiring a disorder or reducing a subject’s risk of
`acquiring a disorder.
`The term “therapeutically effective amount” refers to the
`amount of a compound that, when administered, is sufficient
`to prevent development of oralleviate to some extent, one or
`more of the symptoms of the disorder being treated. The term
`“therapeutically effective amount” also refers to the amount
`of a compound that is sufficient to elicit the biological or
`medical response of a cell, tissue, system, animal, or human
`that is being sought by a researcher, Veterinarian, medical
`doctor, or clinician.
`The term “subject” refers to an animal, including, but not
`limited to, a primate (e.g., human, monkey, chimpanzee,
`gorilla, and the like), rodents (e.g., rats, mice, gerbils, ham
`sters, ferrets, and the like), lagomorphs, Swine (e.g., pig,
`miniature pig), equine, canine, feline, and the like. The terms
`“subject' and “patient” are used interchangeably herein in
`reference, for example, to a mammalian Subject, such as a
`human patient.
`The term “combination therapy’ means the administration
`of two or more therapeutic agents to treat a therapeutic dis
`order described in the present disclosure. Such administration
`encompasses co-administration of these therapeutic agents in
`a Substantially simultaneous manner, Such as in a single cap
`Sule having a fixed ratio of active ingredients or in multiple,
`separate capsules for each active ingredient. In addition, Such
`administration also encompasses use of each type of thera
`peutic agent in a sequential manner. In either case, the treat
`ment regimen will provide beneficial effects of the drug com
`bination in treating the disorders described herein.
`The term "chronic hyperkinetic movement disorders'
`refers to disorders characterized by non-purposeful, repeti
`tive, disordered motor acts, variously termed “compulsive'.
`“rhythmical’, or “stereotyped.” In humans, chronic hyperki
`netic movement disorders can be psychogenic (e.g., tics),
`idiopathic (as in, e.g., Tourette's syndrome and Parkinson's
`Disease, genetic (as in, e.g., the chorea characteristic of Hun
`tington's Disease), infectious (as in, e.g., Sydenham's Cho
`rea), or, as in tardive dyskinesia, drug-induced. Unless other
`wise stated, "chronic hyperkinetic movement disorders'
`refers to and includes all psychogenic, idiopathic, genetic,
`and drug-induced movement disorders.
`
`40
`
`45
`
`50
`
`60
`
`65
`
`Apotex Ex. 1001
`
`

`

`US 8,524,733 B2
`
`7
`The term “stereotyped refers to a repeated behavior that
`appears repetitively with slight variation or, less commonly,
`as a complex series of movements.
`The term “VMAT2 refers to vesicular monoamine trans
`porter 2, an integral membrane protein that acts to transport 5
`monoamines—particularly neurotransmitters such as
`dopamine, norepinephrine, serotonin, and histamine—from
`cellular cytosol into synaptic vesicles.
`The term “VMAT2-mediated disorder” refers to a disorder
`that is characterized by abnormal VMAT2 activity. A 10
`VMAT2-mediated disorder may be completely or partially
`mediated by modulating VMAT2. In particular, a VMAT2
`mediated disorder is one in which inhibition of VMAT2
`results in Some effect on the underlying disorder e.g., admin
`istration of a VMAT2 inhibitor results in some improvement 15
`in at least Some of the patients being treated.
`The term “VMAT2 inhibitor, “inhibit VMAT2, or “inhi
`bition of VMAT2 refers to the ability of a compound dis
`closed herein to alter the function of VMAT2. A VMAT2
`inhibitor may block or reduce the activity of VMAT2 by 20
`forming a reversible or irreversible covalent bond between
`the inhibitor and VMAT2 or through formation of a nonco
`valently bound complex. Such inhibition may be manifest
`only in particular cell types or may be contingent on a par
`ticular biological event. The term “VMAT2 inhibitor, 25
`“inhibit VMAT2', or “inhibition of

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