throbber
(12) United States Patent
`Tung
`
`(10) Patent No.:
`(45) Date of Patent:
`
`US 7,678,914 B2
`Mar. 16, 2010
`
`USOO7678914B2
`
`(54) DEUTERATED BENZODI1,3-DIOXOL
`DERVATIVES
`
`(75) Inventor: Roger Tung, Lexington, MA (US)
`
`(73) Assignee: Concert Pharmaceuticals Inc.,
`Lexington, MA (US)
`
`- r
`(*) Notice:
`
`-
`Subject to any disclaimer, the term of this
`patent is extended or adjusted under 35
`
`(21) Appl. No.: 11/704,554
`
`(22) Filed:
`
`Feb. 8, 2007
`
`(65)
`
`Prior Publication Data
`US 2007/0191432 A1
`Aug. 16, 2007
`O
`O
`Related U.S. Application Data
`(63) Continuation-in-part of application No. 1 1/498.334,
`filed on Jul. 31, 2006.
`(60) Provisional application No. 60/704,073, filed on Jul.
`29, 2005.
`s
`(51) Int. Cl.
`(2006.01)
`CO7D 2L/22
`(2006.01)
`AOIN 43/62
`(52) U.S. Cl. ....................................... 546/197; 514/321
`(58) Field of Classification Search ................. 546/197;
`514/321
`See application file for complete search history.
`References Cited
`U.S. PATENT DOCUMENTS
`4.007,196 A
`2f1977 Christensen et al. ........
`5557.826 A
`1/1997 Howard et al.
`5,874.447 A
`2f1999 Benneker et al.
`6,436,938 B1
`8/2002 Howard, Jr.
`6,720,003 B2
`4/2004 Chen et al.
`2002fOO13372 A1
`1/2002 Ekins
`2007, 0112031 A1
`5, 2007 Gant et al.
`
`(56)
`
`OTHER PUBLICATIONS
`Leis et al J. Mass Spectrom. 2001, 36,923-928.*
`Foster et al Trends in Pharma. Sci. 1984, 5,524-527.*
`U.S. Appl. No. 1 1/498.334, filed Jul. 31, 2006, Tung.
`Fisher, M.B. et al., Complexities Inherent in Attempts tO Decrease
`Drug Clearance by Blocking Sites of CYP-mediated Metabolism,
`Current Opinion. In Drug Discovery & Development, vol. 9(1), pp.
`101-109 (2006).
`FDA Center for Drug Evaluation and Drug Research, NDA No.
`21-299, Clinical Pharmacology and Biopharmaceutics Review(s),
`
`Fukuto, J.M. et al., “Determination of the Mechanism of
`Demethylenation of (Methylenedioxy)phenyl Compounds by
`Cytochrome P450 Using Deuterium Isotope Effects”. J. Med. Chem.
`34:2871-2876 (1991).
`Kaye, C.M. et al., “A review of the metabolism and pharmacokinetics
`of paroxetine in man'. Acta Psychiatr. Scand. 80(Supp. 350):60-75
`(1989).
`Bertelsen, K.M. et al., "Apparent Mechanism-Based Inhibition of
`Human CYP2D6 In Vitro Bv Paroxetine: Comparison with
`Fluoxetine and Quinidine'. Dr. Metab. Dispos. Eo.
`(2003).
`k . cited by examiner
`Primary Examiner Rita J Desai
`Assistant Examiner John Mabry
`(74)Attorney, Agent, or Firm Foley & Lardner LLP, Steven
`G. Davis
`
`(7)
`
`ABSTRACT
`
`The present invention relates to an isotopologue of Com
`pound 1 substituted with deuterium at the methylene carbon
`of the benzodioxol ring. The isotopologues of this invention
`selective serotonin reuptake inhibitors (SSRIs) and possess
`unique biopharmaceutical and metabolic properties com
`pared to Compound 1. They may also be used to accurately
`determine the concentration of Compound 1 in biological
`fluids and to determine metabolic patterns of Compound 1
`and its isotopologues. The invention further provides compo
`sitions comprising these deuterated isotopologues and meth
`ods of treating diseases and conditions that are responsive to
`increased neuronal serotonin transmission, alone and in com
`
`2008.OO33011 A1* 2, 2008 Tung - - - - - - - - - - - - - - - - - - - - - - - - - - 514,321
`
`bination with additional agents.
`
`WO
`
`FOREIGN PATENT DOCUMENTS
`WO95/26325
`* 10/1995
`
`4 Claims, No Drawings
`
`Auspex Exhibit 2007
`Apotex v. Auspex
`IPR2021-01507
`Page 1
`
`

`

`1.
`DEUTERATED BENZODI1,3-DIOXOL
`DERVATIVES
`
`US 7,678,914 B2
`
`CROSS REFERENCE TO RELATED
`APPLICATIONS
`
`This application is a continuation-in-part of U.S. patent
`application Ser. No. 1 1/498.334, filed Jul. 31, 2006, which
`claims benefit of U.S. provisional application 60/704,073,
`filed Jul. 29, 2005, the contents of each is incorporated by
`reference herein.
`
`10
`
`TECHNICAL FIELD OF THE INVENTION
`
`15
`
`The present invention relates to novel isotopologues of
`Compound 1, its acceptable acid with additional deuterium
`and C atoms in place of the normally abundant hydrogen
`and C, respectively addition salts, solvates, hydrates and
`polymorphs thereof, substituted with deuterium on the meth
`ylene carbon atom situated between the oxygens of the ben
`Zodioxol ring, and optionally substituted. The compounds of
`this invention are selective serotonin reuptake inhibitors (SS
`RIs) and are poorer substrates for metabolism by cytochrome
`2D6, and possess unique pharmacokinetic and biopharma
`ceutical properties compared to the corresponding non-iso
`topically Substituted compounds. The invention also provides
`compositions comprising a compound of this invention and
`the use of Such compositions in methods of treating diseases
`and conditions beneficially treated by SSRIs, particularly
`those relating to major depressive disorder, obsessive com
`30
`pulsive disorder, panic disorder, social anxiety disorder, gen
`eralized anxiety disorder, post-traumatic stress disorder, and
`premenstrual dysphoric disorder. The invention further pro
`vides methods for the use of a compound of this invention to
`determine concentrations of Compound 1, particularly in bio
`35
`logical fluids, and to determine metabolism patterns of Com
`pound 1.
`
`25
`
`BACKGROUND OF THE INVENTION
`
`Compound 1, chemically described variously as (-)-trans
`4R-(4-fluorophenyl)-3S-(3',4'-methylenedioxyphenoxy)
`methylpiperidine;
`(3S,4R)-3-((benzod1.3dioxol-5-
`yloxy)methyl)-4-(4-fluorophenyl)piperidine;
`trans-(-)-3-
`(1,3-benzodioxol-5-yloxy)methyl-4-(4-fluorophenyl)
`piperidine, and its pharmaceutically acceptable
`
`40
`
`45
`
`Compound 1
`
`50
`
`55
`
`60
`
`65
`
`N
`
`addition salts, hydrates, and polymorphs thereof, are known
`as a useful selective serotonin reuptake inhibitor (SSRI). This
`compound and pharmaceutical compositions comprising it
`have utility in the treatment of depression, obsessive-compul
`sive disorder, generalized anxiety, post-traumatic stress,
`
`2
`major depression, panic disorder, social phobia, premenstrual
`syndrome, cardiac disorders, non-cardiac chest pain, Smok
`ing (both to cause cessation and prevent relapses), reducing
`platelet activation states, alcoholism and alcohol dependence,
`psychiatric syndromes (including anger, rejection sensitivity,
`and lack of mental or physical energy), late luteal phase
`dysphoric disorder, premature ejaculation, senile dementia,
`obesity, Parkinson's Disease, and canine affective aggres
`sion. See US Food and Drug Administration product label for
`New Drug Application (NDA) Nos. 020031, 020710, and
`020936: Christensen J A and Squires R F, U.S. Pat. No.
`4,007,196, to Ferrosan; Lassen JB, U.S. Pat. No. 4,745,122 to
`Ferrosan; Johnson AMU.S. Pat. No. 5,371,092 to Beecham
`Group: Crenshaw RT and Wiesner MG, U.S. Pat. No. 5,276,
`042: Dodman NH, U.S. Pat. Nos. 5,788,986 and 5,554,383 to
`Trustees of Tufts College: Norden MJ U.S. Pat. No. 5,789,
`449; Gleason M, U.S. Pat. No. 6,121,291 to SmithKline Bee
`cham; Cook L., U.S. Pat. No. 6,071,918 to DuPont Pharma
`ceuticals; Serebruany V L., U.S. Pat. No. 6,245,782 to
`Heartdrug Research: Steiner MX, U.S. Pat. No. 6,300,343 to
`SmithKline Beecham; Krishnan K R et. al., U.S. Pat. No.
`6,316,469 to Duke University; Jenner P N, U.S. Pat. No.
`6,372,763 to SmithKline Beecham.
`Additionally disclosed uses for Compound 1 include meth
`ods of inhibiting cancer cell growth, stimulating bone forma
`tion by osteoblast stimulation, treatment of dermatological
`diseases or disorders such as hyperproliferative or inflamma
`tory skin diseases, and treatment of premature female
`orgasm: see US Patent Applications 20040127573 (Telerman
`A et al.); 20040127573 (Stashenko P and Battaglino R):
`200500 13853 and 20040029860 (Gil-Ad I and Weizman A);
`and 2005.0054688 (May K E and Quinn P).
`Definitions and descriptions of these conditions are known
`to the skilled practitioner and are further delineated, for
`instance, in the above patents and patent applications and
`references contained therein. See also: Harrison's Principles
`of Internal Medicine 16th Edition, Kasper D Let. al. Eds.
`2004, McGraw-Hill Professional; and Robbins & Cotran
`Pathologic Basis of Disease, Kumar Vet. al. Eds., 2004, W.B.
`Saunders.
`The combination of Compound 1 with additional agents
`extends or enhances its utility in the treatment or prevention
`of depression, hypertension, generalized anxiety disorder,
`phobias, posttraumatic stress syndrome, avoidant personality
`disorder, sexual dysfunction, eating disorders (including
`bulimia, anorexia nervosa, and binge eating), obesity, chemi
`cal dependencies, clusterheadache, migraine, pain (including
`neuropathic pain, diabetic nephropathy, post-operative pain,
`psychogenic pain disorders, and chronic pain syndrome),
`Alzheimer's disease, obsessive-compulsive disorder, panic
`disorder with or without agoraphobia, memory disorders,
`Parkinson's diseases, endocrine disorders, vasospasm, cer
`ebellar ataxia, gastrointestinal tract disorders, negative symp
`toms of Schizophrenia, premenstrual syndrome, Fibromyal
`gia Syndrome, urinary incontinence (including stress
`incontinence), Tourette's syndrome, trichotillomania, klepto
`mania, male impotence, cancer, chronic paroxysmal hemic
`rania and headache in a mammal, sleep-related breathing
`disorders, cognitive deficits due to aging, stroke, head trauma,
`neurodegenerative diseases, schizophrenia, anxiety, aggres
`Sion, stress, disorders of thermoregulation, respiratory dis
`ease, bipolar disorder, psychosis, sleep disorders, mania (in
`cluding acute mania), bladder disorder, genitourinary
`disorder, cough, emesis, nausea, psychotic disorders such as
`paranoia and manic-depressive illness, tic disorder, diabetic
`cardiomyopathy, diabetic retinopathy, cataracts, myocardial
`infarction, prolonged fatigue, chronic fatigue, chronic fatigue
`
`Auspex Exhibit 2007
`Apotex v. Auspex
`IPR2021-01507
`Page 2
`
`

`

`US 7,678,914 B2
`
`5
`
`10
`
`15
`
`25
`
`3
`syndrome, premature ejaculation, dysphoria, post partum
`depression, Social phobia, disruptive behavior disorders,
`impulse control disorders, borderline personality disorder,
`attention deficit disorders without hyperactivity, Shy-Drager
`Syndrome, cerebral ischemia, spinal cord trauma, Hunting
`ton's Chorea, amyotrophic lateral sclerosis, AIDS-induced
`dementia, muscular spasms, convulsions, perinatal hypoxia,
`hypoxia, cardiac arrest, hypoglycemic neuronal damage,
`ocular damage and retinopathy, brain edema, tardive dyski
`nesia, cerebral deficits Subsequent to cardiac bypass Surgery
`and grafting, affective disorders, mood disorders, agorapho
`bia without history of panic disorder, and acute stress disor
`ders. These additional agents are also useful for reducing the
`side effects of Compound 1, enhancing or potentiating its
`activity, or increasing its duration of pharmacological action.
`U.S. Pat. Nos. 5,776,969 (James SP) to Eli Lilly; 5,877,171
`(McLeod MN); 5,977,099 (Nickolson VJ) to Akzo Nobel:
`5,962,514 and 6,169,098 (Evenden J and Thorberg S-O) to
`Astra; 5,958,429 (Wong DT) to Eli Lilly; 5,945,416 (Shan
`non H E and Womer DE) to Eli Lilly: 6,066,643 (Perry KW)
`to Eli Lilly; 5,817,665 and 6,034,091 (Dante LG) to Nagle J
`S; 5.990,159 (Meulemans A L G et. al.) to Janssen Pharma
`ceutica; 6,001,848 (Noble EP) to The Regents of the Univer
`sity of California; 6,011,054 (Oxenkrug G F and Requintina
`PJ) to St. Elizabeth's Medical Center of Boston; 6,080,736
`(Landry D W and Klein D F) to Janus Pharmaceuticals;
`6,162,805 (Hefti FF) to Merck Sharp & Dohme: 6,136,861
`(Chenard BL) to Pfizer: 6,147,072 (Bymaster F Pet. al.) to
`Eli Lilly; 6,218,395 (Swartz CM); 6,169,105 (Wong DT and
`Oguiza JI) to Eli Lilly: 6,191,133 (Coppen AJ) to Scarista;
`6,239,126 and 6.242,448 (Kelly M G et. al.) to American
`Home Products; 6,372,919 (Lippa AS and Epstein JW) to
`DOV; 6,369,051 (Jenkins S N) to American Home Products:
`6,358,944 (Lederman Set. al.) to Vela Pharmaceuticals;
`6,121,259; 6,174,882; 6,348.455; 6,352,984; and 6,468,997
`(Yelle WE) to Sepracor; 6,403,597 (Wilson L. Fet. al.) to
`Vivus: 6,395,788 and 6,541,523 (Iglehart I W III) to Vela
`Pharmaceuticals; 6,127,385 and 6,395,752 (Midha K. Ket.
`al.) to Pharmaquest Limited; 6.380.200 (Mylari B L) to
`Pfizer; 6.387,956 (Shapira N A et al.) to University of Cin
`cinnati; 6,444,665 (Helton D Ret. al.) to Eli Lilly: 6,541,478
`(O'Malley Set. al.) to Yale University; 6.541,043 (Lang PC)
`to DexGen Pharmaceuticals; 6,562,813 (Howard H R) to
`Pfizer: 6,579,899 (Wurtman JJ and Wurtman RJ) to Massa
`chusetts Institute of Technology: 6,627,653 (Plata-Salaman C
`45
`Ret. al.) to Ortho-McNeil; 6,649,614 (Carlson E J and Rup
`niak N M) to Merck Sharp & Dohme: 6,667,329 (MajJ) to
`Boehringer Ingelheim; 6,727,242 (Radulovacki M and Car
`ley D W) to The Board of Trustees of the University of
`Illinois; 6,656,951: 6,780,860; 6,815,448; 6,821,981; and
`6,861,427 (Stack; Gary Pet. al.) to Wyeth: 6.878,732 (Wrob
`leski ML) to Schering Corporation; and 6,894,053 (Childers
`W E et al.) to Wyeth.
`Further disclosed are additional combinations of Com
`pound 1 with other agents extending or enhancing its utility in
`55
`the treatment or prevention of autism, dyskinesia, disthymic
`disorder, obesity due to genetic or environmental causes,
`polycystic ovary disease, craniopharyngeoma, Prader-Willi
`Syndrome. Frohlich's Syndrome, Type II diabetes, growth
`hormone deficiency, Turner's Syndrome; pro-inflammatory
`cytokine secretion or production, jet lag, insomnia, hyper
`Somnia, nocturnal enuresis, restless-legs syndrome, vaso-oc
`clusive events, hyperglycemia, hyperinsulinaemia, hyperlipi
`daemia, hypertriglyceridemia, diabetes, insulin resistance,
`impaired glucose metabolism, conditions of impaired glu
`cose tolerance (IGT), conditions of impaired fasting plasma
`glucose, glomerulosclerosis, syndrome X, coronary heart dis
`
`4
`ease, angina pectoris, vascular restenosis, endothelial dys
`function, impaired vascular compliance, or congestive heart
`failure; or to increase the onset of action of Compound 1. US
`Patent
`Applications
`20020032197,
`20020002137,
`20020086865, 20020077323, 20020103249, 20020094960,
`2003.0109544, 20030092770, 2003.014.4270, 20030158173,
`2003.0139395, 20030055070, 2003.0139429, 20040044005,
`20010014678, 20040044005, 20030235631, 20030027817,
`20030229001, 20030212060, 20040132797, 20040204469,
`20040204401, 2004.0171664, 20040229940, 20040229941,
`20040229942, 20040229911, 20040224943, 20040229866,
`20040224.942, 20040220153, 20040229849, 20050069596,
`20050059654, 20050014848, 20050026915, 20050026946,
`20050143350, 20020035105, 20050143314, 20050137208,
`20040010035, 20040013741, 20050136127, 200501 19248,
`200501 19160, 20050085477, 20050085475, 20010003749,
`20050009815, 20040248956, 20050014786, 20050009870,
`2005.0054659, 20050143381, 20050080087, 20050070577,
`and 20050080084.
`Compound 1 has been characterized by in vitro studies of
`binding to rat cortical membranes, wherein radiolabeled
`Compound 1 was found to bind to a single, high affinity,
`saturable site. See e.g. Habert E et. al., Eur. J. Pharmacol.
`1985 118: 107.
`Compound 1 has also been characterized in a number of
`animal model systems. For instance, in models of depression,
`obesity, and anxiety, treatment with Compound 1 accurately
`produced results that are correlated with human clinical
`effects. See, e.g. Akegawa Y et. al. Methods Find Exp Clin
`Pharmacol 1999 21:599; Lassen JB, U.S. Pat. No. 4,745,122
`to Ferrosan; and Hascoet Met. al., Pharmacol. Biochem.
`Behav. 2000 65: 339.
`In human clinical studies, Compound 1 demonstrated good
`tolerability and statistical efficacy in patients suffering from
`major depression, minor depression and dysthymia, obses
`sive-compulsive disorder, panic disorder, Social anxiety dis
`order, generalized anxiety disorder, and post-traumatic stress
`disorder. Compound 1 is highly effective, for instance dem
`onstrating Superior antidepressant effects to other compounds
`with the same mechanism of action in a number of direct
`comparison studies. See, e.g. US Food and Drug Administra
`tion product label for New Drug Application (NDA) Nos.
`020031,020710, and 020936: WagstaffAJet. al., Drugs 2002
`62: 655; Katona C and Livingston G. J. Affect. Disord. 2002
`69: 47.
`Following oral administration to humans, Compound 1 is
`well absorbed, after which it undergoes extensive oxidative
`and phase II metabolism. Its major metabolic pathway pro
`ceeds by oxidative cleavage of the benzodioxol ring to form
`ing a catechol metabolite. Subsequent phase II metabolism
`involves mainly methylation, glucuronidation and Sulfation.
`See Scheme I. In vitro measurements indicate that these
`metabolites possess <2% of the potency of Compound 1 and
`therefore do not contribute pharmacodynamically to its
`action. During a 10-day post-dosing period following a 30 mg
`oral solution dose of radiolabeled Compound 1 in healthy
`volunteers, approximately 64% of Compound 1 was found to
`be excreted in the urine, comprising 2% as the parent com
`pound and 62% as metabolites. About 3.6% was excreted in
`the feces, mostly as metabolites andless than 1% as the parent
`compound during this period. US FDA approved label for
`NDA # 020031, approved Jan. 12, 2005.
`
`30
`
`35
`
`40
`
`50
`
`60
`
`65
`
`Auspex Exhibit 2007
`Apotex v. Auspex
`IPR2021-01507
`Page 3
`
`

`

`5
`The benzodioxol ring Scission is carried out in significant
`part by cytochrome 2D6 (CYP2D6), which acts as a high
`affinity, but relatively low capacity, oxidant. Compound 1
`also acts as a highly potent, mechanism based inactivator of
`CYP2D6, possibly through formation of a carbene interme
`diate during the metabolic oxidation step or by formation of
`an ortho-quinone and Subsequent reaction with active-site
`nucleophiles. Bertelsen K M et. al., Drug Metab. Dispos.
`200331: 289; Murray M, Curr. Drug Metab. 20001: 67: Ortiz to
`de Montellano and Correi MA in “Cytochrome P450 Struc
`ture, Mechanism and Biochemistry” (Ortiz de Montellano PR
`ed) pp. 305-366, 1995 Plenum Press, New York; Wu et al.,
`Biochem. Pharmacol. 199753: 1605; Bolton J Let. al., 1994
`Chem. Res. Toxicol. 7: 443.
`Clinically, this mechanism-based inactivation manifests in
`several ways. For instance, Compound 1 displays signifi
`cantly non-linearity pharmacokinetics, with steady state
`doses several times the levels expected from a single dose as 20
`a result of auto-inhibition of its metabolism. Compound 1
`also causes a dose-dependent, highly significant reduction in
`CYP2D6 activity. CYP2D6 comprises the main metabolic
`pathway for a number of other clinically important drugs,
`including for instance anti-cancer agents, other anti-depres
`sants, and antipsychotics; as well as drugs of abuse Such as the
`widely used drug "Ecstasy”. Co-dosing Compound 1 with
`those agents causes clinically significant increases in their
`blood levels, leading to the potential for increased toxicity. 30
`Jeppesen Uet. al., Eur. J. Clin. Pharmacol. 199651: 73; US
`FDA approved label for NDA # 020935, approved Jan. 12,
`2005; Laugesen Set. al., Clin Pharmacol Ther. 2005 77:312:
`Jin Yet. al., J. Natl. Cancer Inst. 200597:30; Joos AAB et al.,
`Pharmacopsychiat. 1997.30,266; Segura Met. al., Clin Phar
`macokinet. 2005 44: 649.
`Compound 1 is Subject to Substantial inter-patient varia
`tion. Patients possessing relatively low and relatively high
`levels of CYP2D6 activity have been shown to metabolize 40
`Compound 1 at Substantially different rates, leading to an
`approximately 3-fold longer half-life in a European cohort of
`poor metabolizers (PMs) with low CYP2D6-mediated oxida
`tive efficiency versus extensive metabolizers (EMs) with
`higher CYP2D6 activity: Sindrup S Het. al., Clin. Pharmacol. 45
`1992 51: 278. Even when measured at steady state, at which
`time variability is Substantially less than on initial dosing,
`high variability of Compound 1 was observed in a test popu
`lation (about 30-70% coefficients of variability across maxi- so
`mal and minimal plasma concentrations (Cmax and Cmin)
`and overall exposure measured as area under the plasma
`concentration-time curve (AUC)). Kaye C Met. al., Acta
`Psychiatr. Scand. 80(Suppl. 350): 60.
`CYP2D6 is the source of substantial variability in the phar
`macokinetics of a number of drugs due to well-known poly
`morphisms resulting in low CYP2D6 activity in a substantial
`percentage of the population, including about 2% of Asians
`and 7-8% of Caucasians (Wolf C R and Smith G, IARC Sci.
`Publ. 1999 148: 209 (chapter 18); Mura Cet. al., Br. J. Clin.
`Pharmacol. 1993 35: 161; Shimizu Tet. al., Drug Metab.
`Pharmacokinet. 2003 18: 48). Notably, different CYP2D6
`polymorphisms exist across racial types, and it is possible that
`the even greater variability may exist in other patient popu- 65
`lations with different pharmacogenomic backgrounds. Shi
`mada Tet. al., Pharmacogenetics 2001 11: 143.
`
`25
`
`55
`
`60
`
`US 7,678,914 B2
`
`5
`
`15
`
`Scheme I
`Compound 1
`Enzymatic
`Oxidation
`
`F
`
`OH
`
`OH
`
`O
`
`N
`H
`
`/ Penis
`
`\
`
`F
`
`OH
`
`F
`
`Y,
`
`F
`
`O
`
`W
`
`O
`
`w
`
`O
`H
`- w
`
`N
`H
`
`N
`H
`
`N
`H
`
`O-Glucuronidation and Sulfation, Excretion
`
`It is therefore desirable to create a compound displaying
`the beneficial activities of Compound 1, but with a decreased
`metabolic liability for CYP2D6, to further extend its pharma
`cological effective life in extensive metabolizers, decrease
`population pharmacokinetic variability and/or decrease its
`potential for dangerous drug-drug interactions.
`
`SUMMARY OF THE INVENTION
`
`The present invention solves the problems set forth above
`by providing an isolated compound of Formula I:
`
`Formula I
`
`N
`
`Auspex Exhibit 2007
`Apotex v. Auspex
`IPR2021-01507
`Page 4
`
`

`

`US 7,678,914 B2
`
`5
`
`10
`
`15
`
`7
`or a salt thereof, or a prodrug, or a salt of a prodrug thereof; or
`a hydrate, solvate, or polymorph thereof; wherein:
`D is deuterium;
`each Y is independently selected from deuterium or hydro
`gen,
`each hydrogen is independently optionally replaced with
`deuterium; and
`each carbon is independently optionally replaced with C.
`A compound of Formula I reduces the efficiency of benzo
`dioxol ring cleavage by CY2D6 and beneficially decreases
`the rate of mechanism-based CYP2D6 inhibition relative to
`Compound 1.
`The decreased CYP2D6 inhibition is important in reducing
`the pharmacokinetic interactions between Compound 1 and
`other drugs metabolized by that enzyme. This provides
`increased safety as compared to Compound 1.
`In particular, this would produce benefits in the treatment
`of co-morbidities and the use of combinations of medications,
`which is common in patients suffering from depression, anxi
`ety and other psychiatric disorders. Moreover, it would be
`useful in patients taking Compound 1, while being treated by
`different healthcare providers without disclosing all of their
`medications to each of them. It would also be beneficial in
`patients who are using drugs of abuse while taking Com
`pound 1 without the knowledge of their physician.
`25
`The decreased substrate efficiency for CYP2D6 at the
`methylenedioxy portion of the benzodioxol ring demon
`strated by the compounds of this invention will provide the
`further benefit of reducing inter-patient pharmacokinetic
`variability observed for Compound 1.
`The compounds of the present invention comprising addi
`tional deuterium for hydrogen replacement at the methylene
`dioxy carbon demonstrate the added benefit of reduced
`metabolism by other cytochrome P450 enzymes. This is
`important for poor metabolizers of Compound 1, wherein the
`main metabolic pattern of Compound 1 proceeds largely by
`Scission of the benzodioxol ring, likely due to oxidative attack
`by another cytochrome enzyme. Also, a relatively minor
`amount of ring Scission (complete cleavage of the benzo
`dioxol ring, forming 4-(4-fluorophenyl)-3-hydroxymethylpi
`peridine) observed in normal metabolizers, which could
`result from oxidation of the methylene carbon attached to the
`piperidine ring, may become more predominant if the benzo
`dioxol ring is metabolically stabilized. Therefore, com
`pounds of this invention that are deuterated at that carbon will
`45
`also be beneficial to the clearance rate of the compound.
`The compounds of this invention, and compositions com
`prising them, are useful for treating or lessening the severity
`of disorders characterized by reduced serotonin-dependent
`neurological activity. Preferred applications for compounds
`of formula I include methods of use in treating depression,
`anxiety, stress, phobias, panic, dysphoria, and other psychi
`atric disorders, and pain.
`The compounds and compositions of this invention are also
`useful as analytical reagents for determining the concentra
`tion of the Compound 1 in solution. “Compound 1 as used
`herein refers to a compound wherein all hydrogen and all
`carbon atoms are present at their natural isotopic abundance
`percentages. It is recognized that some variation of natural
`isotopic abundance occurs depending upon the origin of
`chemical materials. The concentration of naturally abundant
`stable hydrogen and carbon isotopes, notwithstanding this
`variation, is Small and immaterial with respect to the degree of
`stable isotopic Substitution of compounds of this invention.
`See for instance Wada E and HanbaY. Seikagaku 1994.66:15:
`Ganes L. Zet. al., Comp. Biochem. Physiol. A Mol. Integr.
`Physiol. 1998 119: 725.
`
`55
`
`8
`Incorporation of deuterium in place of hydrogen is known
`in certain instances to have significant effects on the physi
`ological and pharmacological activities of the Substituted
`compound. For instance, N-nitrosamines Substituted with
`deuterium can display increased, decreased, or unchanged
`carcinogenicity depending on where in the compound hydro
`gen is replaced with deuterium and on the identity of the
`compound to which Substitutions are made (Lijinsky Wet. al.,
`Food Cosmet. Toxicol. 1982 20:393: Lijinsky Wet. al., JCNI
`1982 69: 1127). Similarly, both increases and decreases in
`bacterial mutagenicity of deuterium-substituted aza-amino
`acids are known, depending on the identity of the amino acid
`derivative and position of substitution (Mangold J B et. al.,
`Mutation Res. 1994.308:33). Reduced hepatotoxicity of cer
`tain deuterium-substituted compounds is known (Gordon W
`Pet. al., Drug Metab. Dispos. 1987 15:589: Thompson DC
`et. al., Chem. Biol. Interact. 1996 101: 1). Deuterium substi
`tution can affect compounds odors (Turin L. Chem. Senses
`1996 21: 773) and plasma protein binding (Echmann MLet.
`al., J. Pharm. Sci. 1962 51: 66; Cherrah Y. et al., Biomed.
`Environm. Mass Spectrom. 1987 14: 653; Cherrah Y. et al.,
`Biochem. Pharmacol. 198837: 1311). Changes in the biodis
`tribution and clearance of certain deuterium-Substituted com
`pounds suggests changes in their recognition by active trans
`port mechanisms (Zello GA et al., Metabolism 199443: 487;
`Gately SJet. al., J. Nucl. Med. 1986 27:388; Wade D, Chem.
`Biol. Interact. 1999 117: 191).
`Replacement of hydrogen with deuterium at sites Subject to
`oxidative metabolism by, for instance, heme proteins such as
`cytochrome P450 and peroxidase enzymes, is known in cer
`tain, but not all, cases to produce a significant reduction in the
`rate of metabolism due to the primary isotope effect of break
`ing the C H versus C H bond (see, e.g., Guengerich FP
`et. al., J. Biol. Chem. 2002 277: 33711; Kraus, J A and
`Guengerich, F P. J. Biol. Chem. 2005 280: 19496; Mitchell K
`Het. al., Proc. Natl. Acad. Sci. USA 2003 109:3784; Nelson
`SD and Trager WF, Drug Metab. Dispos. 200331: 1481; Hall
`L R and Harzlik R. P. J. Biol. Chem. 1990 265: 12349; Oka
`Zaki 0 and Guengerich F P. J. Biol. Chem. 268, 1546; Iwar
`nura Set. al., J. Pharmacobio-Dyn. 1987 10: 229). If the
`C-H bond breaking step is rate-limiting, a Substantial iso
`tope effect can be observed. If other steps determine the
`overall rate of reaction, the isotope effect may be insubstan
`tial. In cases where a rate-limiting step of a reaction involves
`rehybridization of the attached carbon from sp2 to sp3, deu
`terium Substitution often creates a negative isotope effect,
`speeding up the reaction rate. Introducing deuterium into a
`compound at a site subject to enzymatic oxidation does not
`predictably produce a significant pharmacokinetic change.
`See for instance Mamada Ket. al., Drug Metab. Dispos. 1986
`14: 509; Streeter A Jet. al., Arch. Toxicol. 1990 64: 109:
`Morgan D Set. al., Int. Arch. Occup. Environ. Health 1993
`65(1 Suppl.): S139.
`Although incorporation of deuterium into specific organic
`compounds can change their pharmacological properties,
`general exposure to and incorporation of deuterium is safe
`within levels potentially achieved by use of compounds of
`this invention as medicaments. For instance, the weight per
`centage of hydrogen in a mammal (approximately 9-10%)
`and natural abundance of deuterium (approximately 0.015%)
`indicates, for instance that an average adult US male normally
`contains approximately 1.2 grams of deuterium (see e.g.
`Harper V Wet. al. “Review of Physiological Chemistry' 16"
`Edition, 1977 Lange Medical Publications: Ogden C Let. al.
`CDC Adv. Data 2004 347: 1; www.cdc.gov/nchs/data/ad/
`ad347.pdf).
`
`30
`
`35
`
`40
`
`50
`
`60
`
`65
`
`Auspex Exhibit 2007
`Apotex v. Auspex
`IPR2021-01507
`Page 5
`
`

`

`9
`Furthermore, replacement of up to about 15% of normal
`hydrogen with deuterium has been effected and maintained
`for a period of days to weeks in mammals, including rodents
`and dogs, with minimal observed adverse effects (Czajka D
`Mand Finkel AJ, Ann. N.Y. Acad. Sci. 1960 84: 770; Thom
`son J. F. Ann. N.Y. Acad. Sci. 196084: 736: Czaka D Met. al.,
`Am. J. Physiol. 1961 201: 357). Higher deuterium concen
`trations, usually in excess of 20%, can be toxic in animals.
`However, acute replacement of as high as 15%-23% of the
`hydrogen in humans fluids with deuterium was found not to
`cause toxicity (Blagojevic Net. al. in “Dosimetry & Treat
`ment Planning for Neutron Capture Therapy'. Zamenhof R,
`Solares G and Harling O Eds. 1994. Advanced Medical Pub
`lishing, Madison Wis. pp. 125-134.). These authors report a
`clinical protocol in their practice involving oral administra
`tion of up to 1 liter per day of deuterated water (DO) for up
`to 5 days, followed by intravenous administration of 4 liters of
`deuterated waterprior to radiation procedures; this deuterated
`water is readily incorporated throughout the body beyond the
`fluid compartment, including in glucose and glycogen, fats,
`and cholesterol and thus cell walls (e.g. see Diabetes Metab.
`1997 23:251).
`In a 70kg human, 15% replacement of the hydrogen in the
`fluid compartment with deuterium corresponds to incorpora
`tion of approximately 1 kg of deuterium or the equivalent of
`approximately 5 kg of deuterated water. These quantities are
`orders of magnitude beyond the conceived level of adminis
`tration of any of the deuterium-containing compounds of this
`invention.
`Deuterium tracers including as deuterium-labeled drugs
`and doses, in Some cases repeatedly, of thousands to tens of
`thousands of milligrams of deuterated water, are also used in
`healthy humans of all ages including neonates and pregnant
`women, without reported incident (e.g. Pons G and Rey E.
`Pediatrics 1999 104: 633: Coward WAet. al., Lancet 1979 7:
`13; Schwarcz HP, Control. Clin. Trials 19845(4 Suppl): 573:
`Eckhardt C Let. al. Obes. Res. 2003 11: 1553; Rodewald LE
`et. al., J. Pediatr. 1989 114: 885; Butte N F et. al., Br. J. Nutr.
`1991 65: 3; MacLennan A Het. al., Am. J. Obstet. Gynecol.
`1981 139:948). Thus, it is clear that any deuterium released,
`for instance during the metabolism of compounds of this
`invention, poses no health risk.
`The compounds of this invention display decreased mecha
`nism-based inactivation of CYP2D6 than Compound 1 and
`therefore display a reduced rate of oxidative metabolism and
`decreased mechanism-based inactivation of CYP2D6. This
`reduces the extent of undesirable metabolic drug-drug inter
`actions observed with Compound 1, reducing the need for
`dose adjustments of other drugs taken by patients treated with
`these agents.
`The altered properties of the compounds of this invention
`will not obliterate their ability to bind to their protein target.
`This is because such binding is primarily dependent upon
`non-covalent binding between the protein and the inhibitor
`which may be impacted both positively and negatively by
`isotopic Substitution, depending on the specific Substitution
`involved, and any negative effects that a heavy atom of this
`invention may have on the highly optimized non-covalent
`binding between compounds

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