WO2011008597A1 - Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors - Google Patents

Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors Download PDF

Info

Publication number
WO2011008597A1
WO2011008597A1 PCT/US2010/041122 US2010041122W WO2011008597A1 WO 2011008597 A1 WO2011008597 A1 WO 2011008597A1 US 2010041122 W US2010041122 W US 2010041122W WO 2011008597 A1 WO2011008597 A1 WO 2011008597A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
alkoxy
heteroaryl
cycloalkyl
Prior art date
Application number
PCT/US2010/041122
Other languages
French (fr)
Inventor
Ginny D. Ho
William M. Seganish
Deen B. Tulshian
Cornelis Marius Timmers
Rachel Deborah Van Rijn
Hubert Jan Jozef Loozen
Original Assignee
Schering Corporation
N. V. Organon
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Schering Corporation, N. V. Organon filed Critical Schering Corporation
Publication of WO2011008597A1 publication Critical patent/WO2011008597A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the present invention relates to dihydroimidazoisoquinoline derivatives, to the use of the compounds as phosphodiesterase 10 (PDElO) inhibitors for the treatment of PDElO- modulated disorders, and to pharmaceutical compositions comprising the compounds.
  • PDElO phosphodiesterase 10
  • PDElO is known to be a dual cAMP/cGMP phosphodiesterase; see, for example, Kehler et al, "The potential therapeutic use of phosphodiesterase 10 inhibitors", Expert Opin. Ther. Patents (2007) 17(2): 147-158.
  • PDElO is expressed at high levels in all striatal medium spiny neurons (MSNs), but is expressed at much lower or undetectable levels elsewhere in the brain and periphery.
  • MSNs striatal medium spiny neurons
  • PDElO inhibition will mimic D2 dopamine receptor antagonism in the indirect striatopallidal output pathway and will increase the activity of the direct striatonigral output pathway, thus more fully normalizing the reduced striatal output that characterizes schizophrenia.
  • PDElO inhibition should improve the cognitive dysfunction that characterizes schizophrenia.
  • the discrete localization of PDElO should lead to an improved side effect profile: typical side effects include extrapyramidal syndrome, diabetes, weight gain, hyperprolactinemia, sedation and QT C prolongation.
  • PDElO inhibitors have also been reported to be useful in treating in other CNS (central nervous system) disorders such as psychosis, cognitive disorders (such as
  • Alzheimer's disease bipolar disorder
  • depression depression
  • diet-induced obesity diabetes and metabolic syndrome.
  • Papaverine has been identified as a PDElO inhibitor, and has been shown to be effective in animal models of schizophrenia.
  • triazolopyridines and derivatives thereof are disclosed in WO 2007/113226.
  • Imidazo- and triazolopyridine keratin dyeing compounds are disclosed in US 2005/0229333.
  • Imidazo- and triazolopyridines useful in treating diseases associated with 11-beta-hydroxysteroid dehydrogenase type I are disclosed in WO 2006/135795.
  • Imidazopyridines having phosphatidylinositol 3 kinase inhibitory activity are disclosed in EP 1277754.
  • the present invention provides a novel class of substituted dihydroimidazoisoquinoline PDElO inhibitor compounds and derivatives thereof represented by Formula I, below, pharmaceutical compositions comprising one or more of said compounds of Formula I, and methods of treating PDElO inhibitor mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome using said compounds of Formula I or pharmaceutical compositions comprising it.
  • CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome using said compounds of Formula I or pharmaceutical compositions comprising it.
  • Novel compounds of Formula I of the invention have the structural formula:
  • R 1 is R 12 -Ci_ 6 alkyl, C 3-10 cycloalkyl, C 1-6 alkoxyalkyl-, -CF 3 or -SF 5 ;
  • R 2 is H, OH, alkoxy, Ci_ 6 alkoxyalkoxy, -OCF 3 , -OSF 5 , -O- C 3 -iocycloalkyl, or -O(CH 2 ) n R;
  • n 0, 1 or 2;
  • R 3 is H, R 12 -Ci_ 6 alkyl, Ci_ 6 alkoxy, C 3 _iocycloalkyl, Ci_ 6 alkoxyalkoxy, OH, -CF 3 , - OCF 3 , -SF 5 , -OSF 5 , halo, -O- C 3 _i 0 cycloalkyl, benzyloxy, -N(R 10 XR 11 ) or CN;
  • R 4 , and R 5 , R 6 , R 7 are independently selected from the group consisting of H, R 12 - Ci_ ⁇ alkyl, R 13 - C ⁇ -ioaryl and R 13 - C 5 _ioheteroaryl; or R 4 and R 5 , together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring; or R 6 and R 7 , together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring;
  • R 8 is H, R 12 - Ci_ 6 alkyl, C 3 -i 0 cycloalkyl, Ci_ 6 alkoxy, Ci_ 6 alkoxyalkoxy, OH, -CF 3 , - OCF 3 , -SF 5 , -OSF 5 , halo, -O- C 3 _i 0 cycloalkyl, benzyloxy, -N(R 17 ) 2 , CN, R 13 -C 6 _i 0 aryl or R 13 - C 5 _ioheteroaryl;
  • R 9 is R 12 -Ci_ 6 alkyl, R 12 -C 3 _i 0 cycloalkyl, R 13 -C 6 -i 0 aryl, R 13 -C 5 _i 0 heteroaryl or
  • R is selected from the group consisting of
  • R 10 and R 11 are independently selected from the group consisting of H,
  • R 10 and R 11 together with the nitrogen to which they are attached, form a 5 to 7 membered ring, wherein one carbon ring member not adjacent to the nitrogen can optionally be replaced by -O-, -S- or -NR 14 -;
  • R 12 is 1 or 2 substituents independently selected from the group consisting of H, OH,
  • R 13 is 1 or 2 substituents selected from the group consisting of H, halo, Ci_6alkyl, C 2 . galkenyl, OH, hydroxyalkyl, Ci_ 6 alkoxy, CN, -CF 3 , -OCF 3 , -SF 5 , -OSF 5 , -N(R 17 ) 2 ,
  • each R 14 is independently selected from the group consisting of H, Ci_ 6 alkyl, C 3 . iocycloalkyl and C ⁇ -ioarylalkyl;
  • R 15 is 1 or 2 substituents independently selected from the group consisting of H, halo, Ci_ 6 alkyl, Ci_ 6 alkoxy, -CF 3 , C 3 _iocycloalkyl, Ci_ 6 alkoxyalkoxy, OH, hydroxyalkyl, -OCF 3 , -O- C 3 _iocycloalkyl, benzyloxy, -C(O)O C h alky!, -O- Ci_ 6 alkyl-CO 2 H, -C(O)N(R 10 ) 2 , - N(R 10 ) 2 , - Ci_ 6 alkyl-N(R 10 ) 2 , -NR 10 -C(O)N(R 10 ) 2 , -N(R 10 )C(O)O Ci_ 6 alkyl, -N(R 10 )SO 2 - C 1 . ealkyl, phenyl, CN, -SF 5 , -OS
  • R 16 is H, Ci_ 6 alkyl, Ci_ 6 alkoxy
  • each R 17 is independently selected from the group consisting of H, d_ 6 alkyl, C 3 . l ocycloalkyl, C 3 _ioheterocycloalkyl and benzyl.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of Formula I or a pharmaceutically acceptable salt thereof in a
  • the present invention relates to a method of treating PDElO mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome comprising administering a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof to a mammal in need of such treatment.
  • CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome
  • administering a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof to a mammal in need of such treatment.
  • the invention relates to a method of treating PDElO mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome comprising administering to a mammal in need of such treatment a pharmaceutical composition comprising a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome
  • a pharmaceutical composition comprising a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • R 1 is alkyl, preferably methyl.
  • R 2 is H, OH, alkoxy (preferably methoxy or ethoxy) or -O(CH 2 ) n R, preferably -O-CH 2 -quinolyl. More preferably, R 2 is methoxy.
  • -OR 1 and R 2 together form -0-CH 2 -O-.
  • R 3 is H or alkoxy (preferably methyoxy). More preferably, R 3 is H.
  • R 4 , R 5 , R 6 and R 7 are independently H or alkyl; preferably R 4 , R 5 , R 6 and R 7 are each H.
  • R 8 is H, alkyl or aryl; preferably, R 8 is H, alkyl (preferably methyl) or phenyl; more preferably, R 8 is H or methyl.
  • R 9 is R 12 -cycloalkyl, R 13 - C ⁇ -ioaryl or R 13 - C 5 -ioheteroaryl, preferably R 13 - C ⁇ -ioaryl or R 13 - Cs-ioheteroaryl.
  • a preferred R 12 - cycloalkyl group is cyclohexyl.
  • R 13 - C ⁇ -ioaryl groups are R 13 -phenyl, wherein R 13 is H, 1 or 2 alkoxy groups (preferably methoxy), 1 or 2 halo atoms, -OCF 3 , -O-SO 2 -alkyl (preferably -O-SO 2 -methyl), Cs-ioheterocycloalkyl, (preferably
  • R 14 is H or methyl
  • -N(R 17 ) 2 preferably Preferred R 13 -heteroaryl groups are those wherein the heteroaryl portion is thiazolyl, oxazolyl, imidazolyl, pyridyl, pyrimidinyl or quinolyl; more preferred heteroaryl groups are thiazolyl and pyridyl.
  • R 13 substituents on the carbon atoms of the heteroaryl groups are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl (preferably cyclopropyl), CN, alkenyl (preferably vinyl), hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl),-aminobenzyl, heterocycloalkyl (preferably morpholinyl, pyrrolidinyl or piperidinyl), -alkyl-N(R 10 )(R ⁇ ) (preferably -CH 2 -morpholinyl or -CH 2 -homomorpholinyl), -C(O)N(R 1 ⁇ (R 11 ) (preferably -C(O)-N(alkyl) 2 , -C(O)- morpholinyl,
  • heteroaryl (wherein heteroaryl is preferably pyridyl, imidazolyl, pyrimidinyl or indolyl).
  • Preferred R 13 substituents on the nitrogen atoms of the heteroaryl groups are H, methyl and benzyl, more preferably H.
  • R 9 is R 13 -thiazolyl
  • preferred R 13 substituents are H, 1 or 2 halo atoms, hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl), C 3 _ioheterocycloalkyl, (preferably morpholinyl), -alkyl-N(R 10 )(R ⁇ ) (preferably -CH 2 -morpholinyl or -CH 2 - homomorpholinyl), -C(O)N(R 1 ⁇ (R 11 ) (preferably -C(O)-N(alkyl) 2 , -C(O)-morpholinyl, - C(O)-homomorpholinyl, -C(O)NH-benzyl or
  • R 9 is R 13 -pyridyl
  • preferred R 13 substituents are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl
  • heteroaryl is preferably pyridyl, imidazolyl, pyrimidyl or indolyl.
  • R 1 is alkyl
  • R 2 is H, OH, alkoxy or
  • R 3 is H or alkoxy
  • R 4 , R 5 , R 6 and R 7 are independently H or alkyl
  • R 8 is H, alkyl or aryl.
  • R 1 is alkyl; R 2 is alkoxy; R 3 is H; R 4 , R 5 , R 6 and R 7 are independently H or alkyl; and R 8 is H or alkyl.
  • R 1 is alkyl, preferably methyl; R 2 is alkoxy
  • R 3 is H or alkoxy (preferably methoxy);
  • R 4 , R 5 , R 6 and R 7 are independently H or alkyl, preferably H; and R 8 is H or alkyl, preferably H or methyl.
  • R 9 is preferably R 13 -phenyl; or R 13 -heteroaryl, wherein heteroaryl is preferably thiazolyl or pyridyl; preferred R 13 substituents are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl (preferably cyclopropyl), CN, alkenyl (preferably vinyl), hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl), -aminobenzyl, heterocycloalkyl (preferably morpholinyl, pyrrolidinyl or piperidinyl), -alkyl-N(R 10 )(R ⁇ ) (preferably -CH 2 -morpholinyl or -CH 2 -homomorpholinyl), -C(O)N(R 1 ⁇ (R 11 ) (preferably -C(O)-N(alkyl) (preferably
  • R 1 is methyl, R 2 is methoxy, R 3 to R 7 are H, R 9 is shown by the structure, and R 8 is as defined:
  • R 8 H Preferred compounds of Formula I are those in Examples IA, IB, ID, IE, IH, IN, IP, IQ, IR, IS, IU, IW, IY, IAA, ICC, IFF, IHH, HA, HC, HD, HE, HF, HG, HH, IU, HK, HL, HM, UN, HQ, HR, IIS, IIIA, IIIB, IUC, HID, HIE, IVA, IVB, IVC, IVD, VD, VIA, VIB, VIC, VID, VIE, VIF, VIIA, VIIB, VIIC, VIID, VIIE, VIIF, IX, X, XI, XII, XIV, XXII, XIII, and XXIV.
  • More preferred compounds of Formula I are those in Examples ID, IP, IW, ICC, IFF, HD, HG, IU, HL, HQ, HR, IIIB, IUC, HID, IVD, VIA, VIB, VIC, VID, VIE, VIIA, VIIC, VIID, VIIE, XXII, and XXIV. As used herein, the following terms are as defined below unless otherwise indicated:
  • Mammal means humans and other mammalian animals.
  • alkyl refers to “alkyl” as well as the “alkyl” portions of "hydroxyalkyl”, “haloalkyl”, “alkoxy”, etc.
  • Alkyl means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain.
  • Hydroxyalkyl represents an alkyl group as defined substituted by 1 to 3 hydroxy groups. The bond to the parent is through the alkyl group.
  • Alkoxy means an alkyl-O- group in which the alkyl group is as previously described.
  • suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy.
  • the bond to the parent moiety is through the ether oxygen.
  • Halogen represents fluoro, chloro, bromo and iodo.
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • stable compound or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • isolated or “in isolated form” for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof.
  • purified or “in purified form” for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
  • Cycloalkyl means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 3 to about 7 carbon atoms.
  • suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • suitable multicyclic cycloalkyls include 1-decalinyl, norbornyl, adamantly and the like.
  • Heterocycloalkyl means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system.
  • Preferred heterocycloalkyls contain about 5 to about 6 ring atoms.
  • the prefix aza, oxa or thia before the heterocycloalkyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom.
  • Any - NH in a heterocycloalkyls ring may exist protected such as, for example, as an -N(Boc), - N(CBz), -N(Tos) group and the like; such protections are also considered part of this invention.
  • the nitrogen or sulfur atom of the heterocycloalkyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide.
  • the heterocycloalkyl ring can be joined to the parent moiety by a ring carbon atom or a ring nitrogen atom.
  • Non-limiting examples of suitable monocyclic heterocycloalkyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1 ,4-dioxanyl, tetrahydrofuranyl,
  • Aryl means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms.
  • suitable aryl groups include phenyl and naphthyl.
  • Heteroaryl means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination.
  • Preferred heteroaryls contain about 5 to about 6 ring atoms.
  • the prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom.
  • a nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide.
  • Non-limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N- substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[l,2-a]pyridinyl, imidazo[2,l-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quina
  • heteroaryl also refers to partially saturated heteroaryl moieties such as, for example, tetrahydroisoquinolyl, tetrahydroquinolyl and the like.
  • Aralkyl or “arylalkyl” means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2-phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
  • hetero-atom containing ring systems of this invention there are no hydroxyl, halo, amino or thio groups on ring carbon atoms adjacent to a N, O or S.
  • ring carbon atoms adjacent to a N, O or S there are no hydroxyl, halo, amino or thio groups on ring carbon atoms adjacent to a N, O or S.
  • protecting groups When a functional group in a compound is termed "protected”, this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene et al, Protective Groups in Organic Synthesis (1991), Wiley, New York.
  • variable e.g., alkyl, halo, etc.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • Prodrugs, solvates and co-crystals of the compounds of the invention are also contemplated herein.
  • the term "prodrug”, as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof.
  • a discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 1_4 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
  • Solvate means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • Solvate encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like.
  • “Hydrate” is a solvate wherein the solvent molecule is H 2 O.
  • a co-crystal is a crystalline superstructure formed by combining an active
  • Co-crystals are often made between a dicarboxlyic acid such as fumaric acid, succinic acid etc. and a basic amine such as the one represented by compound I or II of this invention in different proportions depending on the nature of the co-crystal. (Rmenar, J. F. et. al. JAm. Chem. Soc. 2003, 125, 8456).
  • Effective amount or “therapeutically effective amount” is meant to describe an amount of compound or a composition of the present invention effective as PDElO inhibitors and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
  • the compounds of Formula I can form salts which are also within the scope of this invention.
  • Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated.
  • the term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases.
  • zwitterions inner salts may be formed and are included within the term "salt(s)" as used herein.
  • Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Exemplary acid addition salts include acetates, ascorbates, benzoates,
  • benzenesulfonates bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates,
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like.
  • Basic nitrogen- containing groups may be quaternized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
  • lower alkyl halides e.g. methyl, ethyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates e.g. dimethyl, diethyl, and dibutyl sulfates
  • long chain halides e.g. decyl, lauryl, and stearyl chlorides
  • All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds including those of the salts, solvates, co-crystals and prodrugs of the compounds as well as the salts and solvates, co-crystals of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl).
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected,
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the use of the terms "salt”, “solvate” "prodrug” and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds.
  • Isomers can be prepared using conventional techniques, either by reacting optically pure or optically enriched starting materials or by separating isomers of a compound of Formula I. Isomers may also include geometric isomers, e.g., when a double bond is present.
  • the term "at least one compound of Formula I" means that one to three different compounds of Formula I may be used in a pharmaceutical composition or method of treatment. Preferably one compound of Formula I is used.
  • the present invention also embraces isotopically-labelled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 Cl, respectively.
  • Certain isotopically-labelled compounds of Formula I are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances.
  • Isotopically labelled compounds of Formula I can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples hereinbelow, by substituting an appropriate isotopically labelled reagent for a non- isotopically labelled reagent.
  • Compounds of Formula I are prepared by methods known in the art. Typical procedures for preparing the compounds are shown in reaction Schemes 1 and 2; in the schemes, the exemplified compound of Formula I is that wherein R 1 is methyl, R 2 is methoxy, R 3 -R 7 are H and R 8 is H (Scheme 1) or methyl (Scheme 2).
  • Scheme 1 is methyl, R 2 is methoxy, R 3 -R 7 are H and R 8 is H (Scheme 1) or methyl (Scheme 2).
  • Formamide 2 (21 g) was dissolved in dry THF 140 (mL) and cooled to 0 0 C. Et 3 N (70 mL) was added, followed by POCl 3 (10.2 mL in 25 mL of THF) dropwise over 10 min. The reaction was stirred at 0 0 C for 1 h. Ice water (500 mL) was added and the reaction was stirred for 10 min. The mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried (MgSO 4 ) and evaporated. The oily residue was purified via column chromatography (25-50% EtOAc/hexanes) to give a colorless oil (17 g), which solidified on standing to a white solid. MS (ESI) 192 (M+H).
  • Example VI-A was prepared according to the procedure outlined for Example IV-A above except that bromide H-J was used in place of bromide H-H.
  • the crude reaction residue was purified via column chromatography (0-5% MeOH-NH 3 /DCM) to give VI-A as a white solid.
  • Example VII-A was prepared according to the procedure outlined for Example V-A above with the exception that bromide H-J was used in place of bromide H-H.
  • the crude reaction residue was purified via column chromatography (0-5% Me0H-NH 3 /DCM) to give VII-A as a white solid.
  • Example IX was prepared according to the procedure outlined for Example VIII above except that bromide H-J (300 mg) was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-10% MeOH-NHs/DCM) to give IX as a white solid (240 mg). LCMS (ESI) 2.84 min (347, M+H).
  • Example XI was prepared according to the procedure outlined for Example X above except that bromide H-J (200 mg) was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-5% MeOH-NHs/DCM) to give XI as a white solid (140 mg). LCMS (ESI) 1.55 min (348, M+H).
  • Example XIII was prepared according to the procedure outlined for Example XII above except that alkene XI (50 mg) was used in place of alkene X. The reaction mixture was filtered through celite and the solvent evaporated to give Example XIII as a white solid (50 mg). LCMS (ESI) 2.55 min (350, M+H).
  • THP ether 9 (73 g) was dissolved in dry THF (700 mL). The solution was cooled to - 78 0 C and stirred at -78 0 C for 20 min. A solution of nBuLi in hexanes (2.5 M, 127 mL) was added slowly. After the addition was complete, the reaction was allowed to stir at -78 0 C for 30 min. B(OMe) 3 (35 mL) was added and the reaction was allowed to warm to ambient temperature over 2 h. IM HCl (600 mL) was added and the reaction vigorously stirred overnight. The layers were separated, and the aqueous layer was extracted with DCM. The combined organic layers were evaporated to a brown oil, and re-dissolved in DCM.
  • Alcohol 13 (19 g) was dissolved in DCM (75 mL) and cooled to 0 0 C.
  • Et 3 N (28 mL) and DMAP (1.5 g) were added, followed by/?-toluenesulfonyl chloride (TsCl) (35 g).
  • TsCl trimethyl methoxylate
  • the reaction was allowed to slowly warm to RT over 2 h.
  • the reaction was cooled to 0 0 C again and water (50 mL) was added and the reaction was vigorously stirred for 1 h at 0 0 C.
  • the layers were separated, and the aqueous layer was extracted with DCM.
  • the combine organic extracts were evaporated and dissolved in acetone (75 mL) and IM NaOH (100 mL) and stirred at ambient temperature overnight.
  • Step 7 Cyclized compound 14 (11 g) was dissolved in DMF (250 niL) and cooled to 0 °C. NBS (8.8 g) was added in one portion and the reaction was stirred at 0 0 C for 2 h. The reaction was diluted with EtOAc and washed with water. The organic phase was dried (MgSO 4 ), and evaporated. The residue was purified via column chromatography (0-10% MeOH/DCM) to give bromide 15 as a white solid (14 g). LCMS (ESI) 2.78 min (338, M+H). Step 8:
  • Example XVIII was prepared according to the general procedure outlined for Example II, except 3-(3-methoxy-phenyl)-propionitrile was used in place of 3-(3,4- dimethoxy-phenyl)-propionitrile (3).
  • Example XIX was prepared according to the general procedure outlined for Example II, except 3-benzo[l,3]dioxol-5-yl-propionitrile was used in place of 3-(3,4-dimethoxy- phenyl)-propionitrile (3).
  • Example XXI was prepared according to the general procedure outlined for Example II, except ammonium benzoate was used in place of ammonium acetate.
  • a typical recommended dosage regimen can range from about 10 mg/dose to about 100 mg/dose, preferably about 10 to about 50 mg/dose, and more preferably about 20 to about 25 mg/dose.
  • inert, pharmaceutically acceptable carriers can be either solid or liquid.
  • Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and
  • the powders and tablets may be comprised of from about 5 to about 95 percent active ingredient.
  • Suitable solid carriers are known in the art, e.g. magnesium carbonate, magnesium stearate, talc, sugar or lactose. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration. Examples of
  • Liquid form preparations include solutions, suspensions and emulsions.
  • solutions include solutions, suspensions and emulsions.
  • water or water-propylene glycol solutions for parenteral injection or addition of sweeteners and opacif ⁇ ers for oral solutions, suspensions and emulsions.
  • Liquid form preparations may also include solutions for intranasal administration.
  • Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas, e.g. nitrogen. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
  • the compounds of the invention may also be deliverable transdermally.
  • the transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
  • the compound is administered orally.
  • the pharmaceutical preparation is in a unit dosage form.
  • the preparation is subdivided into suitably sized unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
  • the actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage regimen for a particular situation is within the skill of the art. For convenience, the total daily dosage may be divided and administered in portions during the day as required.
  • a typical recommended daily dosage regimen for oral administration can range from about 1 mg/day to about 300 mg/day, preferably 1 mg/day to 75 mg/day, in two to four divided doses.
  • the activity of the compounds of Formula I can be determined by the following procedures.
  • PDElOAl Human recombinant PDElOAl was purchased from BPS Bioscience, Inc. The reaction mixture contained PDElOAl ( 0.02 nM), 10 nM [ 3 H]cAMP ([5 ',8- 3 H] Adenosine 3 ',5 '-cyclic phosphate, ammonium salt], Amersham) and various concentrations of compound in 50 mM Tris-HCl, pH 7.5, 8.3 mM MgCl 2 , 17 mM EGTA and 0.2% bovine serum albumen in a total volume of 30 1.
  • the assay was initiated with the addition of substrate and was allowed to proceed for 30 minutes at room temperature before being stopped by the addition of 300 g yttrium SPA PDE beads.
  • the reaction mixtures were thoroughly mixed, and the beads were allowed to settle for 30 minutes.
  • the plates were then counted in a TopCount scintillation counter.
  • Ki values were determined as described by Cheng and Prusoff (1973).
  • VIE VIF, VIIA, VIIB, VIIC, VIID, VIIE, VIIF, IX, X, XI, XII, XIV, XXII, XXIII, and
  • Ki values of less than 10 nM: Examples ID, IP, IW, ICC, IFF, HD, HG, IU, HL, HQ, HR, IIIB, IIIC, HID, IVD, VIA,

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to dihydroimidazoisoquinolines and derivatives thereof, the use of the compounds as phosphodiesterase 10 (PDE10) inhibitors for the treatment of PDE10-modulated disorders, to pharmaceutical compositions comprising the compounds.

Description

DIHYDROIMIDAZOISOQUINOLINE DERIVATIVES
USEFUL AS PDElO INHIBITORS Field of the Invention
The present invention relates to dihydroimidazoisoquinoline derivatives, to the use of the compounds as phosphodiesterase 10 (PDElO) inhibitors for the treatment of PDElO- modulated disorders, and to pharmaceutical compositions comprising the compounds. Background of the Invention
PDElO is known to be a dual cAMP/cGMP phosphodiesterase; see, for example, Kehler et al, "The potential therapeutic use of phosphodiesterase 10 inhibitors", Expert Opin. Ther. Patents (2007) 17(2): 147-158.
PDElO is expressed at high levels in all striatal medium spiny neurons (MSNs), but is expressed at much lower or undetectable levels elsewhere in the brain and periphery. By increasing cAMP and cGMP levels in all striatal MSNs, PDElO inhibition will mimic D2 dopamine receptor antagonism in the indirect striatopallidal output pathway and will increase the activity of the direct striatonigral output pathway, thus more fully normalizing the reduced striatal output that characterizes schizophrenia. By increasing corticostriatal transmission, PDElO inhibition should improve the cognitive dysfunction that characterizes schizophrenia. Furthermore, the discrete localization of PDElO should lead to an improved side effect profile: typical side effects include extrapyramidal syndrome, diabetes, weight gain, hyperprolactinemia, sedation and QTC prolongation.
PDElO inhibitors have also been reported to be useful in treating in other CNS (central nervous system) disorders such as psychosis, cognitive disorders (such as
Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome.
Papaverine has been identified as a PDElO inhibitor, and has been shown to be effective in animal models of schizophrenia.
8-Substituted triazolopyridine phosphodiesterase 4 inhbitors useful in the treatment of skin diseases are disclosed in WO 2008/125111. 6,7-Di-aryl/heteroaryl-substituted triazolopyridines are disclosed in US 2006/0287324. 6-Aminomethyl-substituted
triazolopyridines and derivatives thereof are disclosed in WO 2007/113226. Imidazo- and triazolopyridine keratin dyeing compounds are disclosed in US 2005/0229333. Imidazo- and triazolopyridines useful in treating diseases associated with 11-beta-hydroxysteroid dehydrogenase type I are disclosed in WO 2006/135795. Imidazopyridines having phosphatidylinositol 3 kinase inhibitory activity are disclosed in EP 1277754.
Pharmaceutically useful imidazoisoquinolines are disclosed in US Patent 6,159,985 and WO 00/69860.
Summary of the Invention
In its several embodiments, the present invention provides a novel class of substituted dihydroimidazoisoquinoline PDElO inhibitor compounds and derivatives thereof represented by Formula I, below, pharmaceutical compositions comprising one or more of said compounds of Formula I, and methods of treating PDElO inhibitor mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome using said compounds of Formula I or pharmaceutical compositions comprising it.
Novel compounds of Formula I of the invention have the structural formula:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 is R12-Ci_6alkyl, C3-10 cycloalkyl, C1-6alkoxyalkyl-, -CF3 or -SF5;
R2 is H, OH, alkoxy, Ci_6alkoxyalkoxy, -OCF3, -OSF5, -O- C3-iocycloalkyl, or -O(CH2)nR;
or -OR1 and -R2 together are -O-(CH2)P-O-, wherein p is 1 or 2, and form a ring with the carbon atoms to which they are attached;
n is 0, 1 or 2;
R3 is H, R12-Ci_6alkyl, Ci_6alkoxy, C3_iocycloalkyl, Ci_6alkoxyalkoxy, OH, -CF3, - OCF3, -SF5, -OSF5, halo, -O- C3_i0cycloalkyl, benzyloxy, -N(R10XR11) or CN;
R4, and R5, R6, R7 are independently selected from the group consisting of H, R12- Ci_ βalkyl, R13- Cβ-ioaryl and R13- C5_ioheteroaryl; or R4 and R5, together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring; or R6 and R7, together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring;
R8 is H, R12- Ci_6alkyl, C3-i0cycloalkyl, Ci_6alkoxy, Ci_6alkoxyalkoxy, OH, -CF3, - OCF3, -SF5, -OSF5, halo, -O- C3_i0cycloalkyl, benzyloxy, -N(R17)2, CN, R13-C6_i0aryl or R13- C5_ioheteroaryl;
R9 is R12-Ci_6alkyl, R12-C3_i0cycloalkyl, R13-C6-i0aryl, R13-C5_i0heteroaryl or
-N(R10XR11);
R is selected from the group consisting of
Figure imgf000004_0001
R10 and R11 are independently selected from the group consisting of H,
R12-Ci_6alkyl, C3_i0cycloalkyl, C3_i0heterocycloalkyl or benzyl;
or R10 and R11, together with the nitrogen to which they are attached, form a 5 to 7 membered ring, wherein one carbon ring member not adjacent to the nitrogen can optionally be replaced by -O-, -S- or -NR14-;
R12 is 1 or 2 substituents independently selected from the group consisting of H, OH,
Ci_6alkoxy, Ci_6alkyl, Ci_6alkyl-S-, Ci_6alkyl-SO2-, C6-ioaryl, C6-ioaryl-S02-, C5_ioheteroaryl, - C(O)O-R14 and -C(O)N(R14)2;
R13 is 1 or 2 substituents selected from the group consisting of H, halo, Ci_6alkyl, C2. galkenyl, OH, hydroxyalkyl, Ci_6alkoxy, CN, -CF3, -OCF3, -SF5, -OSF5, -N(R17)2,
-aminobenzyl, - Ci_6alkyl-N(R10)(Rπ), -C(O)N(R10)(Rπ), -0-SO2- Ci_6alkyl, C3_i0cycloalkyl, (CH2)nC3_ioheterocycloalkylR16, (CH2)nC6-i0arylR16, C6-ioarylalkyl- and
(CH2)nC5_ioheteroarylR16 when R13 is attached to a ring carbon atom, and when R13 is attached to a ring nitrogen, it is H, C^alkyl or benzyl;
each R14 is independently selected from the group consisting of H, Ci_6alkyl, C3. iocycloalkyl and Cβ-ioarylalkyl;
R15 is 1 or 2 substituents independently selected from the group consisting of H, halo, Ci_6alkyl, Ci_6alkoxy, -CF3, C3_iocycloalkyl, Ci_6alkoxyalkoxy, OH, hydroxyalkyl, -OCF3, -O- C3_iocycloalkyl, benzyloxy, -C(O)O Chalky!, -O- Ci_6alkyl-CO2H, -C(O)N(R10)2, - N(R10)2, - Ci_6alkyl-N(R10)2, -NR10-C(O)N(R10)2, -N(R10)C(O)O Ci_6alkyl, -N(R10)SO2- C1. ealkyl, phenyl, CN, -SF5, -OSF5, -SO2R10, -SR10 and trimethylsilyl;
R16 is H, Ci_6alkyl, Ci_6alkoxy; and
each R17 is independently selected from the group consisting of H, d_6alkyl, C3. locycloalkyl, C3_ioheterocycloalkyl and benzyl.
The present invention also relates to a pharmaceutical composition comprising at least one compound of Formula I or a pharmaceutically acceptable salt thereof in a
pharmaceutically acceptable carrier.
In another embodiment, the present invention relates to a method of treating PDElO mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome comprising administering a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof to a mammal in need of such treatment. In another embodiment, the invention relates to a method of treating PDElO mediated disorders, for example CNS disorders such as schizophrenia, psychosis, cognitive disorders (such as Alzheimer's disease), bipolar disorder, depression, diet-induced obesity, diabetes and metabolic syndrome comprising administering to a mammal in need of such treatment a pharmaceutical composition comprising a therapeutically effective amount of at least one compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
Detailed Description
In one embodiment of Formula I, R1 is alkyl, preferably methyl.
In one embodiment of Formula I, R2 is H, OH, alkoxy (preferably methoxy or ethoxy) or -O(CH2)nR, preferably -O-CH2-quinolyl. More preferably, R2 is methoxy.
In one embodiment of Formula I, -OR1 and R2 together form -0-CH2-O-.
In one embodiment of Formula I, R3 is H or alkoxy (preferably methyoxy). More preferably, R3 is H.
In one embodiment of Formula I, R4, R5, R6 and R7 are independently H or alkyl; preferably R4, R5, R6 and R7 are each H.
In one embodiment of Formula I, R8 is H, alkyl or aryl; preferably, R8 is H, alkyl (preferably methyl) or phenyl; more preferably, R8 is H or methyl. In one embodiment of Formula I, R9 is R12-cycloalkyl, R13- Cβ-ioaryl or R13- C5-ioheteroaryl, preferably R13- Cβ-ioaryl or R13- Cs-ioheteroaryl. A preferred R12- cycloalkyl group is cyclohexyl. Preferred R13- Cβ-ioaryl groups are R13-phenyl, wherein R13 is H, 1 or 2 alkoxy groups (preferably methoxy), 1 or 2 halo atoms, -OCF3, -O-SO2-alkyl (preferably -O-SO2-methyl), Cs-ioheterocycloalkyl, (preferably
— N O — N NR 14
or N— f , wherein R14 is H or methyl), -N(R17)2 (preferably
Figure imgf000006_0001
Preferred R13-heteroaryl groups are those wherein the heteroaryl portion is thiazolyl, oxazolyl, imidazolyl, pyridyl, pyrimidinyl or quinolyl; more preferred heteroaryl groups are thiazolyl and pyridyl.
Preferred R13 substituents on the carbon atoms of the heteroaryl groups are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl (preferably cyclopropyl), CN, alkenyl (preferably vinyl), hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl),-aminobenzyl, heterocycloalkyl (preferably morpholinyl, pyrrolidinyl or piperidinyl), -alkyl-N(R10)(Rπ) (preferably -CH2-morpholinyl or -CH2-homomorpholinyl), -C(O)N(R1^(R11) (preferably -C(O)-N(alkyl)2, -C(O)- morpholinyl,
Figure imgf000006_0002
heteroaryl (wherein heteroaryl is preferably pyridyl, imidazolyl, pyrimidinyl or indolyl). Preferred R13 substituents on the nitrogen atoms of the heteroaryl groups (e.g., imidazolyl) are H, methyl and benzyl, more preferably H.
When R9 is R13-thiazolyl, preferred R13 substituents are H, 1 or 2 halo atoms, hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl), C3_ioheterocycloalkyl, (preferably morpholinyl), -alkyl-N(R10)(Rπ) (preferably -CH2-morpholinyl or -CH2- homomorpholinyl), -C(O)N(R1^(R11) (preferably -C(O)-N(alkyl)2, -C(O)-morpholinyl, - C(O)-homomorpholinyl, -C(O)NH-benzyl or
Figure imgf000006_0003
When R9 is R13-pyridyl, preferred R13 substituents are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl
(preferably cyclopropyl), CN, alkenyl (preferably vinyl), aryl (preferably phenyl), -aminobenzyl, heterocycloalkyl (preferably morpholinyl, pyrrolidinyl or piperidinyl), or R16-heteroaryl (wherein heteroaryl is preferably pyridyl, imidazolyl, pyrimidyl or indolyl).
In one embodiment of Formula I, R1 is alkyl; R2 is H, OH, alkoxy or
-O(CH2)nR, or -OR1 and R2 together form -0-CH2-O-; R3 is H or alkoxy; R4, R5, R6 and R7 are independently H or alkyl; and R8 is H, alkyl or aryl.
In another embodiment of Formula I, R1 is alkyl; R2 is alkoxy; R3 is H; R4, R5, R6 and R7 are independently H or alkyl; and R8 is H or alkyl.
In one embodiment of Formula I, R1 is alkyl, preferably methyl; R2 is alkoxy
(preferably methoxy or ethoxy); R3 is H or alkoxy (preferably methoxy); R4, R5, R6 and R7 are independently H or alkyl, preferably H; and R8 is H or alkyl, preferably H or methyl. For compounds in this embodiment, R9 is preferably R13-phenyl; or R13-heteroaryl, wherein heteroaryl is preferably thiazolyl or pyridyl; preferred R13 substituents are H, 1 or 2 halo atoms, 1 or 2 alkyl groups (preferably methyl or ethyl), alkoxy (preferably methoxy), cycloalkyl (preferably cyclopropyl), CN, alkenyl (preferably vinyl), hydroxyalkyl (preferably hydroxymethyl), aryl (preferably phenyl), -aminobenzyl, heterocycloalkyl (preferably morpholinyl, pyrrolidinyl or piperidinyl), -alkyl-N(R10)(Rπ) (preferably -CH2-morpholinyl or -CH2-homomorpholinyl), -C(O)N(R1^(R11) (preferably -C(O)-N(alkyl)2, -C(O)- morpholinyl, -C(O)-homomorpholinyl, -C(O)NH-benzyl or
Figure imgf000007_0001
or R1 - C5- ioheteroaryl (wherein heteroaryl is preferably pyridyl, imidazolyl, pyrimidyl or indolyl).
Another embodiment of the invention is represented by compounds of Formula 1-1
Figure imgf000007_0002
wherein R1 is methyl, R2 is methoxy, R3 to R7 are H, R9 is shown by the structure, and R8 is as defined:
Figure imgf000007_0003
R8=H R8=H R8= H
Figure imgf000007_0004
Figure imgf000008_0001
Figure imgf000008_0002
Figure imgf000008_0003
R8=CH3 R8=CH3 R8=CH3
Figure imgf000008_0004
RB=H R8= H Rb=H R8= H R8=H R8=H R8=H
/ Ni r-S n
Figure imgf000008_0005
R8=H Preferred compounds of Formula I are those in Examples IA, IB, ID, IE, IH, IN, IP, IQ, IR, IS, IU, IW, IY, IAA, ICC, IFF, IHH, HA, HC, HD, HE, HF, HG, HH, IU, HK, HL, HM, UN, HQ, HR, IIS, IIIA, IIIB, IUC, HID, HIE, IVA, IVB, IVC, IVD, VD, VIA, VIB, VIC, VID, VIE, VIF, VIIA, VIIB, VIIC, VIID, VIIE, VIIF, IX, X, XI, XII, XIV, XXII, XXIII, and XXIV.
More preferred compounds of Formula I are those in Examples ID, IP, IW, ICC, IFF, HD, HG, IU, HL, HQ, HR, IIIB, IUC, HID, IVD, VIA, VIB, VIC, VID, VIE, VIIA, VIIC, VIID, VIIE, XXII, and XXIV. As used herein, the following terms are as defined below unless otherwise indicated:
Mammal means humans and other mammalian animals.
The following definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Therefore, the definition of "alkyl" applies to "alkyl" as well as the "alkyl" portions of "hydroxyalkyl", "haloalkyl", "alkoxy", etc.
Alkyl means an aliphatic hydrocarbon group which may be straight or branched and comprising about 1 to about 20 carbon atoms in the chain. Preferred alkyl groups contain about 1 to about 12 carbon atoms in the chain. More preferred alkyl groups contain about 1 to about 6 carbon atoms in the chain. Branched means that one or more lower alkyl groups such as methyl, ethyl or propyl, are attached to a linear alkyl chain.
Hydroxyalkyl represents an alkyl group as defined substituted by 1 to 3 hydroxy groups. The bond to the parent is through the alkyl group.
Alkoxy means an alkyl-O- group in which the alkyl group is as previously described. Non-limiting examples of suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy and n-butoxy. The bond to the parent moiety is through the ether oxygen.
Halogen represents fluoro, chloro, bromo and iodo.
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. By "stable compound" or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The term "isolated" or "in isolated form" for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof. The term "purified" or "in purified form" for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
"Cycloalkyl" means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 3 to about 7 carbon atoms. Non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like. Non-limiting examples of suitable multicyclic cycloalkyls include 1-decalinyl, norbornyl, adamantly and the like.
"Heterocycloalkyl" means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. There are no adjacent oxygen and/or sulfur atoms present in the ring system. Preferred heterocycloalkyls contain about 5 to about 6 ring atoms. The prefix aza, oxa or thia before the heterocycloalkyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom. Any - NH in a heterocycloalkyls ring may exist protected such as, for example, as an -N(Boc), - N(CBz), -N(Tos) group and the like; such protections are also considered part of this invention. The nitrogen or sulfur atom of the heterocycloalkyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide. The heterocycloalkyl ring can be joined to the parent moiety by a ring carbon atom or a ring nitrogen atom. Non-limiting examples of suitable monocyclic heterocycloalkyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1 ,4-dioxanyl, tetrahydrofuranyl,
tetrahydrothiophenyl, lactam, lactone, and the like.
"Aryl" means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms. Non-limiting examples of suitable aryl groups include phenyl and naphthyl.
"Heteroaryl" means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination. Preferred heteroaryls contain about 5 to about 6 ring atoms. The prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom respectively, is present as a ring atom. A nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide. Non-limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, pyridone (including N- substituted pyridones), isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, oxindolyl, imidazo[l,2-a]pyridinyl, imidazo[2,l-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyridyl, isoquinolinyl,
benzoazaindolyl, 1,2,4-triazinyl, benzothiazolyl and the like. The term "heteroaryl" also refers to partially saturated heteroaryl moieties such as, for example, tetrahydroisoquinolyl, tetrahydroquinolyl and the like.
"Aralkyl" or "arylalkyl" means an aryl-alkyl- group in which the aryl and alkyl are as previously described. Preferred aralkyls comprise a lower alkyl group. Non-limiting examples of suitable aralkyl groups include benzyl, 2-phenethyl and naphthalenylmethyl. The bond to the parent moiety is through the alkyl.
When R4 and R5, or R6 and R7, together with the carbons to which they are attached form a 3-6 membered carbocyclic ring, the resultant spirocyclic rings are shown in the following partial structures:
Figure imgf000011_0001
It should be noted that in hetero-atom containing ring systems of this invention, there are no hydroxyl, halo, amino or thio groups on ring carbon atoms adjacent to a N, O or S. Thus, for example, in the ring:
Figure imgf000011_0002
there is no -OH, halo, amino or thio attached directly to carbons marked 2 and 5. It should also be noted that any carbon as well as heteroatom with unsatisfied valences in the text, schemes, examples and Tables herein is assumed to have the sufficient number of hydrogen atom(s) to satisfy the valences.
When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene et al, Protective Groups in Organic Synthesis (1991), Wiley, New York.
When any variable (e.g., alkyl, halo, etc.) occurs more than one time in any constituent or in Formula I or II, its definition on each occurrence is independent of its definition at every other occurrence.
As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
Prodrugs, solvates and co-crystals of the compounds of the invention are also contemplated herein. The term "prodrug", as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of Formula I or a salt and/or solvate thereof. A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems (1987) 1_4 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
"Solvate" means a physical association of a compound of this invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates, and the like. "Hydrate" is a solvate wherein the solvent molecule is H2O.
A co-crystal is a crystalline superstructure formed by combining an active
pharmaceutical intermediate with an inert molecule that produces crystallinity to the combined form. Co-crystals are often made between a dicarboxlyic acid such as fumaric acid, succinic acid etc. and a basic amine such as the one represented by compound I or II of this invention in different proportions depending on the nature of the co-crystal. (Rmenar, J. F. et. al. JAm. Chem. Soc. 2003, 125, 8456).
"Effective amount" or "therapeutically effective amount" is meant to describe an amount of compound or a composition of the present invention effective as PDElO inhibitors and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect.
The compounds of Formula I can form salts which are also within the scope of this invention. Reference to a compound of Formula I herein is understood to include reference to salts thereof, unless otherwise indicated. The term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of Formula I contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term "salt(s)" as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful. Salts of the compounds of the Formula I may be formed, for example, by reacting a compound of Formula I with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
Exemplary acid addition salts include acetates, ascorbates, benzoates,
benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, fumarates, hydrochlorides, hydrobromides, hydroiodides, lactates, maleates,
methanesulfonates, naphthalenesulfonates, nitrates, oxalates, phosphates, propionates, salicylates, succinates, sulfates, tartarates, thiocyanates, toluenesulfonates (also known as tosylates) and the like. Additionally, acids which are generally considered suitable for the formation of pharmaceutically useful salts from basic pharmaceutical compounds are discussed, for example, by P. Stahl et al, Camille G. (eds.) Handbook of Pharmaceutical Salts. Properties, Selection and Use. (2002) Zurich: Wiley- VCH; S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1) 1-19; P. Gould, International J. of Pharmaceutics (1986) 33 201-217; Anderson et al, The Practice of Medicinal Chemistry ( 1996), Academic Press, New York; and in The Orange Book (Food & Drug Administration, Washington, D. C. on their website). These disclosures are incorporated herein by reference thereto. Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases (for example, organic amines) such as dicyclohexylamines, t-butyl amines, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen- containing groups may be quaternized with agents such as lower alkyl halides (e.g. methyl, ethyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, and dibutyl sulfates), long chain halides (e.g. decyl, lauryl, and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
All such acid salts and base salts are intended to be pharmaceutically acceptable salts within the scope of the invention and all acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of the invention.
Compounds of Formula I, and salts, solvates, co-crystals and prodrugs thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates, co-crystals and prodrugs of the compounds as well as the salts and solvates, co-crystals of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention, as are positional isomers (such as, for example, 4-pyridyl and 3-pyridyl). Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected,
stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate" "prodrug" and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, positional isomers, racemates or prodrugs of the inventive compounds. Isomers can be prepared using conventional techniques, either by reacting optically pure or optically enriched starting materials or by separating isomers of a compound of Formula I. Isomers may also include geometric isomers, e.g., when a double bond is present.
Those skilled in the art will appreciate that for some of the compounds of Formula I, one isomer will show greater pharmacological activity than other isomers. Polymorphic forms of the compounds of Formula I, and of the salts, solvates, co- crystals and prodrugs of the compounds of Formula I, are intended to be included in the present invention.
In this specification, the term "at least one compound of Formula I" means that one to three different compounds of Formula I may be used in a pharmaceutical composition or method of treatment. Preferably one compound of Formula I is used.
The present invention also embraces isotopically-labelled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 18O, 170, 31P, 32P, 35S, 18F, and 36Cl, respectively.
Certain isotopically-labelled compounds of Formula I (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Isotopically labelled compounds of Formula I can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples hereinbelow, by substituting an appropriate isotopically labelled reagent for a non- isotopically labelled reagent. Compounds of Formula I are prepared by methods known in the art. Typical procedures for preparing the compounds are shown in reaction Schemes 1 and 2; in the schemes, the exemplified compound of Formula I is that wherein R1 is methyl, R2 is methoxy, R3-R7 are H and R8 is H (Scheme 1) or methyl (Scheme 2). Scheme 1
Figure imgf000016_0001
Scheme 2
Figure imgf000016_0002
Compounds of Formula I can be converted into other compounds of Formula I using methods well known in the art. For examples, see preparative Examples III to XIII, wherein substituents on the R9 variable added or converted to different substituents.
In the scheme above and in the following preparative examples, the following abbreviations are used: RT - room temperature; Ac - acetyl; Me - methyl; Et - ethyl; Ph - phenyl; i-Pr - isopropyl; t-Bu - t-butyl; Ts - tosyl; t-Boc - N-tert-butoxycarbonyl; BINAP - 2,2 ' -bis(diphenylphosphino)- 1 , 1 'binaphthyl; m-CPBA - m-chloroperoxybenzoic acid; DEAD - diethyl azodicarboxylate; DBU - l,8-diazabicyclo[5.4.0]undec-7-ene; DMF - dimethylformamide; EDC - l-(3-dimethylaminopropyl)-3-ethylcarbodiimide; HOBT - 1- hydroxybenzotriazole hydrate; THF - tetrahydrofuran; TFA - trifluoroacetic acid; TLC - thin layer chromatography; DIPEA - diisopropylethylamine; KHMDS - potassium bis(trimethylsilyl)amide; MSA - methane sulfonic acid; HATU - O-(7-azabenzotriazol-l- yl)-Λ/,Λ/,Λf',Λf'-tetramethyluronium hexafluorophosphate; PPTS - pyridinium/?- toluenesulfonate; DCM - dichloromethane; DCE - 1,2-dichloroethane.
Where LC/MS data are presented, analyses were performed using an Applied Biosystems API-150 mass spectrometer and Shimadzu SCL-IOA LC system. Column: Phenomenex Gemini C 18, 5 micron, 50 mm x 4.6 mm ID; Gradient: From 90% water, 10% CH3CN and 0.05%TFA, 5 min to 5% water, 95% CH3CN, 0.05% TFA in 5 minutes. MS data were obtained using Agilent Technologies LC/MSD SL or 1100 series LC/MSD mass spectrometer. Retention times refer to Total Ion Current (TIC) unless uv is indicated.
Following are examples of the preparation of intermediates and compounds of Formula I.
Example I
Figure imgf000017_0001
Figure imgf000017_0002
1 2
2-(3,4-Dimethoxy-phenyl)-ethylamine (18 g) was dissolved in 150 mL of ethyl formate and heated to 70 0C for 18 h. The reaction mixture was concentrated in vacuo to afford the desired formamide product 2 (21 g) which was used directly in the next step without further purification.
Figure imgf000017_0003
Formamide 2 (21 g) was dissolved in dry THF 140 (mL) and cooled to 0 0C. Et3N (70 mL) was added, followed by POCl3 (10.2 mL in 25 mL of THF) dropwise over 10 min. The reaction was stirred at 0 0C for 1 h. Ice water (500 mL) was added and the reaction was stirred for 10 min. The mixture was extracted with EtOAc. The combined organic layers were washed with brine, dried (MgSO4) and evaporated. The oily residue was purified via column chromatography (25-50% EtOAc/hexanes) to give a colorless oil (17 g), which solidified on standing to a white solid. MS (ESI) 192 (M+H).
Step 3:
Figure imgf000018_0001
Ammonium formate (8.75 g), thiazole-5-carbaldehyde (5.23 g), and isonitrile 3 (12.0 g) were dissolved in MeOH (100 mL) and heated to reflux for 4 h. The solution was concentrated to approximately 20 mL of MeOH and EtOAc (200 mL) was added. The mixture was filtered and the solvent evaporated to yield a light brown solid. Trituration with ether provided a white solid (5.70 g), which was used without further purification in the following step. MS (ESI) 350 (M+H).
Figure imgf000018_0002
P2O5 (40.7 g) was added to a clear solution of MSA (160 mL). The mixture was heated to 75 0C for 45 min, at which point most of the P2O5 had dissolved. Formamide 4 (5.56 g) was added as a solid in one portion. The resulting brown solution was stirred at 75 0C for 1 h to consume all of the formamide. The solution was allowed to cool to ambient temperature and then slowly poured onto solid NaHCO3 (400 g). Ice water was added and the aqueous suspension was extracted with EtOAc. The combined organic layers were dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH/DCM) to give I-A as a white solid (3.50 g). LCMS (ESI) 1.85 min (314, M+H).
The following compounds in Table I were prepared in a similar manner:
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Example II
Figure imgf000022_0002
Figure imgf000023_0001
Ammonium acetate (3.01 g) and thiazole-5-carbaldehyde (1.59 g), were dissolved in 34 mL of trifluroethanol. The reaction was heated at 60 0C for 30 min. Isonitrile 3 (3.35 g) was added in one portion, and the reaction was stirred at 60 0C for 4 h. The solution was allowed to cool to ambient temperature, the solvent evaporated, and the residue dissolved in EtOAc and filtered. The EtOAc was removed in vacuo and the residue purified via column chromatography (0-50% acetone/hexane) to provide formamide 5 (1.10 g). MS (ESI) 364 (M+H).
Step 2:
P2O5 (5.5 g) was added to a clear solution of MSA (22 mL). The mixture was heated to 75 0C for 30 min, at which point most of the P2O5 had dissolved. Formamide 5 (790 mg) was added as a solid in one portion. The resulting brown solution was stirred at 75 0C for 3 h to consume all of the formamide. The solution was allowed to cool to ambient temperature and then slowly poured onto solid NaHCOs (65 g). Ice water was added and the aqueous suspension was extracted with EtOAc. The combined organic layers were dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH/DCM) to give H-A as a white solid (230 mg). LCMS (ESI) 2.06 min (328, M+H).
The following compounds in Table II were prepared in a similar manner:
Figure imgf000023_0002
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0003
Example III
Figure imgf000026_0001
Figure imgf000026_0002
I-A 6 6a
Compound I-A (100 mg) was dissolved in 3 niL of THF and cooled to -78 0C. To this solution was added 200 μL of n-BuLi (2.5 M in hexanes). The reaction was stirred for 15 min at -78 0C and was poured onto CO2 pellets. After stirring for 10 min, the solvent was removed in vacuo to provide a mixture of 6 and 6a which was used without further purification in the next step.
Step 2:
Figure imgf000027_0001
Acid 6 from Step 1 (above) was dissolved in 5 mL of DMF. To this solution was added 100 μL of 4-ethylmorpholine, 130 μL of tert-hvXy\ methyl amine, 35 mg of HOBt and 120 mg of HATU. The reaction was stirred for 2 h at ambient temperature to consume all of the starting material (TLC). The reaction was diluted with water and extracted with EtOAc. The combined organic layers were dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH/DCM) to give IH-A (LCMS (ESI) 2.29 min (427, M+H)) and HIF (LCMS (ESI) 3.45 min (540, M+H)).
The following compounds in Table III were prepared in a similar manner:
Figure imgf000027_0002
Figure imgf000028_0001
Example IV
Figure imgf000028_0002
IV-A
Figure imgf000028_0003
IV-A
Bromide H-H (150 mg), and 3-pyridine boronic acid (92 mg) were dissolved in toluene (4 rnL) and MeOH (1.2 rnL). A 2 M aqueous solution OfK2COs was added and the mixture was degassed with argon. Pd(PPtLs)4 was added in one portion and the reaction was sealed and heated to 100 0C for 4 h. After cooling to ambient temperature, the reaction mixture was diluted with EtOAc and washed with water. The solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH- NHs/DCM) to give IV-A as a white solid (133 mg). LCMS (ESI) 2.35 min (399, M+H).
The following compounds in Table IV were prepared in a similar manner:
Figure imgf000028_0004
Figure imgf000029_0001
Example V
Figure imgf000029_0002
V-A
Figure imgf000029_0003
V-A
Bromide H-H (150 mg), Pd(OAc)2 (8.4 mg) BINAP (14 mg) and ΦuONa (50 mg) were placed in a vial and purged with nitrogen. Toluene (2.5 rnL) was added, followed by pyrrolidine (48 μL). The reaction was sealed and heated to 100 0C for 18 h. After cooling to ambient temperature, the reaction mixture was diluted with EtOAc and washed with water. The solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH-NH3/DCM) to give
V-A as a white solid (123 mg). LCMS (ESI) 2.11 min (391, M+H).
The following compounds in Table V were prepared in a similar manner:
Figure imgf000029_0004
Figure imgf000030_0001
Example VI
Figure imgf000030_0002
Example VI-A was prepared according to the procedure outlined for Example IV-A above except that bromide H-J was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-5% MeOH-NH3/DCM) to give VI-A as a white solid. LCMS (ESI) 2.75 min (399, M+H).
The following compounds in Table VI were prepared in a similar manner:
Figure imgf000030_0003
Figure imgf000031_0001
Example VII-A was prepared according to the procedure outlined for Example V-A above with the exception that bromide H-J was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-5% Me0H-NH3/DCM) to give VII-A as a white solid. LCMS (ESI) 1.48 min (391, M+H).
The following compounds in Table VII were prepared in the similar manner:
Figure imgf000032_0001
Example VIII
Figure imgf000032_0002
VIII
Figure imgf000032_0003
Bromide H-H (300 mg), Pd(PPh3)4 (87 mg), and Zn(CN)2 (88 mg) were placed in a vial and purged with nitrogen. DMF (2.5 mL) was added and the reaction was sealed and heated to 80 0C for 18 h. After cooling to ambient temperature, the reaction mixture was diluted with EtOAc and washed with water. The solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-10% MeOH/DCM) to give VIII as a white solid (259 mg). LCMS (ESI) 2.99 min (347, M+H).
Example IX
Figure imgf000033_0001
IX
Figure imgf000033_0002
Example IX was prepared according to the procedure outlined for Example VIII above except that bromide H-J (300 mg) was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-10% MeOH-NHs/DCM) to give IX as a white solid (240 mg). LCMS (ESI) 2.84 min (347, M+H).
Example X
Figure imgf000033_0003
Figure imgf000033_0004
Bromide H-H (200 mg) was placed in a vial and purged with nitrogen. DMF (2.5 mL) was added, followed by vinyl tributyltin (365 μL) and the solution was degassed with argon. Pd(PPtLs)4 (58 mg) was added and the reaction was sealed and heated to 100 0C for 5 h. After cooling to ambient temperature, the reaction mixture was diluted with EtOAc and washed with water. The solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH/DCM) to give Example X as a white solid (139 mg). LCMS (ESI) 1.56 min (348, M+H).
Example XI
Figure imgf000034_0001
Xl
Figure imgf000034_0002
Example XI was prepared according to the procedure outlined for Example X above except that bromide H-J (200 mg) was used in place of bromide H-H. The crude reaction residue was purified via column chromatography (0-5% MeOH-NHs/DCM) to give XI as a white solid (140 mg). LCMS (ESI) 1.55 min (348, M+H).
Example XII
Figure imgf000034_0003
XII
Figure imgf000034_0004
Alkene X (50 mg) was dissolved in 5 mL of EtOH. Pd/C (10% Pd, 30 mg) was added and the reaction mixture was placed under a hydrogen atmosphere. Hydrogenation continued under balloon pressure for 12 h at ambient temperature. The mixture was filtered through celite and the solvent evaporated to give Example XII as a white solid (50 mg). LCMS (ESI) 1.95 min (350, M+H).
Example XIII
Figure imgf000035_0001
Xl XIII
Example XIII was prepared according to the procedure outlined for Example XII above except that alkene XI (50 mg) was used in place of alkene X. The reaction mixture was filtered through celite and the solvent evaporated to give Example XIII as a white solid (50 mg). LCMS (ESI) 2.55 min (350, M+H).
Example XIV
Figure imgf000035_0002
XIV
Figure imgf000035_0003
JV-Bromosuccinamide (NBS) (38 g) was added to a cool (0 0C) solution of dimethoxyphenyl ethanol (7) (39 g) in DMF (800 mL). The red solution was allowed to warm to ambient temperature over 3 h. The solution was poured into water and extracted with EtOAc. The combined organic extracts were washed with sodium thiosulfate (aq), NaHCO3 (aq), and brine. The organic solution was dried (MgSO4) and evaporated to give bromide 8 (56 g) as an orange oil that was used without further purification in the next step.
Step 2:
Figure imgf000036_0001
Bromide 8 (56 g) was dissolved in DCM (250 mL). Dihydropyran (DHP) (29 mL) was added, followed by/?-toluene sulfonic acid (5.4 g). The solution was stirred at ambient temperature for 18 h. The solution was washed with water, NaHCO3 (aq), and brine. The organic solution was dried (MgSO4) and evaporated to give THP ether 9 (73 g) as a red oil that was used without further purification in the next step.
Step 3:
Figure imgf000036_0002
THP ether 9 (73 g) was dissolved in dry THF (700 mL). The solution was cooled to - 78 0C and stirred at -78 0C for 20 min. A solution of nBuLi in hexanes (2.5 M, 127 mL) was added slowly. After the addition was complete, the reaction was allowed to stir at -78 0C for 30 min. B(OMe)3 (35 mL) was added and the reaction was allowed to warm to ambient temperature over 2 h. IM HCl (600 mL) was added and the reaction vigorously stirred overnight. The layers were separated, and the aqueous layer was extracted with DCM. The combined organic layers were evaporated to a brown oil, and re-dissolved in DCM. The organic layer was extracted twice with 1 M NaOH. The combined aqueous layers were washed once with ether, and then acidified with concentrated HCl. The aqueous mixture was extracted with DCM. The combined organic extracts were dried (MgSO4) and evaporated give boronic acid 10 as a light yellow solid (35 g). The boronic acid was used in Step 4 without any further purification.
Figure imgf000036_0003
Boronic acid 10 (33 g) and bromide 11 (28 g) were placed in a flask. Toluene (960 mL) and MeOH (280 mL) were added, followed by 96 mL of 2 M K2CO3 (aq). The solution was degassed with argon. Pd(PPh3)4 (5.6 g) was added and the reaction was refluxed for 18 h. After cooling to ambient temperature, the reaction mixture was extracted with EtOAc. The organic solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-5% MeOH-NH3ZDCM) to give imidazole-aryl 12 as a white solid (28 g). LCMS (ESI) 2.55 min (393, M+H).
Figure imgf000037_0001
Imidazole-aryl 12 (28 g) was dissolved in EtOH (100 mL). 4 M HCl (100 mL) was added and the reaction was heated to reflux for 18 h. The solvents were evaporated, and the residue dissolved in a 50/50 mixture of sat. NaHCO3 (aq) and DCM. The aqueous layer was extracted with DCM, dried (MgSO4), and evaporated to give compound 13 as a white solid (19 g). LCMS (ESI) 1.38 min (263, M+H).
Step 6:
Figure imgf000037_0002
Alcohol 13 (19 g) was dissolved in DCM (75 mL) and cooled to 0 0C. Et3N (28 mL) and DMAP (1.5 g) were added, followed by/?-toluenesulfonyl chloride (TsCl) (35 g). The reaction was allowed to slowly warm to RT over 2 h. The reaction was cooled to 0 0C again and water (50 mL) was added and the reaction was vigorously stirred for 1 h at 0 0C. The layers were separated, and the aqueous layer was extracted with DCM. The combine organic extracts were evaporated and dissolved in acetone (75 mL) and IM NaOH (100 mL) and stirred at ambient temperature overnight. The solution was extracted with DCM, the combined organic extracts were dried (MgSO4), and evaporated. The residue was purified via column chromatography (0-5% MeOH-NH3ZDCM) to give cyclized compound 14 as a white solid (11 g). LCMS (ESI) 2.89 min (245, M+H).
Step 7:
Figure imgf000037_0003
Cyclized compound 14 (11 g) was dissolved in DMF (250 niL) and cooled to 0 °C. NBS (8.8 g) was added in one portion and the reaction was stirred at 0 0C for 2 h. The reaction was diluted with EtOAc and washed with water. The organic phase was dried (MgSO4), and evaporated. The residue was purified via column chromatography (0-10% MeOH/DCM) to give bromide 15 as a white solid (14 g). LCMS (ESI) 2.78 min (338, M+H). Step 8:
Figure imgf000038_0001
XIV
Bromide 15 (90 mg), and 5-pyrmidine boronic acid (70 mg) were dissolved in toluene (4 mL) and MeOH (800 μL). A 2 M aqueous solution Of K2CO3 (300 μL) was added and the mixture was degassed with argon. Pd(PPtLs)4 (32 mg) was added in one portion and the reaction was sealed and heated to 100 0C for 4 h. After cooling to ambient temperature, the reaction mixture was diluted with EtOAc and washed with water. The solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-10% MeOH/DCM) to give XIV as a white solid (63 mg). LCMS (ESI) 1.97 min (323, M+H).
Figure imgf000038_0002
Compound XV was synthesized using a similar procedure to Example XIV, except the substrate alcohol for Step 1 (above) was 2-(4-ethoxy-3-methoxy-phenyl)-ethanol instead of 2-(3,4-dimethoxy-phenyl)-ethanol (7). LCMS (ESI) 2.02 min (336, M+H).
Example XVI
Figure imgf000038_0003
Figure imgf000039_0001
Compound XV (20 mg) was dissolved in 150 μL of concentrated H2SO4. The red solution was heated to 65 0C for 1 h to consume all of the starting material. After cooling to ambient temperature, the reaction mixture was poured into NaHCO3 (aq) and extracted with DCM. The organic extracts were dried (MgSO4) and evaporated to give XVI as a white solid (18 mg). LCMS (ESI) 2.59 min (308, M+H).
Example XVII
Figure imgf000039_0002
XVI XVI l
Compound XVI (35 mg), PPh3 (30 mg), and quinolin-2-yl-methanol (18 mg) were placed in a vial and the vial was purged with nitrogen. THF (700 μL) was added and the reaction was cooled to 0 0C. DEAD (20 mg) in 100 μL of THF was added dropwise and the reaction allowed to stir at ambient temperature for 18 h. The solvent was evaporated and the residue purified via column chromatography (0-10% MeOH-NH3ZDCM) to give XVII as a white solid (34 mg). LCMS (ESI) 3.09 min (449, M+H).
Example XVIII
Figure imgf000039_0003
Example XVIII was prepared according to the general procedure outlined for Example II, except 3-(3-methoxy-phenyl)-propionitrile was used in place of 3-(3,4- dimethoxy-phenyl)-propionitrile (3). LCMS (ESI) 2.05 min (292, M+H).
Example XIX
Figure imgf000040_0001
Example XIX was prepared according to the general procedure outlined for Example II, except 3-benzo[l,3]dioxol-5-yl-propionitrile was used in place of 3-(3,4-dimethoxy- phenyl)-propionitrile (3). LCMS (ESI) 2.01 min (306, M+H).
Example XX
Figure imgf000040_0002
Benzyl imidazole H-B (327 mg) and ammonium formate (515 mg) were dissolved in MeOH. Pd/C (10% Pd, 20 mg) was added and the mixture heated at reflux for 2 h. After cooling to ambient temperature, the reaction mixture was filtered through celite. The solvent was evaporated, and the residue dissolved in DCM. The organic layer was washed with sat. NaHCO3 (aq). The organic solution was dried (MgSO4) and evaporated to give imidazole XX as a white solid (253 mg). LCMS (ESI) 0.96, 1.55 min (311, M+H).
Example XXI
Figure imgf000041_0001
XXI
Example XXI was prepared according to the general procedure outlined for Example II, except ammonium benzoate was used in place of ammonium acetate. LCMS (ESI) 2.47 min (384, M+H).
Example XXII and Example XXIII
Figure imgf000041_0002
Compound I-A (932 mg) was dissolved in dry THF (25 mL) and cooled to -78 0C. A solution of n-BuLi in hexanes (1.49 mL, 2 M) was added dropwise and the reaction was stirred for 30 min. DMF (2.78 mL) was added dropwise and the reaction was allowed to warm to RT overnight. Ice water was added, the layers were separated and the aqueous layer was extracted with EtOAc. The combined organic layers were washed with sat. NH4Cl, brine, and were dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-10% acetone/DCM) to give the desired aldehyde 21 which was used without further purification in the next step.
Step 2:
Aldehyde 21 (305 mg), morpholine (117 mg), and NaBH(OAc)3 (492 mg) were placed into a flask and AcOH (76 μL) and DCE (7 mL) were added. The reaction was stirred at ambient temperature 18 h. The reaction mixture was diluted with DCM and washed with 10% Na2CO3 and brine. The organic solution was dried (MgSO4) and evaporated. The residue was purified via column chromatography (0-60% acetone/DCM) to provide a mixture of Example XXII (LCMS (ESI) 0.93, 1.74 min (413, M+H)) and Example XXIII (LCMS (ESI) 1.07, 1.93 min (344, M+H)).
The following compound was prepared in a similar manner:
Figure imgf000042_0001
LCMS (ESI) 1.05, 1.75 min (427, M+H)
The amount and frequency of administration of the active compound employed and/or the pharmaceutically acceptable salts thereof will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient. A typical recommended dosage regimen can range from about 10 mg/dose to about 100 mg/dose, preferably about 10 to about 50 mg/dose, and more preferably about 20 to about 25 mg/dose.
For preparing pharmaceutical compositions from the compounds described by this invention, inert, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and
suppositories. The powders and tablets may be comprised of from about 5 to about 95 percent active ingredient. Suitable solid carriers are known in the art, e.g. magnesium carbonate, magnesium stearate, talc, sugar or lactose. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration. Examples of
pharmaceutically acceptable carriers and methods of manufacture for various compositions may be found in A. Gennaro (ed.), The Science and Practice of Pharmacy, 20th Edition, (2000), Lippincott Williams & Wilkins, Baltimore, MD.
Liquid form preparations include solutions, suspensions and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection or addition of sweeteners and opacifϊers for oral solutions, suspensions and emulsions.
Liquid form preparations may also include solutions for intranasal administration.
Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas, e.g. nitrogen. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
The compounds of the invention may also be deliverable transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
Preferably the compound is administered orally.
Preferably, the pharmaceutical preparation is in a unit dosage form. In such form, the preparation is subdivided into suitably sized unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage regimen for a particular situation is within the skill of the art. For convenience, the total daily dosage may be divided and administered in portions during the day as required.
The amount and frequency of administration of the compounds of the invention and/or the pharmaceutically acceptable salts thereof will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient as well as severity of the symptoms being treated. A typical recommended daily dosage regimen for oral administration can range from about 1 mg/day to about 300 mg/day, preferably 1 mg/day to 75 mg/day, in two to four divided doses.
The activity of the compounds of Formula I can be determined by the following procedures.
In Vitro PDElO assay
PDElOAl activity was measured in white opaque 384-well Opti-Plates (Perkin Elmer
Life Sciences) using a scintillation proximity assay (GE Healthcare). Human recombinant PDElOAl was purchased from BPS Bioscience, Inc. The reaction mixture contained PDElOAl ( 0.02 nM), 10 nM [3H]cAMP ([5 ',8-3H] Adenosine 3 ',5 '-cyclic phosphate, ammonium salt], Amersham) and various concentrations of compound in 50 mM Tris-HCl, pH 7.5, 8.3 mM MgCl2, 17 mM EGTA and 0.2% bovine serum albumen in a total volume of 30 1. The assay was initiated with the addition of substrate and was allowed to proceed for 30 minutes at room temperature before being stopped by the addition of 300 g yttrium SPA PDE beads. The reaction mixtures were thoroughly mixed, and the beads were allowed to settle for 30 minutes. The plates were then counted in a TopCount scintillation counter.
Under these conditions, less than 30% of the substrate was hydro lyzed in the absence of compound. Ki values were determined as described by Cheng and Prusoff (1973).
Using the test procedures described above, the following compounds of Formula I were found to have Ki values of less than 50 nM: Examples IA, IB, ID, IE, IH, IN, IP, IQ, IR,
IS, IU, IW, IY, IAA, ICC, IFF, IHH, HA, HC, HD, HE, HF, HG, HH, IU, HK, HL, HM, UN,
HQ, HR, IIS, IIIA, IIIB, IIIC, HID, HIE, IVA, IVB, IVC, IVD, VD, VIA, VIB, VIC, VID,
VIE, VIF, VIIA, VIIB, VIIC, VIID, VIIE, VIIF, IX, X, XI, XII, XIV, XXII, XXIII, and
XXIV.
The following compounds of Formula I were found to have Ki values of less than 10 nM: Examples ID, IP, IW, ICC, IFF, HD, HG, IU, HL, HQ, HR, IIIB, IIIC, HID, IVD, VIA,
VIB, VIC, VID, VIE, VIIA, VIIC, VIID, VIIE, XXII, and XXIV.
While the present invention has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present invention.

Claims

1. A compound of Formula I
Figure imgf000045_0001
or a pharmaceutically acceptable salt thereof, wherein
R1 is R12-Ci_6alkyl, C3-1O cycloalkyl, Ci_6alkoxyalkyl-, -CF3 Or -SF5;
R2 is H, OH, alkoxy, C1-6alkoxyalkoxy, -OCF3, -OSF5, -O- C3_i0cycloalkyl, or -0(CH2)nR;
or -OR1 and -R2 together are -0-(CH2)p-0-, wherein p is 1 or 2, and form a ring with the carbon atoms to which they are attached;
n is O, 1 or 2;
R3 is H, R12-Ci_6alkyl, Ci_6alkoxy, C3_iocycloalkyl, Ci_6alkoxyalkoxy, OH, -CF3, - OCF3, -SF5, -OSF5, halo, -O- C3_i0cycloalkyl, benzyloxy, -N(R10XR11) or CN;
R4, and R5, R6, R7 are independently selected from the group consisting of H, R12- Ci_ βalkyl, R13- Cβ-ioaryl and R13- C5_ioheteroaryl; or R4 and R5, together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring; or R6 and R7, together with the carbon atom to which they are attached, form a 3-6 membered carbocyclic ring;
R8 is H, R12- Ci_6alkyl, C3_i0cycloalkyl, C1-6alkoxy, d_6alkoxyalkoxy, OH, -CF3, - OCF3, -SF5, -OSF5, halo, -O- C3_i0cycloalkyl, benzyloxy, -N(R17)2, CN, R13-C6-i0aryl or R13- C5_ioheteroaryl;
R9 is R12-Ci_6alkyl, R12-C3_i0cycloalkyl, R13-C6-i0aryl, R13-C5_i0heteroaryl or
-N(R10XR11);
R is selected from the group consisting of
Figure imgf000045_0002
R10 and R11 are independently selected from the group consisting of H, R12-Ci_6alkyl, C3-iocycloalkyl, C3-ioheterocycloalkyl or benzyl;
or R10 and R11, together with the nitrogen to which they are attached, form a 5 to 7 membered ring, wherein one carbon ring member not adjacent to the nitrogen can optionally be replaced by -O-, -S- or -NR14-;
R12 is 1 or 2 substituents independently selected from the group consisting of H, OH, Ci_6alkoxy, Ci_6alkyl, Ci_6alkyl-S-, Ci_6alkyl-SO2-, Cβ-ioaryl, C6-ioaryl-S02-, C5_ioheteroaryl, - C(O)O-R14 and -C(O)N(R14)2;
R13 is 1 or 2 substituents selected from the group consisting of H, halo, Ci-βalkyl, C2- ealkenyl, OH, hydroxyalkyl, C1-6alkoxy, CN, -CF3, -OCF3, -SF5, -OSF5, -N(R17)2,
-aminobenzyl, - Ci_6alkyl-N(R10)(R11), -C(O)N(R1^(R11), -0-SO2- C1-6alkyl, C3_i0cycloalkyl, (CH2)nC3_ioheterocycloalkylR16, (CH2)nC6.i0arylR16, C6.i0arylalkyl- and
(CH2)nC5_ioheteroarylR16 when R13 is attached to a ring carbon atom, and when R13 is attached to a ring nitrogen, it is H, Ci_6alkyl or benzyl;
each R14 is independently selected from the group consisting of H, Ci_6alkyl, C3. iocycloalkyl and Cβ-ioarylalkyl;
R15 is 1 or 2 substituents independently selected from the group consisting of H, halo, Ci-βalkyl, Ci_6alkoxy, -CF3, C3_iocycloalkyl, Ci_6alkoxyalkoxy, OH, hydroxyalkyl, -OCF3, -O- C3_iocycloalkyl, benzyloxy, -C(O)O C1-6alkyl, -O- C1-6alkyl-CO2H, -C(O)N(R10)2, - N(R10)2, - Ci_6alkyl-N(R10)2, -NR10-C(O)N(R10)2, -N(R10)C(O)O Ci_6alkyl, -N(R10)SO2- C1. 6alkyl, phenyl, CN, -SF5, -OSF5, -SO2R10, -SR10 and trimethylsilyl;
R16 is H, Ci_6alkyl, Ci_6alkoxy; and
each R17 is independently selected from the group consisting of H, Ci_6alkyl, C3. iocycloalkyl, C3_ioheterocycloalkyl and benzyl.
2. The compound of claim 1 wherein R1 is alkyl; R2 is H, OH, alkoxy or
-0(CH2)nR, or -OR1 and R2 together form -0-CH2-O-; R3 is H or alkoxy; R4, R5, R6 and R7 are independently H or alkyl; and R8 is H, alkyl or aryl.
3. The compound of claim 1 wherein R1 is alkyl; R2 is alkoxy; R3 is H or methoxy; R4, R5, R6 and R7 are independently H or alkyl; and R8 is H or alkyl.
4. The compound of claim 1 wherein R is R 12 -cyc ~ilo ~a„1lk1 y 1l, τ R") 13 -aryl or
R13-heteroaryl.
5. The compound of claim 4 wherein R is R -phenyl, wherein R is H, 1 or 2 alkoxy groups, 1 or 2 halo atoms, -OCF3, -0-S02-alkyl, heterocycloalkyl, -N(R 17\ )2 or
R16-heteroaryl.
6. The compound of claim 5 wherein R13 is H, 1 or 2 methoxy groups, 1 or 2 halo atoms, -OCF3, -0-SO2-CH3,
— N .0 — N MR14 -HN-^ O
or >— f , wherein R14 is H or methyl
'
or R16-pyridyl.
7. The compound of claim 4 wherein R9 is R13-heteroaryl, wherein heteroaryl is thiazolyl, oxazolyl, imidazolyl, pyridyl, pyrimidinyl or quinolyl and R13 is H 1 or 2 halo atoms, 1 or 2 alkyl groups, alkoxy, cycloalkyl, CN, alkenyl, hydroxyalkyl, aryl,
-aminobenzyl, heterocycloalkyl, -alkyl-N(R10)(Rπ), -C(O)N(R10)(Rπ) or R16-heteroaryl.
8. The compound of claim 7 wherein R13 is H, or 1 to 2 groups independently selected from halogen, methyl and ethyl, methoxy, cyclopropyl, CN, vinyl, hydroxymethyl, phenyl, - aminobenzyl, morpholinyl, pyrrolidinyl, piperidinyl, -CH2-morpholinyl, -CH2- homomorpholinyl),-C(O)-N(alkyl)2, -C(O)-morpholinyl, -C(O)-homomorpholinyl, -
C(O)NH-benzy 1l,,
Figure imgf000047_0001
oorr RR16--lheteroaryl wherein heteroaryl is pyridyl, imidazolyl, pyrimidinyl or indolyl.
9. The compound of claim 7 wherein R is R -thiazolyl or R -pyridyl.
10. The compound of claim 8 wherein R9 is R13-thiazolyl or R13-pyridyl.
11. The compound of claim 1 wherein R1 is alkyl; R2 is H, OH, alkoxy or
-O(CH2)nR, or -OR1 and R2 together form -0-CH2-O-; R3 is H or alkoxy; R4, R5, R6 and R7 are independently H or alkyl; R8 is H, alkyl or aryl; and R9 is R12-cycloalkyl, R13-aryl or R13-heteroaryl.
12. The compound of claim 1 wherein R1 is alkyl; R2 is alkoxy; R3 is H or methoxy; R4, R5, R6 and R7 are independently H or alkyl; R8 is H or alkyl; R9 is R13-phenyl, wherein R13 is H, 1 or 2 alkoxy groups, 1 or 2 halo atoms, -OCF3, -0-S02-alkyl, heterocycloalkyl, - N(R17)2 or R16-heteroaryl; or R9 is R13-heteroaryl, wherein heteroaryl is thiazolyl, oxazolyl, imidazolyl, pyridyl, pyrimidinyl or quinolyl and R13 is H 1 or 2 halo atoms, 1 or 2 alkyl groups, alkoxy, cycloalkyl, CN, alkenyl, hydroxyalkyl, aryl, -aminobenzyl, heterocycloalkyl, -alkyl-N(R10)(Rπ), -C(O)N(R10)(Rπ) or R16-heteroaryl.
13. A compound which is selected from the group consisting of Examples IA, Table I, Example II, Table II, Example III, Table III, Example IV, Table IV, Example V, Table V, Example VI, Table VI, Example VII, Table VII, Examples VIII, IX, X, XI, XII, XIII, XIV, XV, XVI, XVII, XVIII, XIX, XX, XXI, XXII, XXII, XXIII, and XXIV, or a
pharmaceutically acceptable salt thereof.
14. The compound of claim 13 selected from the group consisting of Examples IA, IB, ID, IE, IH, IN, IP, IQ, IR, IS, IU, IW, IY, IAA, ICC, IFF, IHH, HA, IIC, HD, HE, HF, HG, IIH, IU, IIK, IIL, HM, IIN, HQ, HR, IIS, IIIA, IIIB, IIIC, HID, HIE, IVA, IVB, IVC, IVD, VD, VIA, VIB, VIC, VID, VIE, VIF, VIIA, VIIB, VIIC, VIID, VIIE, VIIF, IX, X, XI, XII, XIV, XXII, XXIII, and XXIV, or a pharmaceutically acceptable salt thereof.
15. A pharmaceutical composition for treating phosphodiesterase 10 modulated disorders comprising an effective amount of at least one compound of claim 1 in a pharmaceutically acceptable carrier.
16. A pharmaceutical composition for treating phosphodiesterase 10 modulated disorders comprising an effective amount of at least one compound of claim 13 in a pharmaceutically acceptable carrier.
17. A method of treating phosphodiesterase 10 modulated disorders comprising administering to a mammal in need of such treatment an effective amount of a compound of claim 1.
PCT/US2010/041122 2009-07-14 2010-07-07 Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors WO2011008597A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US22530609P 2009-07-14 2009-07-14
US61/225,306 2009-07-14

Publications (1)

Publication Number Publication Date
WO2011008597A1 true WO2011008597A1 (en) 2011-01-20

Family

ID=43449690

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/041122 WO2011008597A1 (en) 2009-07-14 2010-07-07 Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors

Country Status (1)

Country Link
WO (1) WO2011008597A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014027078A1 (en) 2012-08-17 2014-02-20 AbbVie Deutschland GmbH & Co. KG Inhibitor compounds of phosphodiesterase type 10a
EP2719689A1 (en) * 2012-10-12 2014-04-16 Les Laboratoires Servier Novel method for synthesising 3-(2-bromo-4,5-dimethoxyphenyl) propanenitrile and use for synthesising ivabradine and the added salts thereof with a pharmaceutically acceptable acid
WO2014079995A2 (en) 2012-11-26 2014-05-30 Abbvie Inc. Novel inhibitor compounds of phosphodiesterase type 10a
WO2014140184A1 (en) 2013-03-14 2014-09-18 AbbVie Deutschland GmbH & Co. KG Novel inhibitor compounds of phosphodiesterase type 10a
US9388180B2 (en) 2012-09-17 2016-07-12 Abbvie Inc. Inhibitor compounds of phosphodiesterase type 10A
US10039764B2 (en) 2013-07-12 2018-08-07 University Of South Alabama Treatment and diagnosis of cancer and precancerous conditions using PDE10A inhibitors and methods to measure PDE10A expression
WO2021188414A1 (en) * 2020-03-16 2021-09-23 Enanta Pharmaceuticals, Inc. Functionalized heterocyclic compounds as antiviral agents
US11198693B2 (en) 2018-11-21 2021-12-14 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
WO2021253091A1 (en) * 2020-06-18 2021-12-23 Alterity Therapeutics Limited Compounds for and methods of treating diseases
US11236108B2 (en) 2019-09-17 2022-02-01 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11377450B2 (en) 2018-09-21 2022-07-05 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11472808B2 (en) 2019-06-04 2022-10-18 Enanta Pharmaceuticals, Inc. Substituted pyrrolo[1,2-c]pyrimidines as hepatitis B antiviral agents
US11738019B2 (en) 2019-07-11 2023-08-29 Enanta Pharmaceuticals, Inc. Substituted heterocycles as antiviral agents
US11760755B2 (en) 2019-06-04 2023-09-19 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
US12011425B2 (en) 2017-08-28 2024-06-18 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051877A1 (en) * 2001-12-18 2003-06-26 Bayer Corporation 2-substituted pyrrolo[2.1-a]isoquinolines against cancer
WO2005087919A1 (en) * 2004-03-12 2005-09-22 Pfizer Products Inc. Crystal structure of 3',5'-cyclic nucleotide phosphodiesterase (pde10a) and uses thereof
WO2009070584A1 (en) * 2007-11-30 2009-06-04 Wyeth Aryl and heteroaryl fused imidazo[1,5-a]pyrazines as inhibitors of phosphodiesterase 10

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003051877A1 (en) * 2001-12-18 2003-06-26 Bayer Corporation 2-substituted pyrrolo[2.1-a]isoquinolines against cancer
WO2005087919A1 (en) * 2004-03-12 2005-09-22 Pfizer Products Inc. Crystal structure of 3',5'-cyclic nucleotide phosphodiesterase (pde10a) and uses thereof
WO2009070584A1 (en) * 2007-11-30 2009-06-04 Wyeth Aryl and heteroaryl fused imidazo[1,5-a]pyrazines as inhibitors of phosphodiesterase 10

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014027078A1 (en) 2012-08-17 2014-02-20 AbbVie Deutschland GmbH & Co. KG Inhibitor compounds of phosphodiesterase type 10a
US9464085B2 (en) 2012-08-17 2016-10-11 AbbVie Deutschland GmbH & Co. KG Inhibitor compounds of phosphodiesterase type 10A
US9388180B2 (en) 2012-09-17 2016-07-12 Abbvie Inc. Inhibitor compounds of phosphodiesterase type 10A
AU2013237671B2 (en) * 2012-10-12 2017-03-02 Les Laboratoires Servier New process for the synthesis of 3-(2-bromo-4,5-dimethoxyphenyl)propanenitrile, and application in the synthesis of ivabradine and additional salts thereof with a pharmaceutically acceptable acid
EP2719689A1 (en) * 2012-10-12 2014-04-16 Les Laboratoires Servier Novel method for synthesising 3-(2-bromo-4,5-dimethoxyphenyl) propanenitrile and use for synthesising ivabradine and the added salts thereof with a pharmaceutically acceptable acid
AU2013237671A1 (en) * 2012-10-12 2014-05-01 Les Laboratoires Servier New process for the synthesis of 3-(2-bromo-4,5-dimethoxyphenyl)propanenitrile, and application in the synthesis of ivabradine and additional salts thereof with a pharmaceutically acceptable acid
FR2996845A1 (en) * 2012-10-12 2014-04-18 Servier Lab NOVEL PROCESS FOR THE SYNTHESIS OF 3- (2-BROMO-4,5-DIMETHOXYPHENYL) PROPANENITRILE, AND APPLICATION TO THE SYNTHESIS OF IVABRADINE AND ITS SALTS OF ADDITION TO A PHARMACEUTICALLY ACCEPTABLE ACID
US8779121B2 (en) 2012-10-12 2014-07-15 Les Laboratoires Servier Process for the synthesis of 3-(2-bromo-4,5-dimethoxyphenyl)propanenitrile, and application in the synthesis of ivabradine and addition salts thereof with a pharmaceutically acceptable acid
AU2013237671C1 (en) * 2012-10-12 2017-06-15 Les Laboratoires Servier New process for the synthesis of 3-(2-bromo-4,5-dimethoxyphenyl)propanenitrile, and application in the synthesis of ivabradine and additional salts thereof with a pharmaceutically acceptable acid
MD4335C1 (en) * 2012-10-12 2015-10-31 Les Laboratoires Servier Process for the synthesis of 3-(2-bromo-4,5-dimethoxyphenyl)propanenitrile and process for the synthesis of ivabradine and addition salts thereof with the use thereof
EP2719689B1 (en) 2012-10-12 2015-12-23 Les Laboratoires Servier Novel method for synthesising 3-(2-bromo-4,5-dimethoxyphenyl) propanenitrile and use for synthesising ivabradine and the added salts thereof with a pharmaceutically acceptable acid
CN103724228B (en) * 2012-10-12 2016-04-20 瑟维尔实验室 The novel method of synthesis 3-(2-bromo-4,5-dimethoxy phenyl) propionitrile and the application in the additive salt of synthesis of ivabradine and itself and pharmaceutically acceptable acid
WO2014057228A1 (en) * 2012-10-12 2014-04-17 Les Laboratoires Servier New method for synthesising -(2-bromo-4,5-dimethoxyphenyl)propanenitrile, and application to the synthesis of ivabradine and the addition salts of same with a pharmaceutically acceptable acid
CN103724228A (en) * 2012-10-12 2014-04-16 瑟维尔实验室 Novel method for synthesising 3-(2-bromo-4,5-dimethoxyphenyl) propanenitrile and use for synthesising ivabradine and the added salts thereof with a pharmaceutically acceptable acid
US9790203B2 (en) 2012-11-26 2017-10-17 Abbvie Inc. Inhibitor compounds of phosphodiesterase type 10A
WO2014079995A2 (en) 2012-11-26 2014-05-30 Abbvie Inc. Novel inhibitor compounds of phosphodiesterase type 10a
WO2014140184A1 (en) 2013-03-14 2014-09-18 AbbVie Deutschland GmbH & Co. KG Novel inhibitor compounds of phosphodiesterase type 10a
US10039764B2 (en) 2013-07-12 2018-08-07 University Of South Alabama Treatment and diagnosis of cancer and precancerous conditions using PDE10A inhibitors and methods to measure PDE10A expression
US12011425B2 (en) 2017-08-28 2024-06-18 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
US11377450B2 (en) 2018-09-21 2022-07-05 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11198693B2 (en) 2018-11-21 2021-12-14 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11891393B2 (en) 2018-11-21 2024-02-06 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
US11760755B2 (en) 2019-06-04 2023-09-19 Enanta Pharmaceuticals, Inc. Hepatitis B antiviral agents
US11472808B2 (en) 2019-06-04 2022-10-18 Enanta Pharmaceuticals, Inc. Substituted pyrrolo[1,2-c]pyrimidines as hepatitis B antiviral agents
US11738019B2 (en) 2019-07-11 2023-08-29 Enanta Pharmaceuticals, Inc. Substituted heterocycles as antiviral agents
US11236108B2 (en) 2019-09-17 2022-02-01 Enanta Pharmaceuticals, Inc. Functionalized heterocycles as antiviral agents
WO2021188414A1 (en) * 2020-03-16 2021-09-23 Enanta Pharmaceuticals, Inc. Functionalized heterocyclic compounds as antiviral agents
US11802125B2 (en) 2020-03-16 2023-10-31 Enanta Pharmaceuticals, Inc. Functionalized heterocyclic compounds as antiviral agents
CN115667256A (en) * 2020-06-18 2023-01-31 艾特里提治疗有限公司 Compounds for treating diseases and methods of treating diseases
WO2021253091A1 (en) * 2020-06-18 2021-12-23 Alterity Therapeutics Limited Compounds for and methods of treating diseases

Similar Documents

Publication Publication Date Title
WO2011008597A1 (en) Dihydroimidazoisoquinoline derivatives useful as pde10 inhibitors
AU2017281228B2 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor M4
EP3416639B1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
EP2417135A1 (en) Substituted triazolopyridines and analogs thereof
EP3319966B1 (en) Bicyclic heterocyclic compounds as pde2 inhibitors
EP3534901B1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4
EP3558309B1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4
JP2012506873A (en) Substituted pyrazoloquinolines and their derivatives
EP3544961B1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4
US20240228489A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
EP3534888B1 (en) Substituted bicyclic heteroaryl allosteric modulators of nicotinic acetylcholine receptors
US20200131180A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
WO2014194519A1 (en) Imidazole derivatives and methods of use thereof for improving pharmacokinetics of drug
US11376254B2 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor M4
US20190330226A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
WO2019241467A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
US10736898B2 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor M4
US12065433B2 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor M1
WO2024086570A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
WO2024086569A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m1
WO2023141511A1 (en) Positive allosteric modulators of the muscarinic acetylcholine receptor m4

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10800329

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 10800329

Country of ref document: EP

Kind code of ref document: A1