WO2006057983A1 - Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease - Google Patents

Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease Download PDF

Info

Publication number
WO2006057983A1
WO2006057983A1 PCT/US2005/042233 US2005042233W WO2006057983A1 WO 2006057983 A1 WO2006057983 A1 WO 2006057983A1 US 2005042233 W US2005042233 W US 2005042233W WO 2006057983 A1 WO2006057983 A1 WO 2006057983A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
aryl
thiazolyl
heteroaryl
Prior art date
Application number
PCT/US2005/042233
Other languages
French (fr)
Inventor
Hemaka A. Rajapakse
Philippe G. Nantermet
Harold G. Selnick
Keith P. Moore
Original Assignee
Merck & Co., Inc.
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 Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to CA002587256A priority Critical patent/CA2587256A1/en
Priority to US11/791,465 priority patent/US7951949B2/en
Priority to JP2007543382A priority patent/JP2008520727A/en
Priority to AT05849049T priority patent/ATE512147T1/en
Priority to AU2005309708A priority patent/AU2005309708A1/en
Priority to EP05849049A priority patent/EP1817312B1/en
Publication of WO2006057983A1 publication Critical patent/WO2006057983A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/08Bridged systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention is directed to the field of compounds which are inhibitors of the the activity of the ⁇ -secretase enzyme, and to the use of the compounds for the treatment of diseases in which the ⁇ - secretase enzyme is involved, such as Alzheimer's disease.
  • Alzheimer's disease is characterized by the abnormal deposition of amyloid in the brain in the form of extra-cellular plaques and intra-cellular neurofibrillary tangles.
  • the rate of amyloid accumulation is a combination of the rates of formation, aggregation and egress from the brain. It is generally accepted that the main constituent of amyloid plaques is the 4kD amyloid protein ( ⁇ A4, also referred to as A ⁇ , ⁇ -protein and ⁇ AP) which is a proteolytic product of a precursor protein of much larger size.
  • the amyloid precursor protein (APP or A ⁇ PP) has a receptor-like structure with a large ectodomain, a membrane spanning region and a short cytoplasmic tail.
  • the A ⁇ domain encompasses parts of both extra-cellular and transmembrane domains of APP, thus its release implies the existence of two distinct proteolytic events to generate its NH 2 - and COOH-termini. At least two secretory mechanisms exist which release APP from the membrane and generate soluble, COOH-trancated forms of APP (APP S ). Proteases that release APP and its fragments from the membrane are termed
  • secretases Most APP 5 is released by a putative ⁇ -secretase which cleaves within the A ⁇ protein to release ⁇ -APP s and precludes the release of intact A ⁇ . A minor portion of APP 3 is released by a ⁇ - secretase (“ ⁇ -secretase”), which cleaves near the NH 2 -terminus of APP and produces COOH-terminal fragments (CTFs) which contain the whole A ⁇ domain.
  • ⁇ -secretase ⁇ -secretase
  • CTFs COOH-terminal fragments
  • the compoimds of the present invention are useful for treating Alzheimer's disease by inhibiting the activity of ⁇ -secretase or BACE, thus preventing the formation of insoluble A ⁇ and arresting the production of A ⁇ .
  • the present invention is directed to novel macrocyclic aminopyridyl compounds represented by general formula (I)
  • the invention is also directed to pharmaceutical compositions which include an effective amount of a compound of formula (T), or pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof, and a pharmaceutically acceptable carrier.
  • the invention is also directed to methods of treating mammals for diseases in which the ⁇ -secretase enzyme is involved, such as Alzheimer's disease, and the use of the compounds and pharmaceutical compositions of the invention in the treatment of such diseases.
  • the present invention is directed to compounds of formula (I):
  • Y is selected from the group consisting of
  • A is selected from the group consisting of
  • aryl and heteroaryl groups are unsubstituted or substituted with one or more (i) halo, (Ii) -OH,
  • Rl is selected from the group consisting of (1) -C6-10 arylene, or j ..' a "r -iu-,,> !!., « , complicat-,. « ceremoni5-!
  • heteroarylene selected from the group consisting of divalent pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benziniidazolyl, indynyl and benzoxazolyl,
  • arylene or heteroarylene is unsubstituted or substituted with one or more (a) halo
  • R2 is selected from the group consisting of: (1) (R5-SO2)N(R6)-, wherein R5 is (a) -Ci-10 alkyl, (b) -C2-10 alkenyl, (c) -C2-IO alkynyl,
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heteroaryl is unsubstituted or substituted with one or more
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, and said aryl and heteroaryl is unsubstituted or substituted with one or more (A) halo,
  • R6 is selected from the group consisting of (a) hydrogen,
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
  • alkyl, alkenyl, alkynyl, aryl or heteroaryl. is unsubstituted or substituted with one or more (i) halo, ( ⁇ ) -OH, (iii) -CN,
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl;
  • R 5 and K.6 may be linked to form a group -CH2(CH2)pCH2 ⁇ ,
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl,pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, wherein said heteroaryl is unsubstituted or substituted with one or more clergy ⁇ • . ⁇ i - i • - - -
  • Q ⁇ is selected from the group consisting of (a) -CH2-
  • Q3 is -CH- or -CH2CH-
  • RY is selected from the group consisting of
  • heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
  • alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heteroaryl RY groups are unsubstituted or substituted with one or more (i) halo,
  • Q4 is selected from the group consisting of
  • R z is selected from the group consisting of (i) hydrogen,
  • heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
  • R4 is -(CH 2 ) S -Q ⁇ -(CH2)t, wherein Q2 is selected from the group consisting of
  • n 0, 1 or 2
  • p is 1, 2 or 3
  • q is 1, 2, 3, 4 or 5
  • r is 0, 1 or 2
  • s is 0 or 1
  • t is 0 or 1
  • u is 0, 1 or 2.
  • Y is hydrogen
  • Rl is unsubstituted or substituted -Cg-io arylene, preferably unsubstituted phenylene.
  • R.4 is -(CH2) S -Q2 -(CEt ⁇ t, wherein Q2 is selected from the group consisting of
  • A is selected from the group consisting of (1) hydrogen
  • heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl.
  • A is -Cl-10 alkyl
  • alkyl (preferably methyl), wherein said alkyl is unsubstituted or substituted with one or more halo (preferably fiuoro).
  • R2 is selected from the group consisting of (R 5 -S ⁇ 2)N(R6)-, wherein R5 is -C ⁇ . ⁇ alkyl, wherein said alkyl is unsubstituted or substituted with one or more
  • R6 is selected from the group consisting of (a) hydrogen,
  • R ⁇ group is-C6-10 ar yl 5 unsubstituted or substituted as described above.
  • Preferred aryl groups are phenyl groups, unsubstituted or substituted with cyano.
  • a preferred R2 substituent is shown below:
  • R2 substituent is heteroaryl, either unsubstituted or substituted as described above.
  • a preferred heteroaryl group is furanyl or oxazolyl, either unsubstituted or substituted as described above.
  • a preferred furanyl or oxazolyl substituent is depicted below:
  • Ql is selected from the group consisting of
  • R x is preferably hydrogen, and n is preferably 1.
  • Q3 is -O - and n is preferably 1.
  • R ⁇ is preferably hydrogen, and n is preferably 1.
  • the dotted line leading to RY is absent and Ry is selected from the group consisting of
  • Q4 is CH2, m is preferably 1 and r is preferably 0.
  • the invention is directed to compounds of Formula (JT):
  • Another embodiment of the present invention includes a compound which is selected from the title compounds of the following Examples and pharmaceutically acceptable salts thereof.
  • alkyl by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C i _ 10 alkyl means an alkyl group having from one to ten carbon atoms).
  • Preferred alkyl groups for use in the invention are Cl-6 alkyl groups, having from one to six carbon atoms.
  • Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl, hexyl, and the like.
  • alkylene by itself or as part of another substituent, means a saturated straight or branched chain divalent hydrocarbon radical having the number of carbon atoms designated.
  • Co alkylene (for example, in the radical "-Coalkylene-C6-l ⁇ aryl") means that the alkylene group is absent.
  • cycloalkyl by itself or as part of another substituent, means a saturated cyclic hydrocarbon radical having the number of carbon atoms designated (e.g., C3.8 cycloalkyl means a cycloalkyl group having from three to eight carbon atoms).
  • exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. IU-.. ....J' ,
  • alkenyl by itself of as part of another substituent, means a straight or branched chain hydrocarbon radical having a single carbon-carbon double bond and having the number of carbon atoms designated (e.g., C2-10 alkenyl means an alkenyl group having from one to ten carbon atoms).
  • Preferred alkenyl groups for use in the invention are C2-6 alkenyl groups, having from two to six carbon atoms.
  • Exemplary alkenyl groups include ethenyl, n-propenyl, isopropenyl, butenyl, and the like.
  • alkynyl by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C2-10 alkynyl means an alkynyl group having from two to ten carbon atoms).
  • Preferred alkynyl groups for use in the invention are C2-6 alkynyl groups, having from two to six carbon atoms.
  • Exemplary alkynyl groups include ethynyl and propynyl.
  • aryl by itself or as part of another substituent, means an aromatic or cyclic radical having the number of carbon atoms designated (e.g., Cg_io aryl means an aryl group having from six to ten carbons atoms).
  • aryl includes multiple ring systems as well as single ring systems.
  • Preferred aryl groups for use in the invention include phenyl and naphthyl.
  • arylene by itself or as part of another substituent, means a divalent aromatic or cyclic radical, having the number of carbon atoms designated (e.g., C6-10 arylene means an arylene group having from six to ten carbons atoms).
  • arylene includes multiple ring systems as well as single ring systems.
  • Preferred arylene groups for use in the invention include phenylene and naphthylene.
  • heteroaryl by itself or as part of another substituent, means an aromatic cyclic radical having at least one ring heteroatom (O, N or S).
  • heteroaryl includes multiple ring systems as well as single ring systems.
  • heteroaryl groups for use in the invention include furyl, pyranyl, benzofuranyl, isobenzofuranyl, chromenyl, thienyl, benzothiophenyl, pyrrolyl, pyrazolyl, imidazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzimidazolyl, quinolinyl, isoquinolinyl, tetrazolyl, indazolyl, napthyridinyl, triazolyl, oxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isoxazolyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indynyl and dihydroindolyl.
  • the substituent When a heteroaryl group as defined herein is substituted, the substituent may be bonded to a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits substitution. Preferably, the substituent is bonded to a ring carbon atom.
  • the point of attachment may be at a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits attachment.
  • the attachment is at a ring carbon atom.
  • heteroarylene by itself or as part of another substituent, means an aromatic cyclic divalent radical having at least one ring heteroatom (O, N or S).
  • halo or “halogen” includes fluoro, chloro, bromo and iodo.
  • the compounds of the instant invention have at least one asymmetric center. Additional asymmetric centers may be present depending upon the nature of the various substituents on the ir iL if . ⁇ • ''Li 1 ⁇ Ji iJ .”.3 ,.' ' ' rE".'. iC' J .,,"S
  • racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated.
  • the separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography.
  • the coupling reaction is often the formation of salts using an enantiomerically pure acid or base.
  • the diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue.
  • the racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art.
  • any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
  • the compounds of the present invention are prepared by the methods outlined in Schemes 1.1 to 5.8, below, and the intermediates and examples herein.
  • the compounds of the present invention are prepared by the methods outlined in Schemes 1.1-5.8, illustrated below.
  • Scheme 1.1 describes the preparation of derivatives of type 1.1a, as well as the corresponding trifiate analogs 1.1b and 1.1c.
  • starting from glycine Schiff base more elaborated bromides of type l.ld and l.le were prepared. IUn il .'' 1 U '-Jt ⁇ !...!' ..ml! ,•" " I! Hv, St.,,, , n Vêt.
  • Scheme 2.1 describes a sequence leading to 2,6-dichloro pyridine derivatives from 2,6- dichoroisonicotinate.
  • Methyl ester hydrolysis gives acid 2.1a, which was transformed via a two step sequence to acylhydrazide 2.1b.
  • Benzyl alcohol 2.1c is readily accessed via ester reduction, and was transformed to the corresponding benzyl bromide with CBr 4 /Ph 3 P.
  • Homo-allylic amies of type 3.1b were prepared via conjugate addition of phthalimide to ⁇ , ⁇ -unsaturated aldehydes, followed by Wittig homologation to give compounds of type 3.1a, as described in Scheme 3.1 (See Bergman, E. D., Migron, Y. Organic Preparations and Procedures Int. 1976, 8, 75-80). Pthalimide deprotection with hydrazine gives primary amine 3.1b, which were Boc protected without further functionalization to afford 3.1c. Alternatively, 3.1b underwent a reductive amination followed by Boc protection to give amines of type 3.1d. Boc protected amine 3.1c can also be alkylated under basic conditions to afford 3.1d. . ,
  • Scheme 3.2 describes an alternate synthesis of amines of type 3.1c and 3.1d.
  • Scheme 3.4 shows the synthesis of alcohols 3.4a and 3.4b from intermediate 3.2b.
  • Decarboxylation of 3.2b using conditions described by Krapcho, ester reduction and alcohol protection affords 3.4a.
  • Lindlar reduction of the alkyne gives homoallylic alcohol 3.4b.
  • Olefins of type 4.3c were accessed via two methods, as described in Scheme 4.3. Suzuki coupling of olefin 3.2f with 1.1b or 1.1 d, followed by oxidation and Wittig homologation gives the desired intermediate. Alternatively, hydroboration of excess 1,5 diene 4.3b, followed by Suzuki coupling enables direct access of 4.3c. Ester hydrolysis gives acid 4.3d, and reduction yields alcohol 4.3e.
  • Etherification of benzylic bromides 2.1d or 2.2c with alcohol 4.1d was accomplished with silver trifluoromethanesulfonate.
  • the azide functionality of adduct 5.1a was reduced under Staudinger conditions. Macroamination yields primary aminopyridine 5.1b. Reductive amination or mono alkylation of the primary amine prior to ring closure gave macroether adducts of type 5.1c.
  • benzyl bromide 2.1d is used for this sequence, further elaboration of the macrocyclic chloroaminopyridines 5.1b and 5.2c is possible utilizing standard Negishi coupling conditions. Deprotection of the Boc group affords adducts 5.1d and 5.1e.
  • Scheme 5.5 Described in Scheme 5.5 is the synthesis of macrocycles of type 5.5b and 5.5c, using a strategy similar to that of Scheme 5.1.
  • the ring closure is accomplished through an intramolecular Heck reaction.
  • Scheme 5.6 Described in Scheme 5.6 is the synthesis of macrocycles of type 5.6b and 5.6c, using a strategy similar to that of Scheme 5.1. The ring closure is accomplished through an intramolecular Heck reaction.
  • Scheme 5.7 is the synthesis of macrocycles of type 5.7b and 5.7c, using a strategy similar to that of Scheme 5.1.
  • the ring closure is accomplished through a palladium catalyzed intramolecular etherification reaction (for conditions, see: Kataoka, N.; Shelby, Q.; Stambuli, J. P.; Hartwig, J. F. J. Org. Chem. 2002, 67, 5553-5566).
  • Scheme 5.8 is the synthesis of macrocycles of type 5.8b and 5.8c, using a strategy similar to that of Scheme 5.1.
  • the ring closure is accomplished through a palladium catalyzed intramolecular etherification reaction (for conditions, see: Kataoka, N.; Shelby, Q.; Stambuli, J. P.; Hartwig, J. F. J. Org. Chem. 2002, 67, 5553-5566).
  • 5.9c can be synthesized utilizing an alternate route.
  • 1,5-pentadiene 3.5b monohydroboration followed by Suzuki coupling affords 5.10a.
  • a second Suzuki coupling with 2.2f affords 5.9c through an alternate route.
  • Described in Scheme 5.11 is the synthesis of macrocycles of type 5.11d and 5.11e.
  • Acid l.lf is alkylated with benzyl bromide derivative 2.2e to afford adduct 5.11a.
  • Bis allylation then gives 5.11b, which can undergo a ring closing metathesis reaction to afford 5.11c.
  • Boc deprotection gives 5.11d.
  • substantially pure means that the isolated material is at least 90% pure, and preferably 95% pure, and even more preferably 99% pure as assayed by analytical techniques known in the art.
  • salts refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids.
  • Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates.
  • Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl- morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.
  • basic ion exchange resins
  • salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids.
  • acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid, and the like.
  • Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids.
  • the present invention is directed to the use of the compounds disclosed herein as inhibitors of ⁇ - secretase enzyme activity or ⁇ -site amyloid precursor protein-cleaving enzyme ("BACE") activity, in a patient or subject such as a mammal in need of such inhibition, comprising the administration of an effective amount of the compound.
  • BACE ⁇ -secretase enzyme
  • ⁇ -site amyloid precursor protein- cleaving enzyme and “BACE” are used interchangeably in this specification. Jh addition to humans, a variety of other mammals can be treated according to the method of the present invention.
  • the present invention is further directed to a method for the manufacture of a medicament or a composition for inhibiting ⁇ -secretase enzyme activity in humans and animals comprising combining a compound of the present invention with a pharmaceutical carrier or diluent.
  • the compounds of the present invention have utility in treating Alzheimer's disease.
  • the compounds may be useful for the prevention of dementia of the Alzheimer's type, as well as for the treatment of early stage, intermediate stage or late stage dementia of the Alzheimer's type.
  • the compounds may also be useful in treating diseases mediated by abnormal cleavage of amyloid precursor protein (also referred to as APP), and other conditions that may be treated or prevented by inhibition of ⁇ -secretase.
  • APP amyloid precursor protein
  • Such conditions include mild cognitive impairment, Trisomy 21 (Down Syndrome), cerebral amyloid angiopathy, degenerative dementia, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type (HCHWA-D), Creutzfeld- Jakob disease, prion disorders, amyotrophic lateral sclerosis, progressive supranuclear palsy, head trauma, stroke, Down syndrome, pancreatitis, inclusion body myositis, other peripheral amyloidoses, diabetes and atherosclerosis.
  • the subject or patient to whom the compounds of the present invention is administered is generally a human being, male or female, in whom inhibition of ⁇ -secretase enzyme activity is desired, but may also encompass other mammals, such as dogs, cats, mice, rats, cattle, horses, sheep, rabbits, monkeys, chimpanzees or other apes or primates, for which inhibition of ⁇ -secretase enzyme activity or treatment of the above noted disorders is desired.
  • the compounds of the present invention may be used in combination with one or more other drugs in the treatment of diseases or conditions for which the compounds of the present invention have utility, where the combination of the drugs together are safer or more effective than either drug alone. Additionally, the compounds of the present invention may be used in combination with one or more other drugs that treat, prevent, control, ameliorate, or reduce the risk of side effects or toxicity of the compounds of the present invention. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with the compounds of the present invention. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to the compounds of the present invention. The combinations may be administered as part of a unit dosage form combination product, or as a kit or treatment protocol wherein one or more additional drugs are administered in separate dosage forms as part of a treatment regimen.
  • combinations of the compounds of the present invention with other drugs in either unit dose or kit form include combinations with anti-Alzheimer's agents, for example other beta-secretase inhibitors or gamma-secretase inhibitors; tau phosphorylation inhibitors; Ml receptor positive allosteric modulators; blockers of A ⁇ oligomer formation; 5-HT modulators, such as PRX-03140, GSK 742467, 1C !l
  • NSAIDs including ibuprofen; vitamin E; anti-amyloid antibodies, including anti-amyloid humanized monoclonal antibodies; anti-inflammatory compounds such as (R)-flurbiprofen, nitroflurbiprofen, rosiglitazone, ND-1251, VP-025, HT-0712 and EHT-202; CB-I receptor antagonists or CB-I receptor inverse agonists; antibiotics such as doxycycline and rifampin; N-methyl-D-aspartate (NMDA) receptor antagonists, such as memantine and neramexane; cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, tacrine, phens
  • NMDA N-methyl-D-aspartate
  • composition as used herein is intended to encompass a product comprising specified ingredients in predetermined amounts or proportions, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • This term in relation to pharmaceutical compositions is intended to encompass a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients.
  • compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for - , K ,, • " u Si O is -
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • compositions include aqueous suspensions, which contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. Oily suspensions may also contain various excipients.
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions, which may also contain excipients such as sweetening and flavoring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension, which may be formulated according to the known art, or may be administered in the form of suppositories for rectal administration of the drug.
  • the compounds of the present invention may also be administered by inhalation, by way of inhalation devices known to those skilled in the art, or by a transdermal patch.
  • pharmaceutically acceptable it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • administering a should be understood to mean providing a compound of the invention to the individual in need of treatment in a form that can be introduced into that individual's body in a therapeutically useful form and therapeutically useful amount, including, but not limited to: oral dosage forms, such as tablets, capsules, syrups, suspensions, and the like; injectable dosage forms, such as IV, IM, or IP, and the like; transdermal dosage forms, including creams, jellies, powders, or patches; buccal dosage forms; inhalation powders, sprays, suspensions, and the like; and rectal suppositories.
  • oral dosage forms such as tablets, capsules, syrups, suspensions, and the like
  • injectable dosage forms such as IV, IM, or IP, and the like
  • transdermal dosage forms including creams, jellies, powders, or patches
  • buccal dosage forms inhalation powders, sprays, suspensions, and the like
  • rectal suppositories rectal suppositories.
  • an effective amount or “therapeutically effective amount” means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • treatment refers to the treatment of the mentioned conditions, particularly in a patient who demonstrates symptoms of the disease or disorder.
  • compositions containing compounds of the present invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • unit dosage form is taken to mean a single dose wherein all active and inactive ingredients are combined in a suitable system, such that the patient or person adminstering the drug to the patient can - . .. • ⁇ -
  • unit dosage forms are tablets or capsules for oral administration, single dose vials for injection, or suppositories for rectal administration. This list of unit dosage forms is not intended to be limiting in any way, but merely to represent typical examples of unit dosage forms.
  • compositions containing compounds of the present invention may conveniently be presented as a kit, whereby two or more components, which may be active or inactive ingredients, carriers, diluents, and the like, are provided with instructions for preparation of the actual dosage form by the patient or person adminstering the drug to the patient.
  • kits may be provided with all necessary materials and ingredients contained therein, or they may contain instructions for using or making materials or components that must be obtained independently by the patient or person administering the drug to the patient.
  • the compounds of the present invention are administered at a daily dosage of from about 0.1 mg to about 100 mg per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form.
  • the total daily dosage is from about 1.0 mg to about 2000 mg, preferably from about 0.1 mg to about 20 mg per kg of body weight. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 mg to about 1,400 mg. This dosage regimen may be adjusted to provide the optimal therapeutic response.
  • the compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
  • Specific dosages of the compounds of the present invention, or pharmaceutically acceptable salts thereof, for administration include 1 mg, 5 mg, 10 mg, 30 mg, 80 mg, 100 mg, 150mg, 300mg and 500 mg.
  • Pharmaceutical compositions of the present invention may be provided in a formulation comprising about 0.5 mg to 1000 mg active ingredient; more preferably comprising about 0.5 mg to 500 mg active ingredient; or 0.5 mg to 250 mg active ingredient; or 1 mg to 100 mg active ingredient.
  • Specific pharmaceutical compositions useful for treatment may comprise about 1 mg, 5 mg, 10 mg, 30 mg, 80 mg, 100 mg, 150 mg, 300mg and 500 mg of active ingredient.
  • FRET fluorescence resonance energy transfer
  • a homogeneous end point fluorescence resonance energy transfer (FRET) assay is employed with the substrate ([TAMRA-S-CO-EEISEVNLDAEF-NHQSY] QFRET), which is cleaved by BACE 1 to release the fluorescence from TAMRA.
  • the Km of the substrate is not determined due to the limit of solubility of the substrate.
  • a typical reaction contains approximately 30 nM enzyme, 1.25 ⁇ M of the substrate, and buffer (50 mM NaOAc, pH 4.5, 0.1 mg/ml BSA, 0.2% CHAPS, 15 mM EDTA and 1 mM deferoxamine) in a total reaction volume of 100 ⁇ l.
  • the reaction is proceeded for 30 min and the liberation of TAMRA fragment is measured in a 96-well plate LJL Analyst AD using an excitation wavelength of 530 nm and an emission wavelength of 580 nm. Under these conditions, less than 10% of substrate is processed by BACE 1.
  • the enzyme used in these studies is soluble (transmembrane domain and cytoplasmic extension excluded) human protein produced in a baculovirus expression system.
  • solutions of inhibitor in DMSO four concentrations of the inhibitors are prepared: ImM, 100 ⁇ M, 10 ⁇ M, 1 ⁇ M) are included in the reactions mixture (final DMSO concentration is 0.8%). All experiments are conducted at rt using the standard reaction conditions described above.
  • HPLC assay A homogeneous end point HPLC assay is used with the substrate (coumarin-CO-REVNFEVEFR), which is cleaved by BACE 1 to release the N-terminal fragment attached with coumarin.
  • the Km of the substrate is greater than 100 ⁇ M and can not be determined due to the limit of solubility of the substrate.
  • a typical reaction contains approximately 2 nM enzyme, 1.0 ⁇ M of the substrate, and buffer (50 mM NaOAc, pH 4.5, 0.1 mg/ml BSA, 0.2% CHAPS, 15 mM EDTA and 1 mM deferoxamine) in a total reaction volume of 100 ⁇ l.
  • the reaction is proceeded for 30 min and is stopped by the addition of 25 ⁇ L of 1 M Tris-HCl, pH 8.0.
  • the resulting reaction mixture is loaded on the HPLC and the product is separated from substrate with 5 min linear gradient. Under these conditions, less than 10% of substrate is processed by BACE 1.
  • the enzyme used in these studies is soluble (transmembrane domain and cytoplasmic extension excluded) human protein produced in a baculovirus expression system.
  • solutions of inhibitor in DMSO (12 concentrations of the inhibitors are prepared and the concentration rage is dependent on the potency predicted by FRET) are included in the reaction mixture (final DMSO concentration is 10 %). All experiments are conducted at rt using the standard reaction conditions described above.
  • To determine the IC50 of the compound four parameters equation is used for curve fitting. The errors in reproducing the dissociation constants are typically less than two-fold.
  • the compounds of the following examples had activity in inhibiting the beta- secretase enzyme in the aforementioned assay, generally with an IC50 from about 1 nM to 100 ⁇ M. Such a result is indicative of the intrinsic activity of the compounds in use as inhibitors of the beta- secretase enzyme activity.
  • HMDS hexamethyldisilazane
  • BSA bovine serum albumin
  • Methyl 2,6-dichloroisonicotinate (10 g, 48.5 mmol), methyl(propylsulfonyl)amine (7.99 g, 58.2 mmol), potassium phosphate (14.4 g, 68 mmol), Xantphos (1.69 g, 2.9 mmol) and tris(dibenzylideneacetone)dipalladium (0.89 g, 0.97 mmol) were added to a dry, argon flushed flask.
  • Step A Sulfonamide reduction Performed as described in Step A of the synthesis of Intermediate H2.C.1, with mesyl methyl sulfonamide being used in place of propyl methyl sulfonamide.
  • Step B Reduction Performed as described in Step B of the synthesis of Intermediate H2.C.1.
  • Step C Silyl ether formation N-f ⁇ -chloro ⁇ -Oiydroxymethy ⁇ pyridin ⁇ -yy-N-methylmethanesulfonamide (2.8 g, 11.1 mmol) from Step B, imidazole (0.91 g, 13.4 mmol), and tert-butyldimethylsilyl chloride (1.85 g, 12.2 mmol) were dissolved in anhydrous methylene chloride (25 mL) and allowed to stir at 25 0 C for 16 hours.
  • Step A Mono-esterif ⁇ cation of diacid 3-Phenylglutaric acid (5 g, 24 mmol) and cesium carbonate (3.9 g, 12 mmol) were dissolved in 200 mL of anhydrous DMF and cooled to 0 0 C. Methyl iodide (1.5 mL, 24 mmol) was added via syringe to the solution, which was allowed to slowly warm to 25 0 C over 16 hours. The reaction was diluted with water, and the pH was adjusted to ⁇ 9 with sat'd NaHCO3.
  • 5-methoxy-5-oxo-3-phenylpentanoic acid (4.2 g, 19 mmol) was dissolved in 200 mL of anhydrous THF and cooled to 0 0 C.
  • Borane (57 mL, 57 mmol, 1 M soln in THF) was added via syringe to the solution, which was allowed to slowly warm to 25 0 C over 16 hours.
  • the reaction was then cooled to 0 0 C and quenched with methanol (25 mL), followed by water (25 mL), followed by saturated solution sodium bicarbonate (25 mL).
  • the product was extracted into ethyl acetate, washed with brine, dried over sodium .
  • Step C Iodination of Alcohol Triphenylphosphine (1.5 g, 5.8 mmol) and imidazole (0.39 g, 5.8 mmol) were added to a dried flask under argon atmoshphere. Anhydrous methylene chloride (50 mL) was added and the solution was cooled to 0 0 C. Iodine (1.45 g, 5.8 mmol) was added in one portion, and the resulting solution was stirred at 0 0 C for 0.5 h.
  • Step A Suzuki Coupling To 0.70Og (2.25 mmol, 1 equiv.) of (but-3-en-l-yloxy)(tert-buryl)diphenylsilane was added 5.9 mL
  • reaction was cooled to rt and filtered through a pad of celite, rinsing with EtOAc. Partitioned filtrate between EtOAc and brine, separated and washed organics with brine, dried over Na 2 SO 4 , filtered and concentrated. Purified residue using silica gel chromatography to isolate desired product as a viscous oil.
  • Step B Silyl Deprotection ⁇ is .•• ⁇ " » »-,: B !? n- ...
  • Step C Allylation To a solution of ⁇ 2-chloro-6-[methyl(methylsulfonyl)amino]pyridine-4-yl ⁇ methyl 3-b ⁇ omo-N-(tert- butoxycarbonyl)- ⁇ -methylphenylalaninate (400 mg, 0.68 mmol) and allyltributylstannane (0.48 mL, 1.6 mmol) in degassed DMF (4 mL) was added bis(triphenylphosphine)palladium( ⁇ ) dichloride (33 mg, 0.048 mmol). The reaction was heated to 120 0 C in a microwave for 0.5 h. Additional catalyst was added and heating was repeated 3x to drive the reaction to completion.
  • Step C Macroamination To a solution of product from Step B (0.015 g, 0.025 mmol, 1 equiv) in 0.60 mL DMA was added
  • Step B and Step C Macroamination and Boc deprotection
  • Step A Zincate coupling of intermediate A and B
  • Step B Reduction of Ester methyl 5- ⁇ 4-( ⁇ [tert-butyl(dimethyl)silyl]oxy ⁇ methyl)-6- ⁇ methyl(methylsulfonyl)amino]pyridin-2-yl ⁇ -3- phenylpentanoate (0.55 g, 1.05 mmol) from step A was dissolved in anhydrous THF (10 mL) and cooled to 0 0 C under argon atmosphere. Lithium borohydride (0.74 mL, 1.49 mmol, 2.0 M solution in THF) was slowly added via syringe. After addition was complete, the temperature was raised to 45 0 C and the reaction was allowed to stir at that temperature for 16 h.
  • Step D Zincate coupling of intermediate I.l.c.l and scaffold r Ii ⁇ ⁇ >u? w 1 ⁇ u8 S ,. ⁇ -Ii ET « * C n »TMi"
  • Step F Hydrolysis of Ester
  • a IM solution of LiOH in water 0.145 mL (0.291 mmol) of a IM solution of LiOH in water. The solution was allowed to stir overnight at 45 0 C for 16 h.
  • Step B Suzuki with product from Step A and intermediate 2.2f Prepared as described in Step A.
  • Step C-F Silyl Deprotection, Ester hydrolysis, Mitsunobu macrolactonization and Boc deprotection sequence performed as described in the synthesis of Example 9.
  • LC/MS [M+H] 446
  • Step A Suzuki with 3 -Methyl- 1,4-pentadiene and 1.1.c.1 Prepared as Step A in the synthesis of Example 9.

Abstract

The present invention is directed to macrocyclic aminopyridyl compounds represented by general formula (I), which are inhibitors of the beta-secretase enzyme and that are useful in the treatment of diseases in which the beta-secretase enzyme is involved, such as Alzheimer's disease. The invention is also directed to pharmaceutical compositions comprising these compounds and the use of these compounds and compositions in the treatment of such diseases in which the beta-secretase enzyme is involved.

Description

TITLE OF THE INVENTION
MACROCYCLIC AMINOPYRIDYL BETA-SECRETASE INHIBITORS FOR THE TREATMENT OF
ALZHEIMER'S DISEASE
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) of U.S. provisional application serial no. 60/630,319, filed November 23, 2004.
FIELD OF THE INVENTION The invention is directed to the field of compounds which are inhibitors of the the activity of the β-secretase enzyme, and to the use of the compounds for the treatment of diseases in which the β- secretase enzyme is involved, such as Alzheimer's disease.
BACKGROUND OF THE INVENTION Alzheimer's disease is characterized by the abnormal deposition of amyloid in the brain in the form of extra-cellular plaques and intra-cellular neurofibrillary tangles. The rate of amyloid accumulation is a combination of the rates of formation, aggregation and egress from the brain. It is generally accepted that the main constituent of amyloid plaques is the 4kD amyloid protein (βA4, also referred to as Aβ, β-protein and βAP) which is a proteolytic product of a precursor protein of much larger size. The amyloid precursor protein (APP or AβPP) has a receptor-like structure with a large ectodomain, a membrane spanning region and a short cytoplasmic tail. The Aβ domain encompasses parts of both extra-cellular and transmembrane domains of APP, thus its release implies the existence of two distinct proteolytic events to generate its NH2- and COOH-termini. At least two secretory mechanisms exist which release APP from the membrane and generate soluble, COOH-trancated forms of APP (APPS). Proteases that release APP and its fragments from the membrane are termed
"secretases." Most APP5 is released by a putative α-secretase which cleaves within the Aβ protein to release α-APPs and precludes the release of intact Aβ. A minor portion of APP3 is released by a β- secretase ("β-secretase"), which cleaves near the NH2-terminus of APP and produces COOH-terminal fragments (CTFs) which contain the whole Aβ domain. Thus, the activity of β-secretase or β-site amyloid precursor protein-cleaving enzyme ("BACE") leads to the abnormal cleavage of APP, production of Aβ, and accumulation of β amyloid plaques in the brain, which is characteristic of Alzheimer's disease (see R. N. Rosenberg, Arch. Neurol., vol. 59, Sep 2002, pp. 1367-1368; H. Fukumoto et al, Arch. Neurol., vol. 59, Sep 2002, pp. 1381-1389; J.T. Huse et al, J. Biol. Chem., vol 277, No. 18, issue of May 3, 2002, pp. 16278-16284; K.C. Chen and WJ. Howe, Biochem. Biophys. Res. Comm, vol. 292, pp 702-708, 2002). Therefore, therapeutic agents that can inhibit β-secretase or BACE may be useful for the treatment of Alzheimer's disease. .
IF IU. Il . ' W .,.„» 1!,,« .„„!' , ' II ii li...« ,,,.,μ ,„,
The compoimds of the present invention are useful for treating Alzheimer's disease by inhibiting the activity of β-secretase or BACE, thus preventing the formation of insoluble Aβ and arresting the production of Aβ.
SUMMARY OF THE INVENTION
The present invention is directed to novel macrocyclic aminopyridyl compounds represented by general formula (I)
Figure imgf000003_0001
(I)
and pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof, which are useful as inhibitors of the β-secretase enzyme.
The invention is also directed to pharmaceutical compositions which include an effective amount of a compound of formula (T), or pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof, and a pharmaceutically acceptable carrier. The invention is also directed to methods of treating mammals for diseases in which the β-secretase enzyme is involved, such as Alzheimer's disease, and the use of the compounds and pharmaceutical compositions of the invention in the treatment of such diseases.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is directed to compounds of formula (I):
Figure imgf000003_0002
(I)
and pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof, wherein: .
Il - It.* I! ,.' <[J .,,,,I! IUI ...Jt . ' TT IU.. IU. „..11 »»J'
Y is selected from the group consisting of
(1) hydrogen,
(2) -C 1-3 alkyl, wherein said alkyl is optionally substituted with one or more halogen, (3) halogen, and
(4) cyano;
A is selected from the group consisting of
(1) hydrogen, (2) -Ci-io alkyl,
(3) -C2-10 alkenyl, and (4) -C2-lθ alkynyl wherein said alkyl, alkenyl or alkynyl is unsubstituted or substituted with one or more
(a) halo, (b) -C3-8 cycloalkyl,
(c) -OH,
(d) -CN,
(e) -O-Ci-io alkyl,
(f) -C6-10 aryl, or (g) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
and said aryl and heteroaryl groups are unsubstituted or substituted with one or more (i) halo, (Ii) -OH,
(Ui) -CN, (iv) -0-Ci_io alkyl,
(v) -Ci-10 alkyl, (vi) -C2-10 alkenyl, (vii) -C2-IO alkynyl, or (viii) -C3_8 cycloalkyl;
Rl is selected from the group consisting of (1) -C6-10 arylene, or j ..' a"r -iu-,,> !!.,« , „-,.« „5-!
(2) heteroarylene selected from the group consisting of divalent pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benziniidazolyl, indynyl and benzoxazolyl,
wherein said arylene or heteroarylene is unsubstituted or substituted with one or more (a) halo,
(b) -Ci-io alkyl, (c) -C2-10 alkenyl, (d) -C2-10 alkynyl,
(e) -OH, (f) -CN,
(g) -0-Ci.io alkyl, or (h) -C3.8 cycloalkyl;
R2 is selected from the group consisting of: (1) (R5-SO2)N(R6)-, wherein R5 is (a) -Ci-10 alkyl, (b) -C2-10 alkenyl, (c) -C2-IO alkynyl,
(d) -C3-8 cycloalkyl,
(e) -C6-10 aryl, or
(f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
wherein said alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heteroaryl is unsubstituted or substituted with one or more
(i) halo,
(ϋ) -OH,
(iii) -CN,
(iv) -0-Ci_io alkyl, (v) -Ci-io alkyl,
(vi) -C2-IO alkenyl,
(vii) -C2-10 alkynyl, (viii) -C3-8 cycloalkyl, (ix) -C6-10 aryl, or
(x) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, and said aryl and heteroaryl is unsubstituted or substituted with one or more (A) halo,
(B) -OH,
(C) -CN, (D) -O-Ci-iθ alkyl,
(E) -C3-8 cycloalkyl, (F) -Ci-iO alkyl,
(G) -C2-IO alkenyl, or (H) -C2-IO alkynyl;
R6 is selected from the group consisting of (a) hydrogen,
(b) -Cl-10 alkyl,
(c) -C2-IO alkenyl,
(d) -C2-IO alkynyl,
(e) -C6-10 aryl, or (f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
wherein said alkyl, alkenyl, alkynyl, aryl or heteroaryl. is unsubstituted or substituted with one or more (i) halo, (ϋ) -OH, (iii) -CN,
(iv) -0-Ci_io alkyl, (v) -C3-8 cycloalkyl, r ιuι>
(vi) -C6-10 aryl, or
(vii) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl;
wherein said cycloalkyl, aryl or heteroaryl is unsubstituted or substituted with one or more (A) halo,
(B) -OH, (C) -CN,
(D) -O-Ci-IO alkyl, (E) -C3.8 cycloalkyl, or (F) -C6-IO aryl,
or R5 and K.6 may be linked to form a group -CH2(CH2)pCH2~,
(2) -C6-10 aryl, wherein said aryl is unsubstituted or substituted with one or more (i) halo,
(ϋ) -OH,
(iii) -CN,
(iv) -0-Ci_io alkyl,
(v) -C3_8 cycloalkyl, (vi) -Ci-10 alkyl,
(vi) -C6-lo aryl,
(3)
Figure imgf000007_0001
(4) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl,pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, wherein said heteroaryl is unsubstituted or substituted with one or more „ ■ • . i - i i • - -
H !(.,.!. is ,.' '!,.,!■ .,,,.Ii im ,„„11 ,•' ir ir,.,., .....ii ...,.n
(i) halo,
(ii) -OH,
(iii) -CN,
(iv) -0-Ci_io alkyl,
(v) -C3.8 cycloalkyl,
(vi) -Ci-io alkyl,
(vii) -CC=O)-O-Ci-I0 alkyl,
(viii) -C(=O)-OH, and
Figure imgf000008_0001
(x) -NRCRd, wherein Rc and Rd are selected from the group consisting of
(A) hydrogen, and
(B) -Ci-IO alkyl,
(5) hydrogen, and
(6) -CF3;
R3 i is
Figure imgf000008_0002
wherein if the dotted line leading to Q3 is absent, then Q^ is selected from the group consisting of (a) -CH2-
Cb) -O-,
(c) -NRX-,
(d) -C(=O)- and
(e) -C(=O) -NRX-, wherein Rχ is selected from the group consisting of t .• .. - ~ ■■■
IC" U...1. It .■' 'I...!' ..,,,It It.JJ ,,m!t .. I!" II,,,.. «»~ --.I' --I'
(i) hydrogen, (ii) -Ci-IO alkyl, (iii)-C2-10 alkenyl, (iv) -C2-IO alkynyl, (v) -C3-8 cycloalkyl,
(vi) -Co-6 alkylene-C6-10 aryl,
(vii) -Co-6 alkylene-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, (viii) -Co-6 alkyl-C3_8 cycloalkyl,
and if the dotted line leading to Q3 represents a bond, then Q3 is -CH- or -CH2CH-,
if the dotted line leading to RY is absent, then RY is selected from the group consisting of
(a) hydrogen, (b) -Ci-iO alkyl,
(c)-C2-lθ alkenyl, (d) -C2-10 alkynyl,
(e) -C3-8 cycloalkyl,
(f) -Co-6 alkylene-C6-10 aryl, or
(g) -Co-6 alkylene-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
and if the dotted line leading to Ry represents a bond, then Ry is selected from the group consisting of (a) =CH-Ci_io alkyl,
(b) =CH-Co-6 alkyl-C6-lO aryl,
wherein said alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heteroaryl RY groups are unsubstituted or substituted with one or more (i) halo,
(ii) -Ci-io alkyl,
(iii) -OH, , u ___. ... .
(iv) -CN, or (v) -0-Ci-io alkyl, or
(vi) -C3_8 cycloalkyl,
Q4 is selected from the group consisting of
Ca) -CH2 - Cb) -O-, and (c) -NRZ- wherein Rz is selected from the group consisting of (i) hydrogen,
(ii) -Ci-io alkyl, (iii)-C2.10 alkenyl, (iv) -C2-IO alkynyl, (v) -C3.8 cycloalkyl, (vi) -Cθ-6 alkylene-Cβ-io aryl, or
(vii) -Cθ-6 alkylene-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
R4 is -(CH2)S-Q^ -(CH2)t, wherein Q2 is selected from the group consisting of
(I)-O-,
(2)-NH-, (3) -O-C(=O>,
C4) -CC=O)-O-,
(S) -NHCC=O)-,
(6) -CC=O)-NH-,
(7) -CH=CH-, (8) -CC=O)-,
Figure imgf000010_0001
(H) r-- L,> if „ Lif ^ Lif ^
Figure imgf000011_0001
m is 0, 1 or 2; n is 0, 1 or 2; p is 1, 2 or 3; q is 1, 2, 3, 4 or 5; r is 0, 1 or 2; s is 0 or 1; t is 0 or 1; and u is 0, 1 or 2.
In a preferred embodiment of the compounds of formula (I), Y is hydrogen.
In another preferred embodiment of the compounds of formula (I), Rl is unsubstituted or substituted -Cg-io arylene, preferably unsubstituted phenylene.
In another preferred embodiment of the compounds of formula (I), R.4 is -(CH2)S-Q2 -(CEtøt, wherein Q2 is selected from the group consisting of
(I)-O-,
(2) -O-C(=O)-,
Figure imgf000011_0002
When Q2 is selected from -O-, -NH-, or -C(=O)-, then s and t are each preferably 1. When Q2 is selected from -O-C(=O)- or -NHC(=O)-, then s is preferably 0 and t is preferably 1. When Q2 is selected from -C(=O)-O- or -(C=O)-NH-, then s is preferably 1 and t is preferably 0. When Q2 is -CH=CH, then preferably s is 0 and t is l,or s is 1 and t is 0. When Q2 is selected from the group consisting of
Figure imgf000011_0003
Figure imgf000012_0001
then s and t are preferably 0.
In preferred embodiments of the compounds of formula (T), A is selected from the group consisting of (1) hydrogen, and
(2) -Ci-iθ alkyl, wherein said alkyl is unsubstituted or substituted with one or more
(a) halo,
(b) -C3-8 cycloalkyl, (c) -CN
Cd) -O-Ci-IO alkyl,
(e) phenyl, or
(f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl. In a more preferred embodiment of the compounds of the formula (I), A is -Cl-10 alkyl
(preferably methyl), wherein said alkyl is unsubstituted or substituted with one or more halo (preferably fiuoro).
In a preferred embodiment of the compounds of formula (I), R2 is selected from the group consisting of (R5-Sθ2)N(R6)-, wherein R5 is -C \.β alkyl, wherein said alkyl is unsubstituted or substituted with one or more
(i) halo, (ii) -OH,
Figure imgf000012_0002
(iv) -O-Ci-6 alkyl, or (v) -Ci_6 alkyl,
R6 is selected from the group consisting of (a) hydrogen,
(b) -Ci_6 alkyl, or Cc) -Ce-IO aiyl, wherein said alkyl and aryl is unsubstituted or substituted with one or more
(i) halo, (ii) -OH,
(iii) -CN, , ,
S- L ii ..'' 'ij Zs ϋjf 3t ,/ -Sr E-. (E^ J .Si
(iv) -O-Ci_6 alkyl, (v) -Ci.6 alkyl, or R5 and Rδ are linked to form a group -CH2(CH2)pCH2-
Another preferred R^ group is-C6-10 aryl5 unsubstituted or substituted as described above. Preferred aryl groups are phenyl groups, unsubstituted or substituted with cyano. A preferred R2 substituent is shown below:
Figure imgf000013_0001
Another preferred R^ substituent is
Figure imgf000013_0002
wherein q is 1, 2 or 3.
Another preferred R2 substituent is heteroaryl, either unsubstituted or substituted as described above. A preferred heteroaryl group is furanyl or oxazolyl, either unsubstituted or substituted as described above. A preferred furanyl or oxazolyl substituent is depicted below:
Figure imgf000013_0003
wherein Ql is selected from the group consisting of
(a) N, and
(b) C-Rb, wherein Rb is selected from the group consisting of
(i) -CN, and (ii) -CC=O)-O-Ci-IO alkyl,
(iii) -C(=O)-OH, and (iv) -C(=O)-NRCRd,
(v) -NR0Rd, wherein Rc and Rd are selected from the group consisting of (A) hydrogen, and
(B) -Ci-io alkyl.
In one embodiment of the compounds of formula (I) when the dottled line leading to Q3 is absent and Q3 is NRX, Rx is preferably hydrogen, and n is preferably 1. .
Il " t|--ι If ,.' 1[,,J' .,.,,1' «,„[( ,„„1' ,. Il !!„,„ IW ,„»11 B-J<
In an alternative embodiment of the compounds of formula (I) when the dottled line leading to Q3 is absent, Q3 is -O - and n is preferably 1.
In another embodiment when the dottled line leading to Q3 is -C(=O) -NRχ, Rχ is preferably hydrogen, and n is preferably 1. In certain embodiments, the dotted line leading to RY is absent and Ry is selected from the group consisting of
(a) hydrogen,
(b) -Ci-io alkyl, or
(c) -Cθ-6 alkyl-C6-10 aryl, wherein said alkyl or aryl or unsusbsituted or substituted with one or more halo (preferably fluoro).
In preferred embodiments, Q4 is CH2, m is preferably 1 and r is preferably 0.
In another embodiment, the invention is directed to compounds of Formula (JT):
Figure imgf000014_0001
(N) and pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof, wherein A, Y, R2, R3 and R4 are as defined above.
Another embodiment of the present invention includes a compound which is selected from the title compounds of the following Examples and pharmaceutically acceptable salts thereof.
As used herein, the term "alkyl," by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C i _ 10 alkyl means an alkyl group having from one to ten carbon atoms). Preferred alkyl groups for use in the invention are Cl-6 alkyl groups, having from one to six carbon atoms. Exemplary alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, pentyl, hexyl, and the like.
As used herein, the term "alkylene," by itself or as part of another substituent, means a saturated straight or branched chain divalent hydrocarbon radical having the number of carbon atoms designated. The term Co alkylene (for example, in the radical "-Coalkylene-C6-lθ aryl") means that the alkylene group is absent.
As used herein, the term "cycloalkyl," by itself or as part of another substituent, means a saturated cyclic hydrocarbon radical having the number of carbon atoms designated (e.g., C3.8 cycloalkyl means a cycloalkyl group having from three to eight carbon atoms). Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like. IU-.. ....J' ,
As used herein, the term "alkenyl," by itself of as part of another substituent, means a straight or branched chain hydrocarbon radical having a single carbon-carbon double bond and having the number of carbon atoms designated (e.g., C2-10 alkenyl means an alkenyl group having from one to ten carbon atoms). Preferred alkenyl groups for use in the invention are C2-6 alkenyl groups, having from two to six carbon atoms. Exemplary alkenyl groups include ethenyl, n-propenyl, isopropenyl, butenyl, and the like.
As used herein, the term "alkynyl", by itself or as part of another substituent, means a saturated straight or branched chain hydrocarbon radical having the number of carbon atoms designated (e.g., C2-10 alkynyl means an alkynyl group having from two to ten carbon atoms). Preferred alkynyl groups for use in the invention are C2-6 alkynyl groups, having from two to six carbon atoms. Exemplary alkynyl groups include ethynyl and propynyl.
As used herein, the term "aryl," by itself or as part of another substituent, means an aromatic or cyclic radical having the number of carbon atoms designated (e.g., Cg_io aryl means an aryl group having from six to ten carbons atoms). The term "aryl" includes multiple ring systems as well as single ring systems. Preferred aryl groups for use in the invention include phenyl and naphthyl. As used herein, the term "arylene," by itself or as part of another substituent, means a divalent aromatic or cyclic radical, having the number of carbon atoms designated (e.g., C6-10 arylene means an arylene group having from six to ten carbons atoms). The term "arylene" includes multiple ring systems as well as single ring systems. Preferred arylene groups for use in the invention include phenylene and naphthylene. As used herein, the term "heteroaryl," by itself or as part of another substituent, means an aromatic cyclic radical having at least one ring heteroatom (O, N or S). The term "heteroaryl" includes multiple ring systems as well as single ring systems. Exemplary heteroaryl groups for use in the invention include furyl, pyranyl, benzofuranyl, isobenzofuranyl, chromenyl, thienyl, benzothiophenyl, pyrrolyl, pyrazolyl, imidazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolyl, benzimidazolyl, quinolinyl, isoquinolinyl, tetrazolyl, indazolyl, napthyridinyl, triazolyl, oxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isoxazolyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indynyl and dihydroindolyl.
When a heteroaryl group as defined herein is substituted, the substituent may be bonded to a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits substitution. Preferably, the substituent is bonded to a ring carbon atom. Similarly, when a heteroaryl group is defined as a substituent herein, the point of attachment may be at a ring carbon atom of the heteroaryl group, or on a ring heteroatom (i.e., a nitrogen, oxygen or sulfur), which has a valence which permits attachment. Preferably, the attachment is at a ring carbon atom.
As used herein, the term "heteroarylene," by itself or as part of another substituent, means an aromatic cyclic divalent radical having at least one ring heteroatom (O, N or S). The term "halo" or "halogen" includes fluoro, chloro, bromo and iodo.
The compounds of the instant invention have at least one asymmetric center. Additional asymmetric centers may be present depending upon the nature of the various substituents on the ir iL if .< ''Li1 ΪJi iJ .".3 ,.' ' ' rE".'. iC' J .,,"S
molecule. Compounds with asymmetric centers give rise to enantiomers (optical isomers), diastereomers (configurational isomers) or both, and it is intended that all of the possible enantiomers and diastereomers in mixtures and as pure or partially purified compounds are included within the scope of this invention. The present invention is meant to encompass all such isomeric forms of these compounds.
The independent syntheses of the enantiomerically or diastereomerically enriched compounds, or their chromatographic separations, may be achieved as known in the art by appropriate modification of the methodology disclosed herein. Their absolute stereochemistry may be determined by the x-ray crystallography of crystalline products or crystalline intermediates that are derivatized, if necessary, with a reagent containing an asymmetric center of known absolute configuration.
If desired, racemic mixtures of the compounds may be separated so that the individual enantiomers are isolated. The separation can be carried out by methods well known in the art, such as the coupling of a racemic mixture of compounds to an enantiomerically pure compound to form a diastereomeric mixture, followed by separation of the individual diastereomers by standard methods, such as fractional crystallization or chromatography. The coupling reaction is often the formation of salts using an enantiomerically pure acid or base. The diastereomeric derivatives may then be converted to the pure enantiomers by cleavage of the added chiral residue. The racemic mixture of the compounds can also be separated directly by chromatographic methods utilizing chiral stationary phases, which methods are well known in the art. Alternatively, any enantiomer of a compound may be obtained by stereoselective synthesis using optically pure starting materials or reagents of known configuration by methods well known in the art.
The compounds of the present invention are prepared by the methods outlined in Schemes 1.1 to 5.8, below, and the intermediates and examples herein. The compounds of the present invention are prepared by the methods outlined in Schemes 1.1-5.8, illustrated below.
Scheme 1.1, describes the preparation of derivatives of type 1.1a, as well as the corresponding trifiate analogs 1.1b and 1.1c. Starting from glycine Schiff base, more elaborated bromides of type l.ld and l.le were prepared. IUn il .'' 1U '-Jt <!...!' ..ml! ,•" " I! Hv, St.,,, ,nV „.„11
Scheme 1.1
Figure imgf000017_0001
Figure imgf000017_0002
Scheme 2.1 describes a sequence leading to 2,6-dichloro pyridine derivatives from 2,6- dichoroisonicotinate. Methyl ester hydrolysis gives acid 2.1a, which was transformed via a two step sequence to acylhydrazide 2.1b. Benzyl alcohol 2.1c is readily accessed via ester reduction, and was transformed to the corresponding benzyl bromide with CBr4/Ph3P.
ir ii..j' ir ..' pι,,j' .,„.!! IU' ...J ,•' 1.,., ...» ι«jι
Scheme 2.1
Figure imgf000018_0001
2.1c 2.1d
Sulfonylation of 2,6-dichloroisinicotinate gave key intermediate 2.2a, which was then be transformed to acid 2.2b or alcohol 2.2c as described above. Acylhydrazide 2.2d was derived from acid 2.2b while alcohol 2.2c was further advanced to bromide 2.2e. Alternatively, siliyl protection of alcohol 2.2c affords silyl ether 2.2f.
Scheme 2.2
Figure imgf000019_0001
LiBH4 1. EDC, BocNHNH2 2. HCI
Figure imgf000019_0002
2.2f
Homo-allylic amies of type 3.1b were prepared via conjugate addition of phthalimide to α,β-unsaturated aldehydes, followed by Wittig homologation to give compounds of type 3.1a, as described in Scheme 3.1 (See Bergman, E. D., Migron, Y. Organic Preparations and Procedures Int. 1976, 8, 75-80). Pthalimide deprotection with hydrazine gives primary amine 3.1b, which were Boc protected without further functionalization to afford 3.1c. Alternatively, 3.1b underwent a reductive amination followed by Boc protection to give amines of type 3.1d. Boc protected amine 3.1c can also be alkylated under basic conditions to afford 3.1d. . ,
|i l|,-i» II ..' W U' 1U' J J1 IP !U. tϊ,.', ,,;]' ,«",B
Scheme 3.1
1. Pthalimide
Ry CHO 2. Ph3PMeBr, Base
Figure imgf000020_0001
1. Reductive Amination .
Figure imgf000020_0002
3.1d 3.1c
Scheme 3.2 describes an alternate synthesis of amines of type 3.1c and 3.1d.
Condensation of diethylmalonate with an aldehyde provides the requisite α,β-unsaturated system 3.2a. Conjugate addition of TMS acetylene Grignard reagent, followed by desilylation affords 3.2b. Ester hydrolysis under basic conditions, followed by Curtius rearrangement affords homo propargylamine 3.2c. Lindlar reduction gives access to 3.1c, which was further alkylated to give 3.1d.
Scheme 3.2
Figure imgf000020_0003
3.1d 3.1c 3.2c 3.2b
Conjugate addition of vinylmagnesium bromide anion to 3.3a, followed by nitro reduction and Boc protection as described in Scheme 3.3 affords another route to 3.1b. As described in Scheme 3.1, this route also enables further substitution of the Boc protected primary amine. , t __ _ _! " i
I' ''. I il ■•' 'L.J' >,'m(l If.Ji ,r,,L[ "It i! -.. t!,u, ,,»,\t ,,u,ii
Scheme 3.3
Figure imgf000021_0001
Scheme 3.4 shows the synthesis of alcohols 3.4a and 3.4b from intermediate 3.2b. Decarboxylation of 3.2b using conditions described by Krapcho, ester reduction and alcohol protection affords 3.4a. Lindlar reduction of the alkyne gives homoallylic alcohol 3.4b.
Scheme 3.4
Figure imgf000021_0002
3.4a 3.4b
3.2b
As shown in Scheme 3.5, glutaπc acid derivatives of type 3.5a were monoesterifϊed. Acid reduction, followed by iodination fo the newly generated alcohol afforded iodides of type 3.5b.
Scheme 3.5
Figure imgf000021_0003
Coupling of intermediates 1.1b and l.ld with terminal olefin 4.1a (For synthesis, see: Boeckman, R. K., Jr.; Charette, A. B.; Asberom, T.; Johnston, B. H. J. Am. Chem. Soc, 1991, 113,
5337-53) can be accomplished using Suzuki conditions to give adduct 4.1b. Ester reduction with LiBH4 gives alcohol 4.1d, while saponification affords acid 4.1e. Scheme 4.1
Figure imgf000022_0001
1. TBAF
2. CBr4, Ph3P
3. NaN3
Figure imgf000022_0002
4.1d 4.1c 4.1 e
Intermediates of type 4.2b and 4.2c can be synthesized using a protocol similar to that described above, as shown in Scheme 4.2. This route can be utilized with either a primary or seconday Boc protected amine.
i ,.
Il <U» Il ..' 'I,,,!' .„. ff IUK ... J .i1 ir iC Bm.. -. J1 iimP
Scheme 4.2
Figure imgf000023_0001
Olefins of type 4.3c were accessed via two methods, as described in Scheme 4.3. Suzuki coupling of olefin 3.2f with 1.1b or 1.1 d, followed by oxidation and Wittig homologation gives the desired intermediate. Alternatively, hydroboration of excess 1,5 diene 4.3b, followed by Suzuki coupling enables direct access of 4.3c. Ester hydrolysis gives acid 4.3d, and reduction yields alcohol 4.3e.
.
Scheme 4.3
Figure imgf000024_0001
4.3e 4.3d
As shown in Scheme 4.4 intermediates of type 4.4a and 4.4b was accessed using a procedure similar to that described in Scheme 4.2.
Scheme 4.4
Figure imgf000024_0002
Etherification of benzylic bromides 2.1d or 2.2c with alcohol 4.1d was accomplished with silver trifluoromethanesulfonate. The azide functionality of adduct 5.1a was reduced under Staudinger conditions. Macroamination yields primary aminopyridine 5.1b. Reductive amination or mono alkylation of the primary amine prior to ring closure gave macroether adducts of type 5.1c. When benzyl bromide 2.1d is used for this sequence, further elaboration of the macrocyclic chloroaminopyridines 5.1b and 5.2c is possible utilizing standard Negishi coupling conditions. Deprotection of the Boc group affords adducts 5.1d and 5.1e.
Scheme 5.1
Figure imgf000025_0001
2. Reductive amination
Figure imgf000025_0002
, ( ...,.t , μ- IUi. Ii ..' 1Ut' ";,7i if,, ii ,,,Ziι „ % id -w u,,s ,,Jι
The synthesis of macrolactones such as 5.2d and 5.2e is depicted in Scheme 5.2, utilizing a similar strategy as described for Scheme 5.1. The central coupling reaction is performed using CsCO3 base.
Scheme 5.2
Figure imgf000026_0001
1. Reductive amination
Figure imgf000026_0002
Syntheses of macroethers of type 5.3b and 5.3c, and macroesters of type 5.4b and 5.4b and 5.4c are depicted in Schemes 5.3 and 5.4 respectively. The strategy utilized is very similar to that , -
discussed in Scheme 5.1, where ring closure is accomplished through a macroamination strategy. When benzyl bromide 2.1d is utilized in the first step of these Schemes, the resulting 2-chloroaminopyridine functionlaity of 5.3b and 5.4b can be further functionlaized after tertiarycarbinamine protection. Aryl coupling under Negishi conditions, followed by Boc deprotection affords compounds of type 5.3c and 5.4c.
Scheme 5.3
Figure imgf000027_0001
1. HCI
2. Pd(Pt-Bu3)2, K3PO4
Figure imgf000027_0002
, '-.a- Il ,.' 'Ul' .,.„« li.,,1' ,,,.,ll > U ItI It,,.. ,_«B ,.,,Jr
Scheme 5.4
Figure imgf000028_0001
1. HCI
2. Pd(Pt-Bu3)2, K3PO4
Figure imgf000028_0002
Described in Scheme 5.5 is the synthesis of macrocycles of type 5.5b and 5.5c, using a strategy similar to that of Scheme 5.1. The ring closure is accomplished through an intramolecular Heck reaction.
, ,.
Scheme 5.5
Figure imgf000029_0001
Described in Scheme 5.6 is the synthesis of macrocycles of type 5.6b and 5.6c, using a strategy similar to that of Scheme 5.1. The ring closure is accomplished through an intramolecular Heck reaction.
ii,.,i. a n1' ϊi Cii iUσ ^i ..'" ~u- E- C .--I' .,:»i'
Scheme 5.6
Figure imgf000030_0001
Described in Scheme 5.7 is the synthesis of macrocycles of type 5.7b and 5.7c, using a strategy similar to that of Scheme 5.1. The ring closure is accomplished through a palladium catalyzed intramolecular etherification reaction (for conditions, see: Kataoka, N.; Shelby, Q.; Stambuli, J. P.; Hartwig, J. F. J. Org. Chem. 2002, 67, 5553-5566).
„ t- . i „' u Zs U is / !Lihd id J! ,,:!
Scheme 5.7
Figure imgf000031_0001
Described in Scheme 5.8 is the synthesis of macrocycles of type 5.8b and 5.8c, using a strategy similar to that of Scheme 5.1. The ring closure is accomplished through a palladium catalyzed intramolecular etherification reaction (for conditions, see: Kataoka, N.; Shelby, Q.; Stambuli, J. P.; Hartwig, J. F. J. Org. Chem. 2002, 67, 5553-5566).
.•• - _ ■■••ir ιc,,.« «„„> ,,,,,n .
Scheme 5.8
Figure imgf000032_0001
As shown in Scheme 5.9, starting with iodides of type 3.5b, formation of the corresponding zincate with activated zinc, followed by coupling with aryl chloride 2.2f affords 5.9a. Ester reduction, transformation of the resulting alcohol to the corresponding iodide and a second Negishi coupling affords intermediate 5.9c. Silyl ether removal, ester hydrolysis and macrolactonization completes the synthesis of molecules of type 5.9d.
. ..• i r __ it
Scheme 5.9
Figure imgf000033_0001
1.1b or 1.1d
Zn*, Pd(t-Bu3P)2
Figure imgf000033_0002
5.9d 5.9c
As depicted in Scheme 5.10, 5.9c can be synthesized utilizing an alternate route. Starting with 1,5-pentadiene 3.5b, monohydroboration followed by Suzuki coupling affords 5.10a. A second Suzuki coupling with 2.2f affords 5.9c through an alternate route.
Scheme 5.10
Figure imgf000033_0003
5.9c
Described in Scheme 5.11 is the synthesis of macrocycles of type 5.11d and 5.11e. Acid l.lf is alkylated with benzyl bromide derivative 2.2e to afford adduct 5.11a. Bis allylation then gives 5.11b, which can undergo a ring closing metathesis reaction to afford 5.11c. Boc deprotection gives 5.11d. Reduction of the olefin, then Boc deprotection affords 5.11e.
Scheme 5.11
Figure imgf000034_0001
(AIIyI)SnBu3 (Ph3P)PdCI2
B
Figure imgf000034_0002
, a (! α ,.' iup >„,.[' llniU .....Il ■ ' Il I'..... II..... .....Il »
Compounds of type 5.2d or 5.2e can be selectively chorinated on the central pyridyl ring to give compounds of type 5.12a, as described in Scheme 5.12.
Scheme 5.12
Figure imgf000035_0001
The term "substantially pure" means that the isolated material is at least 90% pure, and preferably 95% pure, and even more preferably 99% pure as assayed by analytical techniques known in the art.
The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particularly preferred are the ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl- morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like. When the compound of the present invention is basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p-toluenesulfonic acid, and the like. Particularly preferred are citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, and tartaric acids.
The present invention is directed to the use of the compounds disclosed herein as inhibitors of β- secretase enzyme activity or β-site amyloid precursor protein-cleaving enzyme ("BACE") activity, in a patient or subject such as a mammal in need of such inhibition, comprising the administration of an effective amount of the compound. The terms "β-secretase enzyme," "β-site amyloid precursor protein- cleaving enzyme," and "BACE" are used interchangeably in this specification. Jh addition to humans, a variety of other mammals can be treated according to the method of the present invention.
The present invention is further directed to a method for the manufacture of a medicament or a composition for inhibiting β-secretase enzyme activity in humans and animals comprising combining a compound of the present invention with a pharmaceutical carrier or diluent.
The compounds of the present invention have utility in treating Alzheimer's disease. For example, the compounds may be useful for the prevention of dementia of the Alzheimer's type, as well as for the treatment of early stage, intermediate stage or late stage dementia of the Alzheimer's type. The compounds may also be useful in treating diseases mediated by abnormal cleavage of amyloid precursor protein (also referred to as APP), and other conditions that may be treated or prevented by inhibition of β-secretase. Such conditions include mild cognitive impairment, Trisomy 21 (Down Syndrome), cerebral amyloid angiopathy, degenerative dementia, Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type (HCHWA-D), Creutzfeld- Jakob disease, prion disorders, amyotrophic lateral sclerosis, progressive supranuclear palsy, head trauma, stroke, Down syndrome, pancreatitis, inclusion body myositis, other peripheral amyloidoses, diabetes and atherosclerosis.
The subject or patient to whom the compounds of the present invention is administered is generally a human being, male or female, in whom inhibition of β-secretase enzyme activity is desired, but may also encompass other mammals, such as dogs, cats, mice, rats, cattle, horses, sheep, rabbits, monkeys, chimpanzees or other apes or primates, for which inhibition of β-secretase enzyme activity or treatment of the above noted disorders is desired.
The compounds of the present invention may be used in combination with one or more other drugs in the treatment of diseases or conditions for which the compounds of the present invention have utility, where the combination of the drugs together are safer or more effective than either drug alone. Additionally, the compounds of the present invention may be used in combination with one or more other drugs that treat, prevent, control, ameliorate, or reduce the risk of side effects or toxicity of the compounds of the present invention. Such other drugs may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with the compounds of the present invention. Accordingly, the pharmaceutical compositions of the present invention include those that contain one or more other active ingredients, in addition to the compounds of the present invention. The combinations may be administered as part of a unit dosage form combination product, or as a kit or treatment protocol wherein one or more additional drugs are administered in separate dosage forms as part of a treatment regimen.
Examples of combinations of the compounds of the present invention with other drugs in either unit dose or kit form include combinations with anti-Alzheimer's agents, for example other beta-secretase inhibitors or gamma-secretase inhibitors; tau phosphorylation inhibitors; Ml receptor positive allosteric modulators; blockers of Aβ oligomer formation; 5-HT modulators, such as PRX-03140, GSK 742467, 1C !l
SGS-518, FK-962, SL-65.0155, SRA-333 and xaliproden; p25/CDK5 inhibitors; NK1/NK3 receptor antagonists; COX-2 inhibitors; HMG-CoA reductase inhibitors; NSAIDs including ibuprofen; vitamin E; anti-amyloid antibodies, including anti-amyloid humanized monoclonal antibodies; anti-inflammatory compounds such as (R)-flurbiprofen, nitroflurbiprofen, rosiglitazone, ND-1251, VP-025, HT-0712 and EHT-202; CB-I receptor antagonists or CB-I receptor inverse agonists; antibiotics such as doxycycline and rifampin; N-methyl-D-aspartate (NMDA) receptor antagonists, such as memantine and neramexane; cholinesterase inhibitors such as galantamine, rivastigmine, donepezil, tacrine, phenserine, ladostigil and ABT-089; growth hormone secretagogues such as ibutamoren, ibutamoren mesylate, and capromorelin; histamine H3 antagonists such as ABT-834, ABT 829 and GSK 189254; AMPA agonists or AMPA modulators, such as CX-717, LY 451395 and S-18986; PDE IV inhibitors; GABAA inverse agonists; neuronal nicotinic agonists; selective Ml agonists; microtobubule affinity regulating kinase (MARK) ligands; P-450 inhibitors, such as ritonavir; or other drugs that affect receptors or enzymes that either increase the efficacy, safety, convenience, or reduce unwanted side effects or toxicity of the compounds of the present invention. The foregoing list of combinations is illustrative only and not intended to be limiting in any way.
The term "composition" as used herein is intended to encompass a product comprising specified ingredients in predetermined amounts or proportions, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. This term in relation to pharmaceutical compositions is intended to encompass a product comprising one or more active ingredients, and an optional carrier comprising inert ingredients, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. In general, pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. Accordingly, the pharmaceutical compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier. Pharmaceutical compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for - , K ,," u Si O is -
example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
Compositions for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
Other pharmaceutical compositions include aqueous suspensions, which contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. In addition, oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. Oily suspensions may also contain various excipients. The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions, which may also contain excipients such as sweetening and flavoring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension, which may be formulated according to the known art, or may be administered in the form of suppositories for rectal administration of the drug.
The compounds of the present invention may also be administered by inhalation, by way of inhalation devices known to those skilled in the art, or by a transdermal patch. By "pharmaceutically acceptable" it is meant the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
The terms "administration of or "administering a" compound should be understood to mean providing a compound of the invention to the individual in need of treatment in a form that can be introduced into that individual's body in a therapeutically useful form and therapeutically useful amount, including, but not limited to: oral dosage forms, such as tablets, capsules, syrups, suspensions, and the like; injectable dosage forms, such as IV, IM, or IP, and the like; transdermal dosage forms, including creams, jellies, powders, or patches; buccal dosage forms; inhalation powders, sprays, suspensions, and the like; and rectal suppositories.
The terms "effective amount" or "therapeutically effective amount" means the amount of the subject compound that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician. As used herein, the term "treatment" refers to the treatment of the mentioned conditions, particularly in a patient who demonstrates symptoms of the disease or disorder.
The compositions containing compounds of the present invention may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. The term "unit dosage form" is taken to mean a single dose wherein all active and inactive ingredients are combined in a suitable system, such that the patient or person adminstering the drug to the patient can - . ..•■■ -
open a single container or package with the entire dose contained therein, and does not have to mix any components together from two or more containers or packages. Typical examples of unit dosage forms are tablets or capsules for oral administration, single dose vials for injection, or suppositories for rectal administration. This list of unit dosage forms is not intended to be limiting in any way, but merely to represent typical examples of unit dosage forms.
The compositions containing compounds of the present invention may conveniently be presented as a kit, whereby two or more components, which may be active or inactive ingredients, carriers, diluents, and the like, are provided with instructions for preparation of the actual dosage form by the patient or person adminstering the drug to the patient. Such kits may be provided with all necessary materials and ingredients contained therein, or they may contain instructions for using or making materials or components that must be obtained independently by the patient or person administering the drug to the patient.
When treating Alzheimer's disease or other diseases for which compounds of the present invention are indicated, generally satisfactory results are obtained when the compounds of the present invention are administered at a daily dosage of from about 0.1 mg to about 100 mg per kilogram of animal body weight, preferably given as a single daily dose or in divided doses two to six times a day, or in sustained release form. The total daily dosage is from about 1.0 mg to about 2000 mg, preferably from about 0.1 mg to about 20 mg per kg of body weight. In the case of a 70 kg adult human, the total daily dose will generally be from about 7 mg to about 1,400 mg. This dosage regimen may be adjusted to provide the optimal therapeutic response. The compounds may be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
Specific dosages of the compounds of the present invention, or pharmaceutically acceptable salts thereof, for administration include 1 mg, 5 mg, 10 mg, 30 mg, 80 mg, 100 mg, 150mg, 300mg and 500 mg. Pharmaceutical compositions of the present invention may be provided in a formulation comprising about 0.5 mg to 1000 mg active ingredient; more preferably comprising about 0.5 mg to 500 mg active ingredient; or 0.5 mg to 250 mg active ingredient; or 1 mg to 100 mg active ingredient. Specific pharmaceutical compositions useful for treatment may comprise about 1 mg, 5 mg, 10 mg, 30 mg, 80 mg, 100 mg, 150 mg, 300mg and 500 mg of active ingredient.
It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
The utility of the compounds in accordance with the present invention as inhibitors of β- secretase enzyme activity may be demonstrated by methodology known in the art. Enzyme inhibition is determined as follows. , =. =» r, -*
FRET Assay: A homogeneous end point fluorescence resonance energy transfer (FRET) assay is employed with the substrate ([TAMRA-S-CO-EEISEVNLDAEF-NHQSY] QFRET), which is cleaved by BACE 1 to release the fluorescence from TAMRA. The Km of the substrate is not determined due to the limit of solubility of the substrate. A typical reaction contains approximately 30 nM enzyme, 1.25 μM of the substrate, and buffer (50 mM NaOAc, pH 4.5, 0.1 mg/ml BSA, 0.2% CHAPS, 15 mM EDTA and 1 mM deferoxamine) in a total reaction volume of 100 μl. The reaction is proceeded for 30 min and the liberation of TAMRA fragment is measured in a 96-well plate LJL Analyst AD using an excitation wavelength of 530 nm and an emission wavelength of 580 nm. Under these conditions, less than 10% of substrate is processed by BACE 1. The enzyme used in these studies is soluble (transmembrane domain and cytoplasmic extension excluded) human protein produced in a baculovirus expression system. To measure the inhibitory potency of compounds, solutions of inhibitor in DMSO (four concentrations of the inhibitors are prepared: ImM, 100 μM, 10 μM, 1 μM) are included in the reactions mixture (final DMSO concentration is 0.8%). All experiments are conducted at rt using the standard reaction conditions described above. To determine the IC50 of the compound, competitive equation V0/Vi = 1 + (TJ/[IC50] is used to predict the inhibitory potency of the compounds. The errors in reproducing the dissociation constants are typically less than two-fold.
HPLC assay: A homogeneous end point HPLC assay is used with the substrate (coumarin-CO-REVNFEVEFR), which is cleaved by BACE 1 to release the N-terminal fragment attached with coumarin. The Km of the substrate is greater than 100 μM and can not be determined due to the limit of solubility of the substrate. A typical reaction contains approximately 2 nM enzyme, 1.0 μM of the substrate, and buffer (50 mM NaOAc, pH 4.5, 0.1 mg/ml BSA, 0.2% CHAPS, 15 mM EDTA and 1 mM deferoxamine) in a total reaction volume of 100 μl. The reaction is proceeded for 30 min and is stopped by the addition of 25 μL of 1 M Tris-HCl, pH 8.0. The resulting reaction mixture is loaded on the HPLC and the product is separated from substrate with 5 min linear gradient. Under these conditions, less than 10% of substrate is processed by BACE 1. The enzyme used in these studies is soluble (transmembrane domain and cytoplasmic extension excluded) human protein produced in a baculovirus expression system. To measure the inhibitory potency for compounds, solutions of inhibitor in DMSO (12 concentrations of the inhibitors are prepared and the concentration rage is dependent on the potency predicted by FRET) are included in the reaction mixture (final DMSO concentration is 10 %). All experiments are conducted at rt using the standard reaction conditions described above. To determine the IC50 of the compound, four parameters equation is used for curve fitting. The errors in reproducing the dissociation constants are typically less than two-fold.
In particular, the compounds of the following examples had activity in inhibiting the beta- secretase enzyme in the aforementioned assay, generally with an IC50 from about 1 nM to 100 μM. Such a result is indicative of the intrinsic activity of the compounds in use as inhibitors of the beta- secretase enzyme activity. H' ' «.,.» if .' 'ύ ϊ' '.',.α' Lϊf ϋ .' ~ ir ii,,,,, iw ,, i ,,,Jf
Several methods for preparing the compounds of this invention are illustrated in the Schemes and Examples herein. Starting materials are made according to procedures known in the art or as illustrated herein. The following examples are provided so that the invention might be more fully understood. These examples are illustrative only and should not be construed as limiting the invention in any way. The following abbreviations are used throughout the text:
Me: methyl
Et: ethyl
Bu: butyl t-Bu: tert-butyl Ar: aryl
Ph: phenyl
Ac: acetyl
Bn: benzyl
Py: pyridine Boc: tert-butyloxy carbonyl
TFA: trifluoroacetic acid
DCM: dichloromethane
DMA: dimthylacetamide
DMF: N,N'-dimethyl formamide TBAF: tetra-n-butylarmnonium fluoride
HMDS: hexamethyldisilazane
THF: tetrahydrofuran
DMSO: dimethylsulfoxide
EDTA: ethylene diamine tetraacetic acid TMS: trimethylsilyl
9-BBN: 9-borabicyclo[3.3. l]nonane
EDC : 1 -Ethyl-3 -(3 -dimethylaminopropyl)-carbodiimide
BSA: bovine serum albumin
CHAPS: 3-[(3-cholamidopropyl)dimethylammonio]-2-hydroxy-l-propanesulfonate rt: room temperature h: hours aq: aqueous
HPLC: high performance liquid chromatography
Intermediate I.l.a.l (Scheme 1.1) i .. i .. t r i U- IL11U Il ,.' 'Ul' ",,7r iuir ,,,,Jt ,,' "Tt- if:,,, ft.™. ..-.H ,..-.I'
Figure imgf000042_0001
To a suspension of alphamethyl m-tyrosine methyl ester hydrochloride monohydrate (10.4 g, 39.4 mmol) in THF (300 mL) was added diisopropylethyl amine (7.6 mL, 43.4 mmol) and ditertbutyldicarbonate (9.1 g, 41.4 mmol) and the reaction mixture was stirred at RT for 24 h. The reaction mixture was concentrated in vacuo to 1A volume, diluted with EtOAc and diethyl ether, washed with 10% aq KHSO4, and then alternatively with water and brine until aq pH = 7, dried over Na2SO4, concentrated in vacuo, and purified by flash chromatography (30Og silica, 0-60% EtOAc in hexanes) to provide intermediate Ll.a.1. 1H NMR (400 MHz, CDCI3) δ 7.12 (app. t, J = 8 Hz, IH), 6.72 (dd, J = 8, 2.4 Hz, IH), 6.63 (d,
J = 8 Hz, IH), 6.58 (dd, J = 2.5, 2.4 Hz, IH), 5.35 (br s, IH), 5.16 (br s, IH), 3.75 (s, 3H), 3.28 (m, IH), 3.15 (B ofAB, d, J = 13.3 Hz, IH), 1.55 (br s, 3H), 1.47 (s, 9H).
Intermediate Ll. b.l (Scheme 1.1)
Figure imgf000042_0002
To a solution of intermediate Ll.a.1 (6.62 g, 21.4 mmol) in DCM (50 mL) cooled to 0 0C was added 2,6- lutidine (2.9 mL, 24.6 mmol) and triflic anhydride (4 mL, 23.5 mmol) dropwise. The reaction mixture was stirred at 0 0C for 10 min, diluted with water, extracted with DCM twice. The combined organic fraction was dried over Na2SO4, concentrated in vacuo, and purified by flash chromatography (30Og silica, 0-30% EtOAc in hexanes) to provide intermediate I.l.b.l. 1H NMR (400 MHz, CDCI3) δ 7.35
(app. t, J = 8 Hz, IH), 7.15 (dd, J = 8, 2.4 Hz, IH), 7.11 (d, J = 8 Hz, IH), 7.0 (dd, J = 2.5, 2.4 Hz, IH), 5.19 (br s, IH), 3.77 (s, 3H), 3.52 (A of AB, br d, J = 13.6 Hz, IH), 3.27 (B of AB, d, J = 13.6 Hz, IH), 1.56 (s, 3H), 1.48 (s, 9H).
Intermediate Ll. c.l (Scheme 1.1)
Figure imgf000042_0003
To a solution of intermediate I.l.b.l (1.00 g, 2.27 mmol) in 20 mL anhydrous tetrahydrofuran cooled to O0C under an atmosphere of argon was added lithium borohydride (0.236 mL, 0.473 mmol, 2.0M solution in THF). After warming to room temperature over 2 hr., the reaction was cooled back down to O0C and quenched with MeOH. It was diluted with water (50 mL) and extracted with EtOAc (2 x 50 mL). The organic layers were combined, washed with brine (2 x 50 mL), dried over sodium sulfate, and .• . .- _
concentrated in vacuo. Purification by flash chromatography (90 g silica, 0-45% EtOAc in hexanes) gave intermediate I.l.c.1 as a white solid. IH NMR (400 MHz, CDC13) δ 7.38 (app t, J = 7.9 Hz, IH), 7.23 (d, J = 7.9 Hz, IH), 7.18 - 7.15 (m, IH), 7.12 (s, IH), 4.46 (s, IH), 4.06 (br s, IH), 3.72 (A of ABX, dd, JAB = 11.5 Hz, JAX = 3.9 Hz, IH), 3.63 (B of ABX, dd, JAB = 11.5 Hz, JBX = 8.4 Hz, 1 H), 3.30 (A of AB, d, J = 13.5 Hz, 1 H), 2.89 (B of AB, d, J = 13.5 Hz, 1 H), 1.48 (s, 9H), 1.03 (s, 3H).
Intermediate I.l.b.2 (Scheme 1.1)
Figure imgf000043_0001
Prepared from alphamethyl p-tyrosine methyl ester using a similar procedure as described for the preparation of intermediate Ll.b.l 1H NMR (400 MHz, CDCI3) δ 7.08 (s, 4H), 5.17 (br s, IH), 3.64 (s,
3H), 3.35 (A ofAB, br d, J = 13.4 Hz, IH), 3.20 (B of AB, d, J = 13.4 Hz, IH), 1.43 (s, 3H), 1.38 (s, 9H).
Intermediate I.l.b.3 (Scheme 1.1)
Figure imgf000043_0002
Prepared from m-tyrosine methyl ester using a similar procedure as described for the preparation of intermediate Ll.b.l. 1H NMR (400 MHz, CDCI3) δ 7.38 (app. t, J=8.0 Hz, IH), 7.21-7.14 (m, 2H),
7.06 (s, IH), 5.04 (d, J = 7.2 Hz, IH), 4.62-4.54 (m, IH), 4.23-4.10 (m, 2H), 3.19 (A of ABX, dd, JAB = 13.7 Hz, JAX = 5.8 Hz, IH), 3.10 (B of ABX, dd, JAB = 13.7 Hz, JBX = 5.8 Hz, IH), 1.43 (s, 9H), 1.24 (t, J= 7.1 Hz, 3H).
Intermediate I.l.c.1 (Scheme 1.1)
Figure imgf000043_0003
Step A: Alkylation
To a solution of methyl N-(diphenylmethylene)alaninate (2.6 g, 9.7 mmol) in DMF (20 mL) cooled to 0 0C was added ΝaHMDS (12.2 mL, 12.2 mmol, IM in THF) slowly via syringe and the reaction mixture was stirred at 0 0C for 15 min at which point 3-bromo-benzyl bromide (2.55 g, 10.2 mmol) in DMF (10 .
mL) was added slowly via syringe. The reaction mixture was allowed to warm to rt over 16h, quenched with aq NH4Cl and water, extracted with EtOAc, washed with aq LiCl (x3), dried over Na2SO4, concentrated in vacuo, and purified by flash chromatography (12Og silica, 0-15% EtOAc in hexanes) to provide methyl 3-bromo-N-(diphenylmethylene)-α-methylphenylalaninate.
Step B: Deprotection
To a solution of methyl 3-bromo-N-(diphenylmethylene)-α-methylphenylalaninate (2.95 g, 6.76 mmol) in MeOH (25 mL) and THF (25 mL) was added 6Ν HCl (3.4 mL, 20.3 mmol) and the reaction mixture was stirred at RT for 5 min, concentrated in vacuo and purified by ion exchange chromatography (SCX, 25 g, then 50 g, MeOH then 2M NH3 in MeOH) to provide methyl 3-bromo-α-methylphenylalaninate.
Step C: Boc Protection
To a solution of methyl 3-bromo-α-methylphenylalaninate (1.67 g, 6.1 mmol) in THF (30 mL) and MeOH (5 mL) was added ditertbutyldicarbonate (1.61 g, 7.4 mmol) and the reaction mixture was stirred at 50 0C for 6 h and at RT for 16 h, concentrated in vacuo, and purified by flash chromatography (9Og silica, 0-20% EtOAc in hexanes) to provide Intermediate I.l.c.1. 1H NMR (400 MHz, CDCI3) δ 7.36
(d, J = 7.6 Hz, IH), 7.24 (s, IH), 7.13 (t, J = 7.6 Hz, IH), 6.98 (d, J = 7.6 Hz, IH), 5.16 (br s, IH), 3.77 (s, 3H), 3.39 (A ofAB, br d, J = 13.5 Hz, IH), 3.19 (B of AB, d, J = 13.5 Hz, IH), 1.56 (br s, 3H), 1.49
(s, 9H).
Intermediate II.2.c.l(Scheme 2.2)
Figure imgf000044_0001
Step A: Sulfonamide installation
Methyl 2,6-dichloroisonicotinate (10 g, 48.5 mmol), methyl(propylsulfonyl)amine (7.99 g, 58.2 mmol), potassium phosphate (14.4 g, 68 mmol), Xantphos (1.69 g, 2.9 mmol) and tris(dibenzylideneacetone)dipalladium (0.89 g, 0.97 mmol) were added to a dry, argon flushed flask.
Dioxane (400 mL) was added, the solution degassed with argon and the reaction was heated to 100 0C for
16 hours. The reaction was cooled to rt, filtered through celite and evaporated in vacuo. Flash chromatography (silica, 0-35% EtOAc/hexanes) gave methyl 2-chloro-6- [(methylsulfonyl)(propyl)amino]isonicotinate as a yellow oil: 1H NMR (400 MHz, CD3OD) δ 7.88 (s,
IH), 7.72 (s, IH), 3.96 (s, 3H), 3.91 (t, J = 6.4 Hz, 2H), 3.13 (s, 3H), 1.68-1.53 (m, 2H), 0.93 (t, J = 7.5
Hz, 3H). .. -
Step B: Reduction
To a solution of methyl 2-chloro-6-[(methylsulfonyl)(proρyl)amino]isonicotinate (3.5 g, 11.5 mmol) in THF (50 mL) cooled to 0 0C was added LiBH4 (17.2 mL, 34.4 mmol, 2 M in THF). After 10 min, the reaction mixture was allowed to warm to rt and stirred for 3.5 h. The reaction mixture was carefully quenched with EtOAc, MeOH and water. Following dilution with EtOAc, the organic layer was extracted, washed with brine, dried over sodium sulfate and concentrated in vacuo to provide N-[6- chloro-4-(hydroxymethyl)pyridin-2-yl]-N-propylmethanesulfonamide which was used as is in the bromination step.
Step C: Bromination
To a solution of N-[6-chloro-4-(hydroxymethyl)pyridin-2-yl]-N-propylmethanesulfonamide (740 mg, 2.65 mmol) in dichloromethane (20 mL) cooled to 00C was added carbon tetrabromide (967 mg, 2.92 mmol) and triphenylphosphine (765 mg, 2.92 mmol). After 10 min, the reaction mixture was allowed to warm to rt and stirred for 0.5 h. The reaction mixture was concentrated in vacuo and purified by flash chromatography (silica, 0-25% EtOAc/hexanes) to provide N-[4-(bromomethyl)-6-chloropyridm-2-yl]-N- propylmethanesulfonamide as a white solid. 1H ΝMR (400 MHz, CDCl3) δ 7.34 (s, IH), 7.22 (s, IH), 4.35 (s, 2H), 3.85 (t, J = 7.6 Hz, 2H), 3.04 (s, 3H), 1.64-1.50 (m, 2H), 0.93 (t, J = 7.2 Hz, 3H).
Intermediate II.2.c.2(Scheme 2.2)
Figure imgf000045_0001
Synthesized using a procedure simillar to that described for Intermediate H2.C.1, with mesylmethylamine being used in the place of methyl(propylsulfonyl)amine in Step A.
Intermediate II.2.f.l (Scheme 2.2)
Figure imgf000045_0002
Step A: Sulfonamide reduction Performed as described in Step A of the synthesis of Intermediate H2.C.1, with mesyl methyl sulfonamide being used in place of propyl methyl sulfonamide.
Step B: Reduction Performed as described in Step B of the synthesis of Intermediate H2.C.1.
Step C: Silyl ether formation N-fό-chloro^-Oiydroxymethy^pyridin^-yy-N-methylmethanesulfonamide (2.8 g, 11.1 mmol) from Step B, imidazole (0.91 g, 13.4 mmol), and tert-butyldimethylsilyl chloride (1.85 g, 12.2 mmol) were dissolved in anhydrous methylene chloride (25 mL) and allowed to stir at 25 0C for 16 hours. The solution was washed with 10% potassium monohydrogen sulfate (x2), saturated sodium bicarbonate (x2), water (x2), brine (x2), dried over sodium sulfate, and concentrated in vacuo. The resulting oil was purified by flash chromatography (145 g silica, 0-30% EtOAc in hexanes). The sample was taken to the next step without further purification. LC/MS [M+H]+ = 365.1 (Cl pattern).
Intermediate III.5.b.l
Figure imgf000046_0001
Step A: Mono-esterifϊcation of diacid 3-Phenylglutaric acid (5 g, 24 mmol) and cesium carbonate (3.9 g, 12 mmol) were dissolved in 200 mL of anhydrous DMF and cooled to 0 0C. Methyl iodide (1.5 mL, 24 mmol) was added via syringe to the solution, which was allowed to slowly warm to 25 0C over 16 hours. The reaction was diluted with water, and the pH was adjusted to ~9 with sat'd NaHCO3. The mixture was then washed with hexane (x7), acidified (pH ~4) with IN HCl, extracted into ethyl acetate (x3), washed with LiCl (x3), dried over sodium sulfate and concentrated in vacuo. IH NMR (400 MHz, CDC13) δ 7.3 (m, 2H), 7.2 (m, 3H), 3.65 (m, IH), 3.60 (s, 3H), 2.7 (m, 4H).
Step B: Reduction of Acid
5-methoxy-5-oxo-3-phenylpentanoic acid (4.2 g, 19 mmol) was dissolved in 200 mL of anhydrous THF and cooled to 0 0C. Borane (57 mL, 57 mmol, 1 M soln in THF) was added via syringe to the solution, which was allowed to slowly warm to 250C over 16 hours. The reaction was then cooled to 0 0C and quenched with methanol (25 mL), followed by water (25 mL), followed by saturated solution sodium bicarbonate (25 mL). The product was extracted into ethyl acetate, washed with brine, dried over sodium .
sulfate and concentrated in vacuo. The resulting oil and purified by flash chromatography (300g silica, 10-65% EtOAc in hexanes). The sample was taken to the next step without further purification.
Step C: Iodination of Alcohol Triphenylphosphine (1.5 g, 5.8 mmol) and imidazole (0.39 g, 5.8 mmol) were added to a dried flask under argon atmoshphere. Anhydrous methylene chloride (50 mL) was added and the solution was cooled to 00C. Iodine (1.45 g, 5.8 mmol) was added in one portion, and the resulting solution was stirred at 0 0C for 0.5 h. Methyl 5-hydroxy-3-phenylρentanoate (1.0 g, 4.8 mmol) in anhydrous methylene chloride (10 mL) was added via syringe to the solution and allowed to stir at 25 0C for 16 h. The solution was diluted with methylene chloride (100 mL), washed with 10% Na2SO3 (x2), brine (x2), dried over sodium sulfate, concentrated in vacuo. The resulting oil was purified by flash chromatography (175 g silica, 0-25% EtOAc in hexanes). The sample was taken to the next step without further purification. IH NMR (400 MHz, CDCI3) δ 7.3 (m, 2H), 7.2 (m, 3H), 3.7 (s, 3H), 3.30 (m, IH), 3.15 (m, IH), 2.85
(m, IH), 2.65 (d, J = 7.5 Hz, 2H), 2.25 (m, IH), 2.15 (m, IH).
Intermediate IV.l.b.l
Figure imgf000047_0001
Step A: Suzuki Coupling To 0.70Og (2.25 mmol, 1 equiv.) of (but-3-en-l-yloxy)(tert-buryl)diphenylsilane was added 5.9 mL
(0.358 mmol, 1.3 equiv) of a 0.5M soln of 9-BBN in THF. The solution was heated to 70° C for 1.25h, then the reaction was cooled to rt, and transferred to a solution of Intermediate I.l.b.l (0.995g, 2.25 mmol, 1 equiv.) and Pd(Ph3P)4 (0.131g, 0.113 mmol, 0.05 equiv.) in 1.05 mL 3.2M NaOH (3.38 mmol, 1.5 equiv.) and 3 mL toluene. The reaction was degassed with argon for 5 min, then capped and heated at 85° C for 15h. The reaction was cooled to rt and filtered through a pad of celite, rinsing with EtOAc. Partitioned filtrate between EtOAc and brine, separated and washed organics with brine, dried over Na2SO4, filtered and concentrated. Purified residue using silica gel chromatography to isolate desired product as a viscous oil. 1H NMR (400 MHz, CDCl3) δ 7.63 (m, 4H), 7.42-7.32 (m, 6H), 7.14 (dd, J = 7.6, 4.2 Hz, IH), 7.00 (d, J = 7.6 Hz, IH), 6.87-6.84 (m, 2H), 3.70 (s, 3H), 3.64 (t, J = 6.2 Hz, 2H), 3.28 (br peak, IH), 3.13 (d, J = 13.4 Hz, IH), 2.55 (m, IH), 2.39 (m, IH), 1.89-1.76 (m, 2H), 1.69-1.54 (m, 2H), 1.53 (s, 3H), 1.39 (s, 9H), 1.02 (s, 9H). LC/MS [M+H]+ = 604.
Step B: Silyl Deprotection is .•• " » »-,: B !? n- ...
"irE,.; i!-«; .-_ii . ~B
To solution of product from Step A (0.342g, 0.566 mmol, 1 equiv) in 4 mL THF was added 0.736 mL (0.736 mmol, 1.3 equiv) of a IM solution of tetra-n-butyl ammonium fluoride in THF. After 5h, the reaction was quenched by the addition of satd. aqueous NaHCO3 and diluted with EtOAc. The layers were separated, the aqueous was washed with fresh EtOAc, the combined organics were washed with brine, dried over Na2SO4, filtered and concentrated. Purified residue using silica gel chromatography to isolate desired product as a viscous oil. 1H NMR (400 MHz, CDCl3) δ 7.14 (t, J = 7.6 Hz, IH), 7.00 (d, J = 7.6 Hz, IH), 6.87-6.84 (m, 2H), 3.70 (s, 3H), 3.60 (t, J = 6.4 Hz, 2H), 3.30 (br peak, IH), 3.13 (d, J =
13.3 Hz, IH), 2.58 (t, J - 7.5 Hz, 2H), 1.69-1.45 (m, 7H), 1.45 (s, 9H). LC/MS [M+H]+ = 366.
Step C: Bromination
To a solution of product from Step B (0.09Og, 0.246 mmol, 1.0 equiv), triphenylphosphine (0.129g, 0.493 mmol, 2 equiv.) and imidazole (0.034g, 0.493 mmol, 2 equiv) in 2.5 mL CH2CI2 at 0° C was added carbontetrabromide (0.163g, 0.493 mmol, 2 equiv.). After 2.5h, the bath was removed and after a further 2h at rt, the reaction was poured onto a silica gel column for purification using normal phase chromatography. 1H NMR (400 MHz, CDCl3) δ 7.16 (t, J = 7.6 Hz, IH), 7.02 (d, J = 7.6 Hz, IH), 6.89- 6.87 (m, 2H), 3.73 (s, 3H), 3.38 (t, J = 6.5 Hz, 2H), 3.31 (br peak, IH), 3.14 (d, J = 13.5 Hz, IH), 2.57 (t, J = 7.6 Hz, 2H), 1.86-1.82 (m, 2H), 1.76-1.69 (m, 2H), 1.54 (s, 3H), 1.45 (s, 9H). LC/MS [M+H]+ = 428 (Br pattern).
Step D: Azide Displacement
To a solution of product from Step C (0.092g, 0.215 mmol, 1 equiv.) in 1.5 mL DMF was added sodium azide (0.028g, 0.430 mmol, 2 equiv). After 15h, the reaction was diluted with 3M LiCl and EtOAc. The layers were separated, and the organics were washed with 3M LiCl and brine, dried over Na2SO4, filtered and concentrated. Purification by normal phase chromatography afforded the desired azide (Intermediate IV.l.b.l) as a clear oil. 1H NMR (400 MHz, CDCl3) δ 7.17 (t, J = 7.5 Hz, IH), 7.03 (d, J = 7.6 Hz, IH), 6.89-6.87 (m, 2H), 3.74 (s, 3H), 3.32 (br peak, IH), 3.25 (t, J = 6.6 Hz, 2H), 3.15 (d, J =
13.4 Hz, IH), 2.58 (t, J = 7.2 Hz, 2H), 1.71-1.57 (m, 4H), 1.54 (s, 3H), 1.46 (s, 9H). LC/MS [(M- Boc)+H]+ = 291.
Intermediate IV.l.d.l
Figure imgf000048_0001
To a solution of Intermediate IV.1.c.1 (0.044g, 0.113 mmol, 1 equiv) in 1.3 mL THF was added 2M LiBH4 in THF (0.225 mL, 0.451 mmol, 4 equiv). After 16h, the reaction was cooled to 0° C and quenched by the addition of satd. aqueous NaHCO3 and diluted with EtOAc. The layers were separated, i . . i _
U KmI. i! ,.' >l,,,l' .,,,Jl l|,,,ll (I ,,' Il II,,,,, IU. .,-.!• .....I'
the aqueous was washed with EtOAc (2x) the combined organics were washed with brine, dried over Na2SO4, filtered and concentrated. Purification by normal phase chromatography afforded the desired alcohol (Intermediate IV.l.d.1) as a clear oil. 1H NMR (400 MHz, CDCl3) δ 7.20 (t, J = 7.5 Hz, IH), 7.04 (d, J = 7.5 Hz, IH), (6.98 (d, J = 7.5 Hz, IH), 6.97 (s, IH), 4.05 (s, IH), 4.17 (br s, IH), 3.69-3.62 (m, 2H), 3.26 (t, J = 6.8 Hz, 2H), 3.15 (d, J = 13.3 Hz, IH), 2.75 (d, J = 13.4 Hz, IH), 2.61 (t, J = 7.3 Hz, 2H), 1.71-1.45 (m, 4H), 1.45 (s, 9H), 1.05 (s, 3H). LC/MS [(M-Boc)+H]+ = 263.
Intermediate IV.l.e.l
Figure imgf000049_0001
To a solution of Intermediate IV.l.e.l (0.07Og, 0.179 mmol, 1 equiv) in 2 mL THF was added IM LiOH in THF (0.538 mL, 0.538 mmol, 3 equiv). After 3h, 0.5 mL MeOH was added, and after 15h, the reaction was heated to 45° C for 4.5h. The reaction was cooled to rt and acidified to pH 3 using 10% KHSO4. The aqueous was washed with EtOAc (4x) the combined organics were dried over Na2SO4, filtered and concentrated. The residue was used without further purification. 1H NMR (400 MHz, CDCl3) δ 9.92 (br s, IH), 7.18 (t, J = 7.5 Hz, IH), 7.05 (d, J = 7.5 Hz, IH), 6.97-6.94 (m, 2H), 5.05 (br s, IH), 3.26 (t, J = 6.7 Hz, 4H), 2.59 (t, J = 7.3 Hz, 2H), 1.72-1.58 (m, 4H), 1.55 (s, 3H), 1.47 (s, 9H). LC/MS [(M-Boc)+H]+
= 277.
Intermediate 5.2.a.l
Figure imgf000049_0002
Step A: Coupling
To a solution of Intermediate IV.l.e.l (0.077g, 0.205 mmol, 1 equiv) and Intermediate II.2.C.1 (0.07Og, 0.205 mmol, 1 equiv.) in 1.5 mL DMF was added CsCO3 (0.08Og, 0.245 mmol, 1.2 equiv.). After 1.5h, the reaction was partitioned between H2O and EtOAc, the layers were separated, the organics were Il If,,, •UP a> !(...» ,-.'< ✓ h-ii ιi,,ϋ rid ,,rJ ,„:!
washed with 3M LiCl (2x) and brine, dried over Na2SO4, filtered and concentrated. The residue was purified by normal phase silica gel chromatography to afford the desired product as a clear oil. 1H NMR (400 MHz, CDCl3) δ 7.26 (s, IH), 7.17 (d, J = 7.5 Hz, IH), 7.12 (s, IH), 7.06 (d, J = 7.5 Hz, IH), 6.90- 6.89 (m, 2H), 5.17 (d, J = 14.3 Hz, IH), 5.09 (d, J = 14.3 Hz, IH), 4.90 (br s, IH), 3.82 (t, J = 7.2 Hz, 2H), 3.32-3.24 (m, 3H), 3.16 (d, J = 13.6 Hz, IH), 3.01 (s, 3H), 2.58 (t, J = 7.1 Hz, 2H),1.69-1.54 (m, 6H), 1.49 (s, 3H), 1.44 (s, 9H), 0.90 (t, J = 7.5 Hz, 3H). LC/MS [(M-ffiu)+H]+ = 581.
Step B: Staudinger Reduction
To a solution of product from Step A (0.040 g, 0.063 mmol) in 1 mL THF and 0.1 mL water was added triphenylphosphine (0.026 g, 0.10 mmol). The reaction was heated at 65° C for 1.5h, cooled to rt, then pipetted onto a Ig SCX ion exchange column conditioned with MeOH. The column was eluted with 50 mL MeOH, them 50 mL 2M NH3 in MeOH. The fractions contaning the desired product were collated and concentrated. LC/MS [M+H]+ = 611 (Cl pattern).
Intermediate 5.2.a.2
Figure imgf000050_0001
Synthesized using a reaction sequence similar to that described for the synthesis of Intermediate V.2.a.2, with Intermediate U.2.C.2 being used in place of Intermediate HcI in step A.
Intermediate V.ll.b.l
Figure imgf000050_0002
- , -, ~ f ,r_
Step A: Hydrolysis
Performed on Intermediate I.l.c.l as described for the synthesis of Intermediate IV.l.e.l. LC/MS [M+H]
= 358, 360 (Br pattern)
Step B: Coupling
Product from Step A and Intermediate 2.2.e.2 were coupled as described in Step A of Intermediate V.2.a.l synthesis. LC/MS [M+H] = 590, 592 (Br pattern)
Step C: Allylation To a solution of {2-chloro-6-[methyl(methylsulfonyl)amino]pyridine-4-yl}methyl 3-bτomo-N-(tert- butoxycarbonyl)-α-methylphenylalaninate (400 mg, 0.68 mmol) and allyltributylstannane (0.48 mL, 1.6 mmol) in degassed DMF (4 mL) was added bis(triphenylphosphine)palladium(π) dichloride (33 mg, 0.048 mmol). The reaction was heated to 1200C in a microwave for 0.5 h. Additional catalyst was added and heating was repeated 3x to drive the reaction to completion. Potassium fluoride (134 mg, 2.3 mmol) was added, the reaction was stirred for 3h and diluted with EtOAc. Filtration through celite, concentration and flash chromatography (silica gel, 0-50% EtOAc/hexanes) gave {2-allyl-6- [methyl(methylsulfonyl)amino]pyridine-4-yl}methyl 3-allyl-N-(te?t-butoxycarbonyl)-α- methylphenylalaninate. MS: 558.1 (M+l) ES+
Step D: Ring Closing Metathesis Reaction
To a solution of {2-allyl-6-[methyl(methylsulfonyl)amino]pyridine-4-yl}methyl 3-allyl-N-(tert- butoxycarbonyl)-α-methylphenylalaninate (80 mg, 0.14 mmol) in degassed DCE (2.9 mL) was added Zhan (metathesis) Catalyst I (9.2 mg, 0.014 mmol). The reaction was heated to 12O0C in a microwave for 0.5 h. Concentration and flash chromatography (silica gel, 0-50% EtOAc/hexanes) gave tert-butyl [(3^Z)-14-methyl-8-[methyl(methylsulfonyl)amino]-13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]hemcosa- 1(20), 3, 6(21), 7, 9, 16, 18-heptaen-14-yl]carbamate. MS: 530.4 (M+l) ES+
EXAMPLE l
N-[13-amino-13-methyl-15-oxa-2,20-diazatricyclo[15.3.1.17,l l]docosa-l(21),7(22),8,10,17,19-hexaen- 19-yl] -N-propylmethanesulfonamide ■ _ _..
Step A: Coupling
To a solution of Intermediate IV.l.d.l (0.039g, 0.109 mmol, 1.2 equiv) and Intermediate H2.C.1 (0.031g, 0.091 mmol, 1 equiv.) in 1.5 mL 1,2-dichloroethane was added a spatula tip of activated 4A molecular sieves, polymer bound 2,6-di-tBuPy (0.052g, 0.272 mmol, 3 equiv.) and AgOTf (0.068g, 0.263 mmol, 2.9 equiv.). The solution was microwaved at 90° C for 8.5h, then filtered through a pad of celite, rinsing with EtOAc. The rinsate was concentrated, and the residue was purified by normal phase silica gel chromatography. 1H NMR (400 MHz, CDCl3) δ 7.20-7.16 (m, 2H), 7.03 (d, J = 7.5 Hz, IH), 6.8-6.96 (m, 3H), 4.53 (s, 2H), 4.47 (s, IH), 3.81 (t, J = 7.3 Hz, 2H), 3.65 (d, J = 9.0 Hz, IH), 3.52 (d, J = 9.0 Hz, IH), 3.25 (t, J = 6.7 Hz, 2H), 3.11 (d, J = 13.2 Hz, IH), 3.00 (s, 3H), 2.85 (d, J = 13.2 Hz, IH), 2.59 (t, J = 7.1 Hz, 2H), 1.70-1.52 (m, 6H), 1.45 (s, 9H), 1.23 (s, 3H), 0.90 (t, J = 7.3 Hz, 3H). LC/MS [M+H]+ = 623.
Step B: Staudinger Reduction
To a solution of product from Step A (0.020 g, 0.032 mmol, 1 equiv) in 0.70 mL THF and 0.070 mL water was added triphenylphosphine (0.009 g, 0.035 mmol, 1.1 equiv.). The reaction was heated at 65° C for 15h, cooled to rt, then pipetted onto a Ig SCX ion exchange column conditioned with MeOH. The column was eluted with 50 mL MeOH, them 50 mL 2M NH3 in MeOH. The fractions contaning the desired product were collated and concentrated. 1H NMR (400 MHz, CDCl3) δ 7.19-7.16 (m, 2H), 7.04 (d, J = 7.5 Hz, IH), 6.96-6.95 (m, 3H), 4.54 (s, 2H), 4.52 (s, IH), 3.81 (t, J = 7.1 Hz, 2H), 3.65 (d, J = 9.0 Hz, IH), 3.52 (d, J = 9.0 Hz, IH), 3.11 (d, J = 13.2 Hz, IH), 3.01 (s, 3H), 2.84 (d, J = 13.2 Hz, IH), 2.68 (t, J = 7.0 Hz, 2H), 2.58 (t, J = 7.7 Hz, 2H), 1.66-1.48 (m, 6H), 1.45 (s, 9H), 1.23 (s, 3H), 0.91 (t, J = 7.4 Hz, 3H). LC/MS [M+H]+ = 597.
Step C: Macroamination To a solution of product from Step B (0.015 g, 0.025 mmol, 1 equiv) in 0.60 mL DMA was added
K3PO4 (0.016 g, 0.075 mmol, 3 equiv.) and Pd(t-Bu3)2 (0.004g, 0.008 mmol, 0.3 equiv). The reaction was degassed with Ar, then heated at 100° C for 19h, cooled to rt and diluted with H2O and EtOAc. The layers were separated, and the organics were washed with 3M LiCl (2x) and brine, dried over Na2SO4, filtered and concentrated. The residue was purified using silica gel chromatography to obtain the desired i a-" 111Ii"' .' ti it it:-' rii if- ■' !( Ii r_3> "1 ;
product was a clear glass. 1H NMR (400 MHz, CDCl3) δ 7.10 (t, J = 8.0 Hz, IH) 6.95-6.94 (m, 3H), 6.39 (s, IH), 6.05 (s, IH), 4.84 (s, IH), 4.73 (t, J = 6.4 Hz, IH), 4.50 (d, J = 13.5 Hz, IH), 4.31 (d, J = 13.5 Hz, IH), 3.65 (t, J = 6.3 Hz, 2H), 3.35-3.19 (m, 4H), 3.02 (d, J = 9.3 Hz, IH), 2.97 (s, 3H), 2.67 (d, J = 9.3 Hz, IH), 2.61 (m, IH), 1.67 (m, 2H), 1.56-1.48 (m, 6H), 1.48 (s, 9H), 1.24 (s, 3H), 0.86 (t, J = 7.5 Hz, 3H). LC/MS [MHhH]+ = 561.
Step D: Boc deprotection
To a solution of product from Step C (0.004 g, 0.007 mmol, 1 equiv) in 1.0 mL CH2CI2 was added 4M
HCl in dioxane (0.027 mL, 0.107 mmol, 15 equiv.). After 15h at rt, the reaction was concentrated to afford the hydrochloride salt of N-flS-amino-lS-methyl-lS-oxa^^O-diazatricycloflS.S.l.πjllJdocosa- l(21),7(22),8,10,17,19-hexaen-19-yl]-N-propyl methanesulfonamide as a white solid. 1H ΝMR (400 MHz, ^-MeOH) δ 7.21 (t, J = 7.5 Hz, IH), 7.10 (d, J = 7.5 Hz, IH), 7.01 (d, J = 7.5 Hz, IH), 6.97 (s, IH), 6.77 (s, IH), 6.76 (s, IH), 4.75 (d, J = 13.9 Hz, IH), 4.52 (d, J = 13.9 Hz, IH), 3.72-3.54 (m, 4H), 3.45-3.33 (m, 2H), 3.12 (d, J = 10.4 Hz, IH), 2.79 (d, J = 10.3 Hz, IH), 2.70-2.56 (m, 2H), 1.73-1.1.44 (m, 6H), 1.27 (s, 3H), 0.88 (t, J = 7.4 Hz, 3H). LC/MS [M+H]+ = 461. Exact Mass calc for C24H36N4O3S: 461.2581; measured: 461.2567.
EXAMPLE 2
N-[13-amino-13-methyl-14-oxo-15-oxa-2,20-diazatricyclo[15.3.1.17,l l]docosa-l(21),7(22),8,10,17,19- hexaen-19-yl]-N-propylmethanesulfonamide
Figure imgf000053_0001
Step A: Macroamination
Performed on Intermediate 5.2.a.l described in Step C of the.Example 1 synthesis, with the following modifications: 2 equiv. OfK3PO4 were used, and the reaction was heated at 90° C for 2h.
Step B: Boc deprotection
Performed as described in Step D of the Example 1 synthesis. 1H NMR (400 MHz, ^-MeOH) δ 7.16 (t, J = 7.5 Hz, IH), 7.05 (d, J = 7.5 Hz, IH), 6.90 (d, J = 7.5 Hz, IH), 6.59 (s, IH), 6.29 (br s, IH), 6.23 (s, IH), 5.20 (d, J = 12.3 Hz, IH), 5.01 (d, J = 12.3 Hz, IH), 3.69 (m, 2H), 3.40 (m, 2H), 3.16 (d, J = 13.9 Ir1 >[„.» ii 'LI .','3 Li L"JΓΪF ..' -"IrSn;! id ,, i „;;
Hz, IH), 3.04-2.97 (m, 3H), 2.49 (m, 2H), 1.64 (s, 3H), 1.61-1.43 (m, 6H), 0.83 (t, J = 7.3 Hz, 3H). LC/MS [M+H]+ = 475. Exact Mass calc for C24H34N4O4S: 475.2374; measured: 475.2384.
EXAMPLE 3
N-[13-amino-13-methyl-14-oxo-15-oxa-2,20-diazatricyclo[15.3.1.17,l l]docosa-l(21),7(22),8,10,17,19- hexaen- 19-yl] -N-methylmethanesulfonamide
Figure imgf000054_0001
Prepared from Intermediate 5.2.a.2 utilizing a reaction sequence as described for the synthesis of Example 2.
EXAMPLE 4
N-[13-amino-2-benzyl-13-methyl-14-oxo-15-oxa-2,20-diazatricyclo[15.3.1.17>n]docosa- 1 (21),7(22), 8, 10, 17, 19-hexaen-l 9-yl]-N-methylmethanesulfonamide
Figure imgf000054_0002
Step A: Reductive Amination
To a solution of Intermediate 5.2. a.2 (0.030 g, 0.051 mmol) in 0.4 mL 1,2-dichloroethane was added benzaldehyde (0.006 mL, 0.057 mmol), sodium triacetoxyborohydride (0.014 g, 0.067 mmol) and acetic acid (1 drop). The reaction was allowed to proceed overnight, then contrated and purified by reverse- phase Gilson. LC/MS [M+H]+ = 673 (Cl pattern). ■ f . • _
1I,, ~ ,S !6, j - - t Jf ,,»,l' J
Step B and Step C: Macroamination and Boc deprotection
Performed as described for the synthesis of Example 3. LC/MS [M+H]+ for title compound = 537.
The following compounds were synthesized using a procedure as described for the synthesis of Example 4.
Red. Amin Partner Example Structure LC/MS (M+H)+
Acetone 488
Figure imgf000055_0001
Propanal 488
Figure imgf000055_0002
Cyclopropyl carboxaldehyde 500
Figure imgf000055_0003
EXAMPLE 8
S-methyl-lP-fmethy^methylsulfony^aminoJ^-oxo-M-phenyl-S-oxa-lS-azatricyclo [15.3.1. l~7,ll~]docosa-l(21),7(22),8,10,17,19-hexaen-5-amine
Figure imgf000056_0001
Step A: Zincate coupling of intermediate A and B
Intermediate ]H.5.b.l (1.0 g, 3.14 mmol) and activate zinc (0.225 g, 3.45 mmol) was dissolved in degassed THF (8 mL) and allowed to stir under argon at 25 0C for 3 h. The iodide/activated zinc mixture was added via syringe to a dried flask containing intermediate ϋ.2.f.l (1.15 g, 3.14 mmol) and [Xt-Bu)3P]2Pd (0.16 g, 0.314 mmol) in degassed THF (15 mL) under argon atmosphere. The resulting solution was stirred at 75 0C for 16 h. The solution was then filtered over a pad of celite, washing with ethyl acetate, and concentrated in vacuo. The resulting oil was purified by flash chromatography (175 g silica, 0-25% EtOAc in hexanes). The sample was taken to the next step without further purification. LC/MS [M+H]+ = 520.1.
Step B: Reduction of Ester methyl 5-{4-({[tert-butyl(dimethyl)silyl]oxy}methyl)-6-{methyl(methylsulfonyl)amino]pyridin-2-yl}-3- phenylpentanoate (0.55 g, 1.05 mmol) from step A was dissolved in anhydrous THF (10 mL) and cooled to 0 0C under argon atmosphere. Lithium borohydride (0.74 mL, 1.49 mmol, 2.0 M solution in THF) was slowly added via syringe. After addition was complete, the temperature was raised to 45 0C and the reaction was allowed to stir at that temperature for 16 h. The solution was then cooled to 0 0C and quenched with methanol (5 mL) followed by water (5 mL). The product was extracted into ethyl acetate, washed with brine, dried over sodium sulfate and concentrated in vacuo. The sample was taken to the next step without further purification. LC/MS [M+H]+ = 493.0
Step C: Iodination of alcohol
Prepared in a similar fashion as the preparation of Intermediate ffl.5.b.l, Step C.
Step D: Zincate coupling of intermediate I.l.c.l and scaffold r Ii ■■ >u? w1 <u8 S ,. -Ii ET«* C n »™i"
^^-({[tert-buty^dimethyOsilyyoxylmethy^-ό-CS-iodo-S-phenylpenty^pyridin^-ylj-N- methylmethanesulfonamide (0.395 g, 0.66 mmol) from Step C and activated zinc (0.086 g, 1.3 mmol) was dissolved in degassed THF (2 mL) and allowed to stir under argon at 25 0C for 3 h. The iodide/activated zinc mixture was added via syringe to a dried flask containing intermediate I.l.c.l (0.244 g, 0.66 mmol) and [(t-Bu)3P]2Pd (0.033 g, 0.066 mmol) in degassed THF (5 mL) under argon atmosphere. The resulting solution was stirred at 75 0C for 16 h. The solution was then filtered over a pad of celite, washing with ethyl acetate, and concentrated in vacuo. The resulting oil was purified by flash chromatography (90 g silica, 0-20% EtOAc in hexanes). The sample was taken to the next step without further purification. LC/MS [M+H]+ = 768.2
Step E: Silyl Deprotection
To solution of product from Step D (0.118g, 0.154 mmol) in 2 mL THF was added 0.169 mL (0.169 mmol) of a IM solution of tetra-n-butyl ammonium fluoride in THF. After 5h, the reaction was quenched by the addition of satd. aqueous NaHCO3 and diluted with EtOAc. The layers were separated, the aqueous was washed with fresh EtOAc, the combined organics were washed with brine, dried over Na2SO4, filtered and concentrated. Purified residue using silica gel chromatography to isolate desired product as a viscous oil. LC/MS [M+H]+ = 654.1
Step F: Hydrolysis of Ester To solution of product from Step E (0.038g, 0.058 mmol) in 2 mL THF was added 0.145 mL (0.291 mmol) of a IM solution of LiOH in water. The solution was allowed to stir overnight at 45 0C for 16 h.
The crude reaction mixture was acidified to pH = 4 using IM HCl, extracted into ethyl acetate, washed with 3 M solution of LiCl and dried over sodium sulfate. The solvent was removed in vacuo and taken to the next step without further purification. LC/MS [M+H]+ = 640.2
Step G: Mitsonobu Macrolactionization
To a solution of the product from Step F (0.035 g, 0.055 mmol) and triphenylphosine (0.022 g, 0.082 mmol) in 6 mL of anhydrous THF under argon atmoshphere was added DIAD (0.016 mL, 0.082 mmol) via syringe. The solution was allowed to stir at 25 0C for 3 h. The solution was then concentrated and purified residue using reverse phase (C 18) chromatography to isolate desired product as a viscous oil.
LC/MS [M+H]+ = 622.0.
Step H: Boc deprotection
To a solution of product from Step H (0.028 g, 0.045 mmol) in 1.0 mL CH2CI2 was added 4M HCl in dioxane (0.027 mL, 0.107 mmol). After 15h at rt, the reaction was concentrated to afford the hydrochloride salt of 5-methyl-19-[methyl(methylsulfonyl)amino]-4-oxo-14-phenyl-3-oxa-18- i «„„. Ii „' '[,,,r .„„11 rj, ji „„,{! „ ιr ii,,,,,. if,,,,. ,,.,.n ,,,,.u
azatricyclo[15.3.1.1-7,1 l~]docosa-l(21),7(22),8,10,17,19-hexaen-5-aminium chloride as a white solid. LC/MS [M+H]+ = 522.0. Exact Mass calc for C29H35N3O4S: 522.2421; measured: 522.2418.
EXAMPLE 9
5 -methyl- 19-[methyl(methylsulfonyl)amino]-3 ,4-dioxa- 18-azatricyclo [ 15.3.1.1-7, 11 ~] docosa- l(21),7(22),8,10,17,19-hexaen-5-amine
Figure imgf000058_0001
Step A: Suzuki Coupling
To 5.28 mL (50.4 mmol) of 1, 4-pentadiene was added 14.8 mL (7.4 mmol) of a 0.5 M solution of 9-BBN in THF. The solution was heated to 70 0C for 1.25h, then the reaction was cooled to rt, and transferred to a solution of Intermediate I.l.c.1 (2.5 g, 6.7 mmol), Pd(Ph3P)4 (0.77 g, 0.67 mmol) in 2.2 mL 3.2M NaOH (7.05 mmol) and 10 mL of degassed toluene. The reaction was then degassed with argon for 5 min, then capped and heated at 85 0C for 15h. The reaction was cooled to rt and filtered through a pad of celite, rinsing with EtOAc. Partitioned filtrate between EtOAc and brine, separated and washed organics with brine, dried over Na2SO4, filtered and concentrated. Purified residue using silica gel chromatography to isolate desired product as a viscous oil. LC/MS [M+H]+ = 362.1.
Step B: Suzuki with product from Step A and intermediate 2.2f Prepared as described in Step A.
Step C-F: Silyl Deprotection, Ester hydrolysis, Mitsunobu macrolactonization and Boc deprotection sequence performed as described in the synthesis of Example 9. LC/MS [M+H] = 446
EXAMPLE 10 5-methyl-19-[methyl(methylsulfonyl)amino]-4-oxo-14-methyl-3-oxa-l 8-azatricyclo [15.3.1. l-7,l l~]docosa-l(21),7(22),8,10,17,19-hexaen-5-amine Il 'Ui. U ,.' >!„!' WiT .„(1 ,,,j! . ll-|u! EU "Jf .
Figure imgf000059_0001
Synthesized using a reaction sequence similar to that described for Example 9, with 3 -methyl- 1,4- pentadiene being substituted for 1,4-pentadiene in Step A. LC/MS [M+H] = 460.
EXAMPLE I l
N-[(15Jδ)-5-amino-5,14-dimethyl-4-oxo-3-oxa-18-azatricyclo[15.3.1.17'n]docosa- 1 (21),7(22),8, 10, 15, 17, 19-heptaen-19-yl]-N-methylmethanesulfonamide
Figure imgf000059_0002
Step A: Suzuki with 3 -Methyl- 1,4-pentadiene and 1.1.c.1 Prepared as Step A in the synthesis of Example 9.
Step B: Hydrolysis of Ester
Prepared as Step F in synthesis of Example 9.
Step C: Alkylation
To a solution of the product of Step B (0.515 g, 1.425 mmol) and H.2.C.2 (0.491 g, 1.55 mmol) in DMF (7 mL) was added cesium carbonate (0.302 g, 0.926 mmol). The reaction was allowed to stir at rt for 16 h. The crude mixture was extracted with EtOAc (x3), washed with DI water (x3), sat'd LiCl (x3), dried over sodium sulfate, and concentrated in vacuo. The crude material was purified using flash chromatography (9Og silica, 0-25% EtOAc in hexanes) to afford the corresponding lactone. if » „■
Step D: Intramolecular Heck reaction
To a solution of product from Step C (0.10 g, 0.17 mmol) and potassium phosphate (0.107 g, 0.51 mmol) in 8 mL of degassed anhydrous DMA under argon atmosphere was added [(t-Bu)3P]2Pd (0.009 g, 0.017 mmol). The whole system was then degassed with argon. The temperature was raised to 115 0C and allowed to stir at that temperature for 16 h. The crude reaction was filtered over celite, washing with ethyl acetate. The organic layers were washed with water (x3), brine (x3), dried over sodium sulfate, solvent was removed in vacuo. The resulting oil was purified by flash chromatography (40 g silica, 0- 25% EtOAc in hexanes). The sample was taken to the next step without further purification. LC/MS [M+H]+ = 558.1
Step E: Boc Deprotection
Prepared as Step H in synthesis of Example 8. LC/MS [M+H]+ = 458.1
EXAMPLE 12
N-[14-amino-4,14-dimethyl-5-methylene-13-oxo-12-oxa-7-azatricyclo[14.3.1.16'10]henicosa- 1 (20),6(21),7,9, 16, 18-hexaen-8-yl]-N-methylmethanesulfonamide
Figure imgf000060_0001
Isolated as a secondary product in Step D of the Example 11 synthesis elaborated through step E to afford title compound. LC/MS [MH-H]+ = 458.1.
EXAMPLE 13
N-[(3JB)-14-Amino-14-methyl-13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16, 18-heptaen-8-yl]-N-methylmethanesulfonamide and
N-[(3Z)-14-Amino-14-methyl-13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16, 18-heptaen-8-yl]-N-methylmethanesulfonamide [[,.,.. α ,•' 'U' is If,, it ,,.,,!' .' -!i UTJ. iu ,,;,.u „,„[:
Figure imgf000061_0001
A solution of tert-butyl [(S^^-M-methyl-S-fmethy^methylsulfony^aminoJ-lS-oxo-^-oxa-?- azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16, 18-heptaen-14-yl] carbamate (14 mg, 0.026 mmol) (Intermediate V.I I.e.1) in DCM (0.5 mL) and TFA (1.5 mL) was stirred at rt for 1 h.
Concentration, purification and separation of EZ isomers by reverse phase preparative HPLC (5-95%
CH3CN in water containing 0.1% TFA) gave N-[(3£)-14-Amino-14-methyl-13-oxo-12-oxa-7- azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16, 18-heptaen-8-yl]-N-methylmethanesulfonamide and
N-[(3Z)-14-Amino-14-methyl-13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16,
18-heptaen-8-yl]-N-methylmethanesulfonamide as the TFA salts. EXaCt MaSS CaIC fOr (^ Or Z) C22H27N3O4S: 430.1795; measured: 430.1798. Exact
Mass calc for (Z or ^) C22H27N3O4S: 430.1795; measured: 430.1801.
EXAMPLE 14
N-[14-Amino-14-methyl-13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]henicosa-l(20), 6(21), 7, 9, 16, 18- hexaen~8-yl]-N-methylmethanesulfonamide
Figure imgf000061_0002
Step A: Olefin reduction
To a solution of tert-butyl [(3£Z)-14-methyl-8-[methyl(methylsulfonyl)amino]-13-oxo-12-oxa-7- azatricyclo[14.3.1.16' 10]henicosa-l(20), 3, 6(21), 7, 9, 16, 18-heptaen-14-yl]carbamate (22.5 mg, 0.042 mmol) (Intermediate V.I l.c.l) in degassed THF (2.5 mL) was added palladium on carbon (10%, 3 mg). .. i .►■ 1 N „ r E^.. «3 .ri
The reaction was stirred under a hydrogen atmosphere (1 atm) for 1 h and filtered through celite. Concentration gave fert-butyl [14-methyl-8-[methyl(methylsulfonyl)amino]-13-oxo-12-oxa-7- azatricyclo[14.3.1.16l l0]henicosa-l(20), 6(21), 7, 9, 16, 18-hexaen-14-yl]carbamate. MS: 532.0 (M+l)
ES+
Step B: Boc deprotection
A solution of tert-butyl [14-methyl-8-[methyl(methylsulfonyl)amino]-13-oxo-12-oxa-7- azatricyclo[14.3.1.16' 10]henicosa-l(20), 6(21), 7, 9, 16, 18-hexaen-14-yl]carbamate (22 mg, 0.041 mmol) in DCM (0.5 mL) and TFA (0.5 mL) was stirred at rt for 1.5 h. Concentration and purification by reverse phase preparative HPLC (5-95% CH3CN in water containing 0.1% TFA) gave N-[14-amino-14-methyl- 13-oxo-12-oxa-7-azatricyclo[14.3.1.16' 10]henicosa-l(20), 6(21), 7, 9, 16, 18-hexaen-8-yl]-N- methylmethanesulfonamide as the TFA salt. Exact Mass calc for C22H29Ν3O4S: 432.1952; measured: 432.1956.
EXAMPLE 15
N-[13-amino-18-chloro-13-methyl-14-oxo-15-oxa-2,20-diazatricyclo[15.3.1.17>11]docosa- 1 (21),7(22),8, 10, 17, 19-hexaen- 19-yl]-N-methylmethanesulfonamide
Figure imgf000062_0001
To a solution of Example 3 (0.440 g, 0.785 mmol) in 13 mL CH2Cl2 was added NCS (0.105 g, 0.785 mmol), and the reaction was allowed to proceed at rt for 4 days. The reaction was concentrated and purified by preparative HPLC to afford the title compound. LC/MS [MH-H]+ = 481.
While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention. It is intended, therefore, that the invention be defined by the scope of the claims that follow and that such claims be interpreted as broadly as is reasonable.

Claims

IU- if i-' it ,<■ «,.... ...* ...J'WHAT IS CLAIMED IS:
1. A compound of formula (T):
Figure imgf000063_0002
(I)
wherein:
Y is selected from the group consisting of
(1) hydrogen,
(2) -C 1-3 alkyl, wherein said alkyl is optionally substituted with one or more halogen,
(3) halogen, and
(4) cyano;
A is selected from the group consisting of (1) hydrogen, (2) -Ci-10 alkyl, (3) -C2-10 alkenyl, and
(4) -C2-10 alkynyl wherein said alkyl, alkenyl or alkynyl is unsubstituted or substituted with one or more
(a) halo,
(b) -C3-8 cycloalkyl, (c) -OH,
(d) -CN, (e) -0-Ci_io alkyl,
(f) -C6-10 aryl, or
(g) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
and said aryl and heteroaryl groups are unsubstituted or substituted with one or more
(i) halo, (ϋ) -OH, (Ui) -CN,
(iv) -0-Ci-io alkyl, (v) -Ci-io alkyl,
(vi) -C2-lθ alkenyl,
(vii) -C2-10 alkynyl, or (viii) -C3-8 cycloalkyl;
Rl is selected from the group consisting of
(1) -C6-10 arylene, or
(2) heteroarylene selected from the group consisting of divalent pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
wherein said arylene or heteroarylene is unsubstituted or substituted with one or more (a) halo,
(b) -Ci-10 alkyl, (c) -C2-IO alkenyl,
(d) -C2-IO alkynyl, (e) -OH,
(f) -CN, (g) -O-Ci_l0 alkyl, or (h) -C3.8 cycloalkyl;
R2 is selected from the group consisting of: (1) (R5-SO2)N(R6)-, wherein R5 is
(a) -Ci-io alkyl, (b) -C2-IO alkenyl,
(c) -C2-lθ alkynyl, (d) -C3.8 cycloalkyl, [( 1|«Λ -JI .<" ~ IT II-m- B™, ..,.,1' ,.,..U
(e) -C6-10 aryl, or
(f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
wherein said alkyl, alkenyl, alkynyl, cycloalkyl, aryl and heteroaryl is unsubstituted or substituted with one or more (i) halo,
(ii) -OH, (iii) -CN,
(iv) -0-Ci-io alkyl, (v) -Ci-IO alkyl, (vi) -C2- 10 alkenyl,
(vii) -C2-10 alkynyl, (viii) — C3_8 cycloalkyl, (ix) -C6-10 aryl, or
(x) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, and said aryl and heteroaryl is unsubstituted or substituted with one or more
(A) halo, (B) -OH, (C) -CN,
(D) -O-Ci-iO alkyl, (E) -C3_8 cycloalkyl,
(F) -Ci-io alkyl, (G) -C2-10 alkenyl, or (H) -C2-IO alkynyl;
s selected from the group consisting of
(a) hydrogen, (b) -CLIO alkyl, , . „
Ir- i[«,i> Ii ,.' <t,,,'j .„„[1 (f«,i! ,,...ii ,.' ""irιι,,,n ιι,,,« ,„"» „„;«
(c) -C2-10 alkenyl,
(d) -C2-10 alkynyl,
(e) -C6-10 aryl, or
(f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
wherein said alkyl, alkenyl, alkynyl, aryl or heteroaryl. is unsubstituted or substituted with one or more
(i) halo,
(ii) -OH,
(iii) -CN, (Iv) -O-Ci-IO alkyl,
(v) -C3-8 cycloalkyl,
(vi) -C6-10 aryl. or
(vii) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl;
wherein said cycloalkyl, aryl or heteroaryl is unsubstituted or substituted with one or more
(A) halo,
(B) -OH,
(C) -CN,
(D) -O-Ci-io alkyl, (E) -C3-8 cycloalkyl, or
(F) -Ce-IO aryl,
or R5 and R^ may be linked to form a group -CH2(CH2)pCH2-,
(2) -C6-10 aryl, wherein said aryl is unsubstituted or substituted with one or more
(i) halo, (ϋ) -OH, , , ϋ- ir,,,ι> Ii , >[.„!' ;;,,,n ιi,.,ιι ....u ,.' -ιi n,,.,, IU. .....n ..._p
(iii) -CN,
(iv) -0-Ci_io alkyl, (v) -C3.8 cycloalkyl, (vi) -Ci-io alkyl, (vi) -C6-10 aryl,
(3)
Figure imgf000067_0001
(4) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl,pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, wherein said heteroaryl is unsubstituted or substituted with one or more
(i) halo,
(ϋ) -OH,
(iii) -CN,
(iv) -0-Ci-io alkyl,
(v) -C3-8 cycloalkyl,
(vi) -Ci-io alkyl,
(vii) -C(O)-O-Ci-IO alkyl,
(viii) -C(=O)-OH, and
Figure imgf000067_0002
(x) -NRCRd, wherein Rc and Rd are selected from the group consisting of
(A) hydrogen, and
(B) -Ci-io alkyl,
(5) hydrogen, and
(6) -CF3;
R3 i is
Figure imgf000068_0001
wherein if the dotted line leading to Q3 is absent, then Q3 is selected from the group consisting of (a) -CH2- (b) -O-,
(c) -NRX-,
(d) -CC=O)-, and
(e) -CC=O) -NRX-, wherein Rχ is selected from the group consisting of (i) hydrogen,
(ii) -CM0 alkyl,
(iii)-C2-10 alkenyl, (iv) -C2-10 alkynyl, (v) -C3-8 cycloalkyl, (vi) -Cθ-6 alkylene-C6-10 aryl, or
(vii) -Cθ-6 alkylene-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
(viii) -Cθ-6 alkyl-C3_8 cycloalkyl,
and if the dotted line leading to Q^ represents a bond, then Q^ is -CH- or -CH2-CH-,
if the dotted line leading to RY is absent, then RY is selected from the group consisting of
(a) hydrogen,
(b) -Ci-10 alkyl,
(c)-C2-io alkenyl,
(d) -C2-IO alkynyl,
(e) -C3-8 cycloalkyl, ,'■■
(f) -Co-6 alkylene-C6-10 aryl, or
(g) -Co-6 alkylene-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl,
and if the dotted line leading to RY represents a bond, then RY is selected from the group consisting of (a) =CH-Ci_iθ alkyl, (b) =CH-C0-6 alkyl-C6-10 aryl,
wherein said alkyl, alkenyl, alkynyl, cycloalkyl, aryl or heteroaryl RY groups are unsubstituted or substituted with one or more
(i) halo, (ii) -Ci-10 alkyl,
(iii) -OH,
(iv) -CN, or (V) -O-Ci-IO alkyl, or .
(vi) -C3-8 cycloalkyl,
Q4 is selected from the group consisting of (a) -CH2 -
(b) -O-, and (C) -NRZ- wherein Rz is selected from the group consisting of
(i) hydrogen,
(ii) -Ci-10 alkyl,
(Ui)-C2-IO alkenyl, (iv) -C2-IO alkynyl, (v) -C3-8 cycloalkyl,
(vi) -Co-6 alkyl-C6-10 83V^ or
(vii) -Cθ-6 alkyl-heteroaryl, wherein said heteroaryl is selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl, , \r if,., Ira" ι,,".iι IUl' „7, F ,.' ir ir,», «,.„ ,,,,ji ,„„!!
R4 is -(CH2)S-Q2 -(CH2)t, wherein Q2 is selected from the group consisting of
(i)-o-,
(2)-NH-, (3) -0-C(=O)-, (4) -C(=O)-O-,
(5) -NHC(O)-,
(6) -CC=O)-NH-,
(7) -CH=CH-, (8) -C(O)-, (9) -(CH2)U -,
Figure imgf000070_0001
m is O, 1 or 2; n is O, 1 or 2; p is 1, 2 or 3; q is 1, 2, 3, 4 or 5: r is O, 1 or 2; s is O or 1; t is O or 1; and u is 0, 1 or 2; and pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof.
2. The compound of Claim 1 wherein Y is hydrogen.
3. The compound of Claim 1 wherein Rl is phenylene.
4. The compound of any of Claims 1 -3 wherein R4 is -(CH2)s-Q2-(CH2)t, wherein Q2 is selected from the group consisting of
(1) -O-, wherein s and t are 1,
(2) -O-C(=O)-, wherein s is 1 and t is 0, (3)
Figure imgf000071_0001
(4)
Figure imgf000071_0002
wherein s and t are O.
5. The compound of Claim 4 wherein n and m are each 1.
6. The compound of any of Claims 1-5 wherein A is selected from the group consisting of (1) hydrogen, and
(2) -Ci_iθ alkyl, wherein said alkyl is unsubstituted or substituted with one or more
(a) halo,
(b) -C3.8 cycloalkyl,
(c) -CN
(d) -0-Ci_io alkyl, (e) -C6-10 aryl, or
(f) heteroaryl selected from the group consisting of pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, tetrazolyl, furanyl, imidazolyl, triazinyl, pyranyl, thiazolyl, thienyl, thiophenyl, triazolyl, oxazolyl, isoxazolyl, thiazolyl, oxadiazolyl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, indynyl and benzoxazolyl.
7. The compound of any of Claims 1-6 wherein R.2 is (R5-SO2)N(R6)-, wherein R5 is -Ci_ alkyl, wherein said alkyl is unsubstituted or substituted with one or more
(i) halo, (ϋ) -OH,
(iii) -CN,
(iv) -O-Ci-6 alkyl, or (v) -C 1-6 alkyl, and
R6 is selected from the group consisting of (a) hydrogen,
(b) -Ci_6 alkyl, (c) -C6-lθ aryl, wherein said alkyl and aryl is unsubstituted or substituted with one or more (i) halo, i > _ .
(ii) -OH,
(iii) -CN,
(iv) -O-Ci_6 alkyl,
(v) -Ci_6 alkyl,
8. The compound of any of Claims 1-6 wherein R2 is phenyl, unsusbstituted or substituted with cyano.
9. The compound of any of claims 1 -6 wherein R2 is
Figure imgf000072_0001
; wherein q is 1, 2 or 3.
10. The compound of any of Claims 1-6 wherein R2 is
Figure imgf000072_0002
wherein Ql is selected from the group consisting of
(a) N, and
(b) C-Rb, wherein Rb is selected from the group consisting of
(i) -CN, and (ii) -C(=0)-0-Ci_io alkyl,
(iii) -C(=O)-OH, and (iv) -C(O)-NRCRd
(v) -NR0Rd, wherein Rc and Rd are selected from the group consisting of (A) hydrogen, and
(B) -Ci_io alkyl.
11. The compound of any of Claims 1-10 wherein the dotted line leading to Q3 is absent, and Q3 is NRX, Rχ is hydrogen, and n is 1.
12. The compound of any of Claims 1-10 wherein the dotted line leading to Q3 is absent, and Q3 is O and n is 1. It" 1-.,Ii Il , ' '[,„!' Ljf l'».« ,,,Jl .•' Il 'I,,,,, II,™ .,,Jl ,,,,,It
13. The compound of any of Claims 1-10, wherein the dottled line leading to Q3 is -C(=O) - NRX, Rχ is hydrogen, and n is 1.
14. The compound of any of Claims 1-13, wherein the dotted line leading to Ry is absent and RY is selected from the group consisting of
(a) hydrogen,
O) -Cl-IO alkyl, or
(c) -Co-6 alkylene-C6-10 yh wherein said alkyl or aryl or unsusbsituted or substituted with one or more halogen.
15. The compound of any of Claims 1-13, wherein Q4 is CH2, m is 1 and r is 0.
16. The compound of Claim 1 which is a compound of Formula (II):
Figure imgf000073_0001
(H) and pharmaceutically acceptable salts thereof, and individual enantiomers and diastereomers thereof
17. A pharmaceutical composition comprising a therapeutically effective amount of a compound of Claim 1 and a pharmaceutically acceptable carrier.
18. A method for inhibition of β-secretase activity in a mammal in need thereof which comprises administering to the mammal a therapeutically effective amount of a compound of Claim 1.
19. A method for treating Alzheimer's disease in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Claim 1.
PCT/US2005/042233 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease WO2006057983A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA002587256A CA2587256A1 (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease
US11/791,465 US7951949B2 (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of Alzheimer's disease
JP2007543382A JP2008520727A (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of Alzheimer's disease
AT05849049T ATE512147T1 (en) 2004-11-23 2005-11-18 MACROCYCLIC AMINIOPYRIDYL BETA SECRETASE INHIBITORS FOR THE TREATMENT OF ALZHEIMER'S DISEASE
AU2005309708A AU2005309708A1 (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of Alzheimer's disease
EP05849049A EP1817312B1 (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63031904P 2004-11-23 2004-11-23
US60/630,319 2004-11-23

Publications (1)

Publication Number Publication Date
WO2006057983A1 true WO2006057983A1 (en) 2006-06-01

Family

ID=36498305

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/042233 WO2006057983A1 (en) 2004-11-23 2005-11-18 Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer's disease

Country Status (8)

Country Link
US (1) US7951949B2 (en)
EP (1) EP1817312B1 (en)
JP (1) JP2008520727A (en)
CN (1) CN101061119A (en)
AT (1) ATE512147T1 (en)
AU (1) AU2005309708A1 (en)
CA (1) CA2587256A1 (en)
WO (1) WO2006057983A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006055434A3 (en) * 2004-11-17 2007-07-05 Merck & Co Inc Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008104580A1 (en) 2007-03-01 2008-09-04 Probiodrug Ag New use of glutaminyl cyclase inhibitors
EP2073635A1 (en) * 2006-10-06 2009-07-01 Merck & Co., Inc. Macrocyclic spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
US8183252B2 (en) 2003-12-15 2012-05-22 Schering Corporation Heterocyclic aspartyl protease inhibitors
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
US8729071B2 (en) 2009-10-08 2014-05-20 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions and their use
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104263795A (en) * 2014-08-22 2015-01-07 四川同晟生物科技有限公司 Method for preparing chiral alpha-naphthenic glycine

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006015621A1 (en) * 2004-08-09 2006-02-16 Cellzome Ag Treatment of neurodegenerative diseases by the use of scd4 inhibitors

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001051126A1 (en) * 2000-01-12 2001-07-19 Merck & Co., Inc. Inhibitors of prenyl-protein transferase
US6333410B1 (en) * 2000-08-18 2001-12-25 Immunogen, Inc. Process for the preparation and purification of thiol-containing maytansinoids
CA2450167A1 (en) 2001-06-12 2002-12-19 Elan Pharmaceuticals, Inc. Macrocycles useful in the treatment of alzheimer's disease
AU2003276047A1 (en) 2002-06-17 2003-12-31 Sunesis Pharmaceuticals, Inc. Aspartyl protease inhibitors
WO2004043916A1 (en) * 2002-11-12 2004-05-27 Merck & Co., Inc. Phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
AU2004204692B2 (en) * 2003-01-07 2009-07-09 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for treatment of Alzheimer's disease
EP1644322A1 (en) 2003-06-16 2006-04-12 Sunesis Pharmaceuticals, Inc. Aspartyl protease inhibitors
WO2005004802A2 (en) * 2003-06-30 2005-01-20 Merck & Co., Inc. N-alkyl phenylcarboxamide beta-secretase inhibitors for the treatment of alzheimer's disease
CA2530006A1 (en) * 2003-07-01 2005-01-20 Merck & Co., Inc. Phenylcarboxylate beta-secretase inhibitors for the treatment of alzheimer's disease
CA2535337A1 (en) * 2003-08-14 2005-03-03 Merck & Co., Inc. Macrocyclic beta-secretase inhibitors for the treatment of alzheimer's disease
CA2540452A1 (en) * 2003-10-03 2005-04-14 Merck & Co., Inc. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer's disease
EP1689713B1 (en) 2003-11-24 2011-07-27 Merck Sharp & Dohme Corp. Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer s disease
CA2548849A1 (en) 2003-12-19 2005-07-21 Merck & Co., Inc. Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of alzheimer's disease
AU2005236020A1 (en) 2004-04-20 2005-11-03 Merck & Co., Inc. 1,3,5-substituted phenyl derivative compounds useful as beta-secretase inhibitors for the treatment of Alzheimer's disease
JP4764418B2 (en) 2004-04-20 2011-09-07 メルク・シャープ・エンド・ドーム・コーポレイション 2,4,6-substituted pyridyl derivative compounds useful as β-secretase inhibitors for the treatment of Alzheimer's disease
WO2006055434A2 (en) 2004-11-17 2006-05-26 Merck & Co., Inc. Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006015621A1 (en) * 2004-08-09 2006-02-16 Cellzome Ag Treatment of neurodegenerative diseases by the use of scd4 inhibitors

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8183252B2 (en) 2003-12-15 2012-05-22 Schering Corporation Heterocyclic aspartyl protease inhibitors
US7678783B2 (en) 2004-11-17 2010-03-16 Merck Sharp & Dohme Corp. Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2006055434A3 (en) * 2004-11-17 2007-07-05 Merck & Co Inc Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer's disease
EP2073635A1 (en) * 2006-10-06 2009-07-01 Merck & Co., Inc. Macrocyclic spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
EP2073635A4 (en) * 2006-10-06 2010-12-29 Merck Sharp & Dohme Macrocyclic spiropiperidine beta-secretase inhibitors for the treatment of alzheimer's disease
WO2008055945A1 (en) 2006-11-09 2008-05-15 Probiodrug Ag 3-hydr0xy-1,5-dihydr0-pyrr0l-2-one derivatives as inhibitors of glutaminyl cyclase for the treatment of ulcer, cancer and other diseases
WO2008065141A1 (en) 2006-11-30 2008-06-05 Probiodrug Ag Novel inhibitors of glutaminyl cyclase
WO2008104580A1 (en) 2007-03-01 2008-09-04 Probiodrug Ag New use of glutaminyl cyclase inhibitors
EP2481408A2 (en) 2007-03-01 2012-08-01 Probiodrug AG New use of glutaminyl cyclase inhibitors
EP2865670A1 (en) 2007-04-18 2015-04-29 Probiodrug AG Thiourea derivatives as glutaminyl cyclase inhibitors
WO2011029920A1 (en) 2009-09-11 2011-03-17 Probiodrug Ag Heterocylcic derivatives as inhibitors of glutaminyl cyclase
US8940748B2 (en) 2009-10-08 2015-01-27 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions, and their use
US8729071B2 (en) 2009-10-08 2014-05-20 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions and their use
US9029362B2 (en) 2009-10-08 2015-05-12 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as brace inhibitors, compositions, and their use
US9428475B2 (en) 2009-10-08 2016-08-30 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions, and their use
US9475785B2 (en) 2009-10-08 2016-10-25 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions and their use
US9687494B2 (en) 2009-10-08 2017-06-27 Merck Sharp & Dohme Corp. Iminothiadiazine dioxide compounds as BACE inhibitors, compositions, and their use
WO2011107530A2 (en) 2010-03-03 2011-09-09 Probiodrug Ag Novel inhibitors
WO2011110613A1 (en) 2010-03-10 2011-09-15 Probiodrug Ag Heterocyclic inhibitors of glutaminyl cyclase (qc, ec 2.3.2.5)
WO2011131748A2 (en) 2010-04-21 2011-10-27 Probiodrug Ag Novel inhibitors
WO2012123563A1 (en) 2011-03-16 2012-09-20 Probiodrug Ag Benz imidazole derivatives as inhibitors of glutaminyl cyclase
EP3461819A1 (en) 2017-09-29 2019-04-03 Probiodrug AG Inhibitors of glutaminyl cyclase

Also Published As

Publication number Publication date
CA2587256A1 (en) 2006-06-01
ATE512147T1 (en) 2011-06-15
US20080015213A1 (en) 2008-01-17
CN101061119A (en) 2007-10-24
EP1817312A1 (en) 2007-08-15
US7951949B2 (en) 2011-05-31
EP1817312A4 (en) 2009-11-04
AU2005309708A1 (en) 2006-06-01
EP1817312B1 (en) 2011-06-08
JP2008520727A (en) 2008-06-19

Similar Documents

Publication Publication Date Title
EP1817312B1 (en) Macrocyclic aminopyridyl beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
JP6744355B2 (en) Antifibrotic pyridinone
EP1758854B1 (en) Pyrrolidin-3-yl compounds useful as beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
EP1855679B1 (en) Aminomethyl beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
EP1814537B1 (en) Macrocyclic tertiary amine beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
WO2006078577A1 (en) Tertiary carbinamines having substituted heterocycles, which are active as inhibitors of beta-secretase, for the treatment of alzheimer&#39;s disease
CA2768881A1 (en) Fused aminodihydropyrimidone derivatives
CA2540452A1 (en) Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
KR20180128404A (en) Naphthyridine as an integrin antagonist
WO2018001332A1 (en) Compound having inhibitory activity against mutant isocitrate dehydrogenase, preparation method therefor and use thereof
CN113490668A (en) Compounds and compositions for treating diseases associated with APJ receptor activity
WO2021263278A1 (en) Rev-erb agonists for the treatment of th17-mediated inflammatory disorders
TW202233573A (en) Novel substituted 6,7-dihydro-5h- benzo[7]annulene derivatives, processes for their preparation and therapeutic uses thereof
AU2019424628B2 (en) 1,2,3,4-tetrahydroquinoxaline derivative, preparation method therefor and application thereof
WO2007019080A2 (en) Tricyclic beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
EP1912963B1 (en) Cyclic ketal beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
TWI833819B (en) Compounds and compositions for treating conditions associated with apj receptor activity

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 1772/CHENP/2007

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2005309708

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2587256

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2005849049

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200580039921.8

Country of ref document: CN

Ref document number: 2007543382

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 11791465

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2005309708

Country of ref document: AU

Date of ref document: 20051118

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005849049

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 11791465

Country of ref document: US