WO2009126624A1 - Triazolo compounds useful as dgat1 inhibitors - Google Patents

Triazolo compounds useful as dgat1 inhibitors Download PDF

Info

Publication number
WO2009126624A1
WO2009126624A1 PCT/US2009/039770 US2009039770W WO2009126624A1 WO 2009126624 A1 WO2009126624 A1 WO 2009126624A1 US 2009039770 W US2009039770 W US 2009039770W WO 2009126624 A1 WO2009126624 A1 WO 2009126624A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
alkyl
compounds
substituted
mmol
Prior art date
Application number
PCT/US2009/039770
Other languages
French (fr)
Inventor
Yanting Huang
Chongqing Sun
R. Michael Lawrence
William R. Ewing
Huji Turdi
Original Assignee
Bristol-Myers Squibb Company
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 Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Priority to US12/936,969 priority Critical patent/US8324241B2/en
Publication of WO2009126624A1 publication Critical patent/WO2009126624A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Definitions

  • the monoacylglycerol pathway initiates from acyl coenzyme A:monoacylglycerol acyltransferase (MGAT) (EC 2.3.1.22). Within minutes of its appearance from the digestion of dietary fat in the lumen of the small intestine, 2-monoacylglycerol is acylated by MGAT to form diacylglycerol. Diacylglycerol is further acylated by acyl coenzyme A: diacylglycerol acyltransferase (DGAT) (EC 2.3.1.20) to re-synthesize triacylglycerol, which is packaged into chylomicron lipoprotein particles that eventually are secreted into the lymph.
  • DGAT diacylglycerol acyltransferase
  • DGAT lies at the final step of both triacylglycerol synthesis pathways.
  • Two DGAT enzymes have been identified and have been designated as DGATl and DGAT2 (Cases, S. et al., Proc. Natl Acad.
  • DGATl and DGAT2 are encoded by two different genes that bear little sequence homology. Functionally, these two enzymes might have different physiological importance in vivo. DGATl knockout mice exhibit resistance towards becoming obese when challenged with a high fat (Smith, SJ. et al., Nat. Genet., 25:87-90 (2000)).
  • Patent 7,300,932 B2 discloses fused bicyclic nitrogen-containing heterocyclic compounds that are useful for treating or preventing conditions and disorders associated with DGAT. As may be appreciated, there still remains a need for new compounds that are inhibitors of DGAT and are useful for the treatment of DGAT related conditions and disorders. [0004] Applicants have found triazolopyridine compounds that have activity as inhibitors of DGAT, in particular DGATl, and are thereby useful in therapy.
  • A is hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl, -C(O)R 6 , -C(O)OR 6 , or -C(O)NR 6 R 7 ;
  • L is -(CR 1 R 2 V; n is 1, 2, or 3;
  • B is -OR 5 , -C(O)OR 5 , -OC(O)R 5 , or -OC(O)OR 3 ; one of X and Y is CH or N, and the other of X and Y is CH;
  • R 1 and R 2 are, independently at each occurrence, hydrogen, alkyl, cycloalkyl, aryl, and/or heterocyclyl, or one R 1 and one R 2 form a Cs-C ⁇ cycloalkyl or 4- to 7- membered heterocyclyl ring having one or two heteroatoms;
  • R 3 is hydrogen or alkyl, or R 3 and R 1 form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms;
  • R 4 is hydrogen or alkyl
  • R 5 is hydrogen or alkyl
  • R 6 is alkyl, cycloalkyl, aryl, or heterocyclyl
  • R 7 is hydrogen or alkyl, or R 7 and R 6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms
  • each of said alkyl is substituted with 0-3 R a
  • each of said cycloalkyl is substituted with 0-3 R a
  • each of said aryl is substituted with 0-4 R
  • each of said heterocyclyl is substituted with 0-4 R b ;
  • R a is, independently at each occurrence, F, Cl, Br, -CF 3 , -OH, -OCH 3 , -OCF 3 , -CN, -NR c R d , phenyl, imidazolyl, and/or C 1 - Qalkoxy;
  • R b is, independently at each occurrence, Q ⁇ alkyl, F, Cl, Br, -CF 3 , -OH, -OCH 3 , -OCF 3 , -CN, -NR c R d , and/or C,-C 3 alkoxy;
  • R c and R d are, independently at each occurrence, H and/or C r C 4 alkyl, or R c and R d together with the nitrogen atom to which they are attached, form a 4- to 7- membered heterocyclyl ring with one or two heteroatoms.
  • composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier or diluent.
  • method for treating a condition or disorder comprising administering to a patient in need thereof at least one compound of Formula (I); wherein said condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis.
  • alkyl and “alk” refer to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 12 carbon atoms, preferably from 1 to 6 carbon atoms, and more preferably from 1 to 4 carbon atoms.
  • alkyl and/or “alk” groups include, but are not limited to, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isohexyl, heptyl, octyl, nonyl, decyl, and dodecyl.
  • the term "lower alkyl” refers to an “alkyl” and/or “alk” group containing from 1 to 4 carbon atoms and preferably from 1 to 2 carbon atoms. When a subscript is used with reference to an alkyl or other group, the subscript refers to the number of carbon atoms the group may contain.
  • Co-C4alkyl includes a bond and an alkyl group containing 1 to 4 carbon atoms
  • CrC 4 alkyl refers to alkyl groups containing 1 to 4 carbon atoms.
  • Exemplary lower alkyl groups include, but are not limited to, for example, methyl, ethyl, propyl including n-propyl and isopropyl, and butyl including n-butyl, isobutyl, and t-butyl.
  • the "alkyl” and/or “a ⁇ k” group can be optionally substituted with one or more substituents, preferably 1 to 3 substituents, at any available and substitutable position.
  • substituents include halogen (e.g., a single halo substituent or multiple halo substituents form, in the latter case, groups such as, for example, a perfluoroalkyl group or an alkyl group bearing -CCI 3 or -CF 3 ), hydroxyl, -NH 2 , -NH(alkyl), -CF 3 , -N(alkyl) 2 , cyano, Ci-C 3 alkoxy group, phenyl, imidazolyl, and halogenated alkoxy group such as -OCF 3 .
  • halogen e.g., a single halo substituent or multiple halo substituents form, in the latter case, groups such as, for example, a perfluoroalkyl group or an alkyl group bearing -CCI 3 or -CF 3 ), hydroxyl, -NH 2 , -NH(alkyl), -CF 3 , -N(alkyl) 2 ,
  • cycloalkyl refers to a fully saturated hydrocarbon group containing from 1 to 4 rings and 3 to 8 carbon atoms per ring.
  • exemplary cycloalkyl groups include, but at not limited to, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl.
  • the cycloalkyl group can be optionally substituted with one or more substituents, preferably 1 to 3 substituents, at any available and substitutable point of attachment.
  • substituents include those groups recited for substituted alkyl.
  • aryl refers to cyclic aromatic hydrocarbon groups having from 1 to 2 aromatic rings, such as, for example, phenyl, biphenyl, or naphthyl.
  • the aromatic rings may be joined at a single point (e.g., biphenyl) or fused (e.g., naphthyl and phenanthrenyl).
  • the aryl group can be optionally substituted with one or more substituents, preferably 1 to 4 substituents, at any available and substitutable ring position, or where valence allows on any rings fused or attached thereto.
  • substituents include alkyl and those groups recited for substituted alkyl.
  • heterocyclo represents 3-, 4-, 5 ⁇ , 6-, or 7-membered monocyclic or polycyclic or 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered polycyclic heterocyclic ring that is saturated, partially unsaturated, or fully unsaturated, and that contains carbon atoms and i , 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring.
  • the nitrogen and sulfur heteroatoms may optionally be oxidized to -NO-, -SO-, or -SO 2 -.
  • the heterocyclic ring may be attached to its pendant group at any heteroatora or carbon atom which results in the creation of a stable structure.
  • the heterocycle, heterocyclic, or heterocyclo group can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl. When the term "heterocycle" is used, it is intended to include heteroaryl.
  • heterocyclo, heterocycle, heterocyclic, or heterocyclo group can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl.
  • substituents include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazoly ⁇ , benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnoliny ⁇ , decahydroquinolinyl, 2H,6H ⁇ l,5,2 ⁇ dithiazinyl
  • fused ring and spiro compounds containing, for example, the above heterocycles are fused ring and spiro compounds containing, for example, the above heterocycles.
  • 5- to 10-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, mo ⁇ holinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyi, benzimidazolyl, lH-indazoIyl, benzofuranyl, benzothiofuranyl, benzt
  • the terra "aromatic heterocyclic group” or “heteroaryl” is intended to mean fully unsaturated heterocyclyl rings, including monocyclic and polycyclic aromatic hydrocarbons having at least one heteroatom ring member such as sulfur, oxygen, or nitrogen.
  • Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyi, tetrazolyl indazolyl, indolmyl, 1,2,4- thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, benzodioxolanyl, and benzodioxane.
  • Heteroaryl groups are unsubstituted or substituted. Heteroaryl groups can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl.
  • alkoxy as employed herein alone or as part of another group includes an alkyl as defined above linked through an oxygen atom.
  • halogen or “halo” as used herein alone or as part of another group refers to fluorine, chlorine, bromine, and iodine, with fluorine, chlorine, and bromine being preferred.
  • cyano refers to a -CN group.
  • methylene refers to a -CH 2 - group.
  • the compounds of the present invention can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of the present invention have, for example, at least one basic center, they can form acid addition salts.
  • organic carboxylic acids such as alkanecarboxylic acids of 1 to 4 carbon atoms, for example acetic acid, which are unsubstituted or substituted, for example, by halogen as chloroacetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, nialeic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (Ci-C 4 ) alkyl or arylsulfonic acids which are unsubstituted or substituted,
  • Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center.
  • the compounds of Formula ( ⁇ ) having at least one acid group can also form salts with bases.
  • Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as morpholine, thiornorpholine, piperidine, pyrrolidine, a mono-, di-, or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl- propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine.
  • Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of Formula (I) or their pharmaceutically acceptable salts, are also included.
  • Preferred salts of the compounds of Formula (I) which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate, nitrate or acetate.
  • Preferred salts of the compounds of Formula (I) which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines,
  • modulator refers to a chemical compound with capacity to either enhance (e.g.
  • agonist activity or partially enhance (e.g. , “partial agonist” activity) or inhibit (e.g., “antagonist” activity or “inverse agonist” activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types.
  • biological activity or process e.g., enzyme activity or receptor binding
  • bioactive metabolites include, but are not limited to, -OH, -NH 2 , or functional groups wherein the hydrogen can be replaced with a functional group such as -PO 3 H 2 for example, which, upon biotransformation generates an -OH or -NH 2 functional group of a compound of Formula (I).
  • prodrug as employed herein includes functionalization of bioactive amine- or hydroxyl-containing compounds of Formula (I) to form alkyl-, acyl-, sulfonyl-, phosphoryl-, or carbohydrate-substituted derivatives.
  • Such derivatives are formed by reacting compounds of Formula (I) with alkylating-, acylating-, sulfonylating-, or phosphorylating reagents employing procedures known to those skilled in the art.
  • Alkyiation of amines of Formula (I) may result in, but is not limited to, derivatives that include spacer units to other prodrug moieties such as substituted alkyoxymethyl-, acyloxymethyl-, phosphoryloxymethyl-, or sulfonyloxymethyl- groups.
  • Alkyiation of amines of Formula (I) may result in the generation of quarternary amine salts that act in vivo to provide the bioactive agent (i.e., the compound of Formula (I)).
  • Preferred prodrugs consist of a compound of Formula (I) where a pendant hydroxyl is phosphorylated to generate a phosphate derivative. Such a prodrug may also include a spacer group between the compound of Formula (I) and the phosphate group, such as a methyleneoxy-group. Methods to generate such a prodrug from a compound of Formula (I) are known to those skilled in the art, and are listed in the references below. [0030] Preferred prodrugs also consist of a compound of Formula (I) where a pendant amine, such as a pyridine group, is alkylated with a group, such as methyl, to form a quarternary ammonium ion salt.
  • Bundgaard (Elsevier, 1985); c) A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds., Ch. 5, pp. 113-191 (Harwood Academic Publishers, 1991); d) Hydrolysis in Drug and Prodrug Metabolism, B. Testa and J.M. Mayer (Verlag Helvetica Chimica Acta AG, Zurich, Switzerland; Wiley- VCH, Weinheim,
  • the term "patient” as used herein encompasses all mammalian species including humans, cows, horses, dogs, and cats; and preferably, humans.
  • the term "therapeutically effective” is intended to qualify the amount of each agent, which will treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration.
  • the compounds of this embodiment have structures represented by Formula (Ia): and stereoisomers or salts thereof, wherein: A, L, B, R 3 , and R 4 are as defined hereinabove.
  • the compounds of Formula (Ia) are triazolopyridine derivatives. [0037] In one embodiment, compounds of Formula (I) are provided where X is N and Y is CH.
  • the compounds of this embodiment have structures represented by Formula (Ib):
  • compounds of Formula (Ic) are triazolopyrazine derivatives.
  • L is - (CR 1 R 2 )!,-, n is 1 or 2, and R 1 and R 2 are as defined hereinabove.
  • suitable L groups include, but are not limited to, -CR 1 R 2 - in which n is 1 and -CHR ! - in which n is 1 and R 2 is hydrogen.
  • R J and R 2 are, independently at each occurrence, hydrogen, C 1 -Ce alkyl, C 3 -C 7 cycloalkyl, aryl, and/or 1- or 2-ring heterocyclyl; and each alkyl is substituted with 0-3 R a , each cycloalkyl is substituted with 0-3 R a , each aryl is substituted with 0-4 R b , and each heteroaryl is substituted with 0-4 R b ; and R a and R b are as defined hereinabove.
  • R 1 is hydrogen, Q-Qalkyl, C 3 ⁇ C 7 cycloalkyl, or phenyl; and R 2 is hydrogen or methyl. More preferably, R ! is hydrogen, Q ⁇ alkyl, Cs-Cvcycloakyl, or phenyl; and R 2 is hydrogen.
  • R 1 is hydrogen, C]-C 4 alkyl, hydroxyethyl, cyclohexyl, phenyl, or methyl substituted with phenyl, hydroxyphenyl or imidazolyl; and R 2 is hydrogen.
  • compounds of Formula (I) wherein one R 1 and one R 2 form a C 3 -C 7 cycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein said cycloalkyl ring is substituted with 0-3 R a and said heterocyclyl ring is substituted with 0-4 R b , and R a and R b are as defined hereinabove.
  • R 1 and R 2 form a Cs-C ⁇ cycloalkyl, and more preferably, R 1 and R 2 form a C5-C 7 cycloalkyl.
  • this embodiment provides compounds of Formula (I) wherein R 1 and R 2 form a cyclohexyl group.
  • compounds of Formula (I) are provided wherein n is 2 and L is -CR 1 R 2 CR 1 R 2 -, and R 1 attached to one carbon atom of L and R 2 attached to the other carbon atom of L, form a C 3 -C 7 CyClOaIkVl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein said cycloalkyl ring is substituted with 0-3 R a and said heterocyclyl ring is substituted with 0-4 R b , and R a and R b are as defined hereinabove.
  • L is -CHR 1 CHR 2 -.
  • R 3 is hydrogen or Ci-C ⁇ alkyl, wherein each alkyl is substituted with 0-3 R a and R a is as defined hereinabove.
  • R 3 is hydrogen or Ci-C 4 alkyl, and more preferably hydrogen and Q-Cialkyl.
  • suitable R 3 groups include, but are not limited to, hydrogen and methyl. Still more preferably, R 3 is hydrogen.
  • compounds of Formula (I) are provided wherein R 3 and R 1 form a 4 ⁇ to 7-membered heterocyclyl ring having one or two heteroatoms, wherein the heterocyclyl ring is substituted with 0-4 R b .
  • suitable heteroatoms include nitrogen, oxygen, and sulfur.
  • the heterocyclyl ring is saturated or partially unsaturated.
  • the heterocyclyl ring is saturated.
  • suitable heterocyclyl rings include 5- to 7-membered heterocyclyl rings such as imidazolyl, pyrrol idinyl, and piperiditiyl.
  • R 4 is hydrogen or Cj-Cealkyl, wherein each alkyl is substituted with 0-3 R a and R a is as defined hereinabove.
  • R 4 is hydrogen or Ci-C 4 EIkVl, and more preferably hydrogen and Examples of suitable R 4 groups include, but are not limited to, hydrogen and methyl.
  • R 4 is hydrogen
  • compounds of Formula (I) are provided wherein B is -OR 5 , -C(O)OR 5 , -OC(O)R 5 , or -OC(O)OR 5 ; R 5 is hydrogen or Ci-Qalkyl; and wherein said alkyl is substituted with 0-3 R a ; and R a is defined hereinabove.
  • B is -OR 5 or -C(O)OR 5 ; and more preferably, B is -OH, -C(O)OH, or -C(O)OCH 2 CH 3 .
  • R 5 is hydrogen or Ci-C 4 alkyl.
  • this embodiment provides compounds of Formula (I) wherein B is -OH, -OCH 3 , -C(O)OH, -C(O)OCH 3 , and -C(O)OCH 2 CH 3 .
  • R 6 is Ci-C & alkyl, CrC ⁇ cycloalkyl, aryl, or 1- or 2-ring heterocyclyl; and said alkyl is substituted with 0-3 R a , said cycloalkyl is substituted with 0-4 R b , said aryl is substituted with 0-4 R , and said heterocyclyl is substituted with 0-4 R b ; and R a and R are as defined hereinabove.
  • compounds of Formula (I) or are provided wherein A is hydrogen, Cj-Cealkyl, C 3 -C 7 cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R 6 , -C(O)OR 6 , or -C(O)NR 6 R 7 ;
  • R 6 is alkyl, cycloalkyl, aryl, or heterocyclyl;
  • R 7 is hydrogen or alkyl, or R 7 and R 6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; and each of said alkyl is substituted with 0-3 R a ; each of said cycloalkyl is substituted with 0-3 R a ; each of said aryl is substituted with 0-4 R b ; and each of said heterocyclyl is substituted with 0-4 R b ; and R a and R b are as defined
  • the present embodiment provides compounds of Formula (I) in which A is a 1- or 2-ring heterocyclyl having 1 or 2 heteroatoms selected from nitrogen and sulfur.
  • A is hydrogen, Q-Cealkyl, C 3 -C 7 cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R 6 , -C(O)OR 6 , or -C(O)NR 6 R 7 ;
  • R 1 and R 2 are, independently at each occurrence, hydrogen, CrQalkyl, Cj-C 7 cycloalkyl, aryl, and/or 1- or 2-ring heterocyclyl, or one R 1 and one R 2 form a Q-C ⁇ cycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms;
  • R 3 is hydrogen or Q-Cgalkyl, or R 3 and R 1 form a 4- to 7-membered heterocyclyl ring
  • compounds of Formula (I) are provided wherein: A is hydrogen, C]-C 4 alkyl, phenyl, 1- or 2-ring heterocyclyl having 1- or 2-heteroatoms selected from S and N, -C(O)R 6 , -C(O)OR 6 , or -C(O)NR 6 R 7 ; R 1 is hydrogen, Ci-Cgalkyl, CyC ⁇ cycloalkyl, or phenyl; R is hydrogen or methyl; or R and R form a Cs-Cycycloalkyl ring or 5- to ⁇ -membered heterocyclyl ring having one or two heteroatoms; R 3 is hydrogen or Ci-C 4 alkyl, or R 3 and R 1 form a 5- to 7-membered heterocyclyl ring having one or two heteroatoms; R 4 is hydrogen or C
  • compounds of Formula (I) are provided wherein: A is hydrogen, -C(O)R 6 , -C(O)OR 6 , -C(O)NHR 6 , thiazolyl, or benzothiazolyl; B is -OH or -C(O)OR 5 ; R 1 is hydrogen, C r C 4 alkyl, C 5 -C 7 cycloalkyl, or phenyl; R 2 is hydrogen; or R 1 and R 2 form a Cs-Qcycloalkyl; R 3 is hydrogen or Q-Caalkyl, or R 3 and R 1 form a 5- to 6-merabered heterocyclyl ring having one heteroatom; R 4 is hydrogen or Cj-C ⁇ alkyl; R 5 is hydrogen or and n is 1 or 2; and each alkyl is substituted with 0-2 R a ; each cycloalkyl is substituted with 0-2 R a ; each heterocyclyl is substituted with 0-2
  • compounds of Formula (I) are provided wherein: A is hydrogen, -C(O)-CH 2 CH 3 , -C(O)-benzyl, -C(O)O-CH 3 , -C(O)O-(butyl), -C(O)NH- (trifluoromethylphenyl), -C(O)NH-(trifluoromethoxyphenyl), thiazolyl, or chlorobenzothiazolyl; B is -OH, -C(O)OH, or -C(O)OCH 2 CH 3 ; R 1 is hydrogen, Ci-C 4 alkyl, hydroxyelhyl, cyclohexyl, phenyl, or methyl substituted with phenyl, hydroxyphenyl, or imidazolyl; R 2 is hydrogen; or R 1 and R 2 form a cyclopentyl ring; R 3 is hydrogen, or R 3 and R 1 form a pyrrolidinyl
  • the monoacylglycerol pathway initiates from acyl coenzyme A: monoacylglycerol acyl transferase (MGAT) (EC 2.3.1.22).
  • 2- monoacyl glycerol is acylated by MGAT to form diacylglycerol.
  • Diacylglycerol is further acylated by acyl coenzyme A: diacylglycerol acyl transferase (DGAT) (EC 2.3.1.20) to re-synthesize triacylglycerol, which is packaged into chylomicron lipoprotein particles that eventually are secreted into the lymph.
  • DGAT diacylglycerol acyl transferase
  • DGAT diacylglycerol acyl transferase
  • two fatty acyl coenzyme A molecules are added to glycerol-3- phosphate to form phosphatidate.
  • DGAT lies at the final step of both triacylglycerol synthesis pathways.
  • DGATl and DGAT2 (Cases, S. et al., Proc. Natl Acad. ScL USA, 95:13018-13023 (1998)) (Oelkers, P. et al., J. Biol. Chem., 273:26765-26771 (1998)) (Cases, S. et al., J. Biol Chem., 276:38870-38876 (2001)).
  • DGATl and 2 are encoded by two different genes that bear little sequence homology. Functionally, these two enzymes might have different physiological importance in vivo.
  • DGATl knockout mice exhibit resistance towards becoming obese when challenged with a high fat (Smith, SJ.
  • DGAT2 knockout mice exhibit phenotypes such as lipopenia and skin barrier abnormalities, resulting in death soon after birth (Stone, SJ. et al., J. Biol Chem., 279:11767-11776 (2004)).
  • the compounds of the present invention are useful as DGAT modulators, and include compounds which are, for example, activators or inhibitors of DGAT enzyme. Accordingly, the compounds of the present invention may be useful for the treatment or prevention of diseases and disorders associated with DGAT enzyme activities. Preferably, compounds of the present invention possess activity as inhibitors of DGAT enzyme activities, and may be used in the treatment of diseases or disorders associated with the activity of the DGAT enzyme.
  • the compounds of the present invention or stereoisomers or pharmaceutically acceptable salts thereof can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to metabolic as well as conditions associated with metabolic disorders, (e.g.
  • the compounds of the present invention can possess both DGAT and ACAT inhibitory activities.
  • ACAT inhibition is a known mechanism to provide hypolipidemic effects (which also has anti-atherosclerosis activity) such as disclosed in, Drugs of the Future, 24:9-15 (1999) (Avasimibe); Nicolosi et al., "The ACAT inhibitor, Cl-1011 is effective in the prevention and regression of aortic fatty streak area in hamsters", Atherosclerosis (Shannon, Irel.), 137(1):77 ⁇ 85 (1998); Ghiselli, G., "The pharmacological profile of FCE 27677: a novel ACAT inhibitor with potent hypolipidemic activity mediated by selective suppression of the hepatic secretion of ApoBlOO-contaming lipoprotein", Cardiovasc.
  • the present invention relates to the use of a DGAT inhibitor in the treatment of appetitive or motivational disorders that regulate desires to consume sugars, carbohydrates, alcohol or drugs and more generally to regulate the consumption of ingredients with hedonic value.
  • appetitive disorders are understood as meaning: disorders associated with a substance and especially abuse of a substance and/or dependency on a substance, disorders of eating behaviors, especially those liable to cause excess weight, irrespective of its origin, for example: bulimia nervosa, craving for sugars.
  • the present invention further relates to the use of a DGAT inhibitor for the treatment of bulimia and obesity, including obesity associated with type II diabetes (non-insulin- dependent diabetes), or more generally any disease resulting in the patient becoming overweight.
  • Obesity as described herein, is defined by a body mass index (kg/m 2 ) of at least 26.
  • treating encompasses prevention, partial alleviation, or cure of the disease or disorder. Further, treatment of obesity is expected to prevent progression of medical covariants of obesity, such as arteriosclerosis.
  • Type II diabetes polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders.
  • Respiratory diseases for which DGAT modulators are useful include, but are not limited to, chronic pulmonary obstructive disorder, emphysema, asthma, and bronchitis.
  • DGAT modulators block the activation of lung epithelial cells by moieties such as allergic agents, inflammatory cytokines or smoke, thereby limiting release of mucin, cytokines, and chemokines, or selectively inhibiting lung epithelial cell activation.
  • DGAT is important in the regulation of TNF alpha of adipocytes.
  • Compounds of the present invention are especially of value, for example, in treating obesity associated inflammatory diseases such as arthritis or inflammatory bowel disease.
  • the present compounds are useful for treating autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses leading to kidney damage.
  • a method for treatment of a condition or disorder comprising administering to a patient in need thereof at least one compound of Formula (I) wherein said condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis.
  • Preferred compounds useful in the method of the present embodiment include compounds of Formula (I) wherein: [0063] A is hydrogen, Ci-C ⁇ alkyl, C 3 -C7cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R 6 , -C(O)OR 6 , or -C(O)NR 6 R 7 ; L is -(CR 1 R 2 V; n is 1 , 2, or 3; B is -OR 5 , -C(O)OR 5 , -OC(O)R 5 , or -OC(O)OR 5 ; R 1 and R 2 are, independently at each occurrence, hydrogen, Cj-C ⁇ alkyl, C 3 -C 7 CyClOaIkV
  • the patient is a human.
  • a therapeutically effective amount of the compound of Formula (I) or stereoisomer or a pharmaceutically acceptable salt thereof is administered in the method of this embodiment.
  • the use of a compound of Formula (I) in the manufacture of a medicament for the treatment of a condition or disorder is provided, wherein the condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis.
  • the condition or disorder is obesity.
  • the use of a compound of Formula (I) in the manufacture of a medicament for the treatment of a condition or disorder is provided, wherein the condition or disorder is treatable by modulation of the DGATl enzyme.
  • the modulation can be activation or inhibition of the DGATl enzyme.
  • the condition or disorder is treatable by inhibition of the DGATl enzyme.
  • Conditions or disorder treatable by the DGATl enzyme include, but are not limited to, obesity, dyslipidemia, diabetes, or atherosclerosis.
  • the condition or disorder treated by inhibition of the DGATl enzyme is obesity.
  • a method is provided for treating a condition or disorder in a patient wherein the condition or disorder is dependent upon DGATl inhibition, comprising administering to the patient a compound of Formula (I), The method of this embodiment can be used to treat conditions or disorders including obesity, dyslipidemia, diabetes, or atherosclerosis.
  • the condition or disorder is obesity.
  • a therapeutically effective amount of the compound of Formula (I) is administered in the method of this embodiment.
  • the present invention provides a compound of the present invention for use in therapy.
  • the present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of Formula (I) or a pharmaceutically acceptable salt thereof alone or in combination with a pharmaceutical carrier or diluent.
  • compounds of the present invention can be used alone, in combination with other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-obesity agents; anti-diabetic agents, appetite suppressants; agents used to treat eating disorders, cholesterol/lipid-lowering agents, HDL-raising agents, cognition enhancing agents, agents used to treat neurodegeneration, agents used to treat respiratory conditions, agents used to treat bowel disorders, anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; cardiac glycosides; and anti-tumor agents.
  • Such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the DGAT inhibitors in accordance with the invention.
  • Suitable anti-obesity agents for use in combination with the compounds of the present invention include melanocortin receptor (MC4R) agonists, melanin-concentrating hormone receptor (MCHR) antagonists, growth hormone secretagogue receptor (GHSR) antagonists, galanin receptor modulators, orexin antagonists, CCK agonists, GLP-I agonists, and other Pre-proglucagon-derived peptides; NPYl or NPY5 antagonist, NPY2 and NPY4 modulators, amylin receptor modulators, corticotropin releasing factor agonists, histamine receptor-3 (H3) modulators, aP2 inhibitors, PPAR gamma modulators, PPAR delta modulators, acetyl-CoA carboxylase (ACC) inhibitors, 11- ⁇ -HSD-l inhibitors, adinopectin receptor modulators; beta 3 adrenergic agonists, such as AJ9677 (Takeda/Dain
  • Suitable anti-diabetic agents for use in combination with the compounds of the present invention include: insulin secretagogues or insulin sensitizers, which may include biguanides, sulfonyl ureas, glucosidase inhibitors, aldose reductase inhibitors, PPAR ⁇ agonists such as thiazo ⁇ idinediones, PPAR ⁇ agonists (such as fibric acid derivatives), PPAR ⁇ antagonists or agonists, PPAR ot/ ⁇ dual agonists, 11- ⁇ -HSD-l inhibitors, dipeptidyl peptidase IV (DPP4) inhibitors, SGLT2 inhibitors, glucokinase inhibitors, AMP kinase modulators, glycogen phosphorylase inhibitors, and/or meglitinides, as well as insulin, and/or glucagon-like peptide- 1 (GLP-I), GLP-I agonist, and/or a PTP-
  • the antidiabetic agent may be an oral antihyperglycemic agent preferably a biguanide such as metformin or phenformin or salts thereof, preferably metformin HCl. Where the antidiabetic agent is a biguanide, the compounds of the present invention will be employed in a weight ratio to biguanide within the range from about 0.001 : 1 to about 10:1 , preferably from about 0.01 : 1 to about 5:1. [0072] The antidiabetic agent may also preferably be a sulfonyl urea such as glyburide (also known as glibenclamide), glimepiride (disclosed in U.S. Patent No.
  • the oral antidiabetic agent may also be a glucosidase inhibitor such as acarbose (disclosed in U.S. Patent No. 4,904,769) or miglitol (disclosed in U.S. Patent No. 4,639,436), which may be administered in the same or in a separate oral dosage forms.
  • the compounds of the present invention may be employed in combination with a PPAR ⁇ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect in NIDDM patients) such as rosiglitazone (SKB), pioglkazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No.
  • a PPAR ⁇ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect in NIDDM patients) such as rosiglitazone (SKB), pioglkazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No.
  • Glaxo-Wellcome's GL-262570 englitazone (CP-68722, Pfizer) or darglitazone (CP-86325, Pfizer, isaglitazone (MIT/J&J), JTT-501 (JPNT/P&U), L-895645 (Merck), R-119702 (Sankyo/WL), NN-2344 (Dr. Reddy/NN), or YM-440 (Yamanouchi), preferably rosiglitazone and pioglitazone.
  • the compounds of the present invention may be employed with a
  • PPAR ⁇ / ⁇ dual agonist such as MK-767/KRP-297 (Merck/Kyorin; as described in Yajirna, K. et al., Am. J. Physiol. Endocrinol. Metab., 284:E966 ⁇ E971 (2003)), AZ- 242 (tesaglitazar; Astra-Zeneca; as described in Ljung, B. et al., J. Lipid Res., 43:1855-1863 (2002)); muraglitazar; or the compounds described in U.S. Patent No. 6,414,002.
  • the compounds of the present invention may be employed in combination with anti-hyper lipidemia agents, or agents used to treat arteriosclerosis.
  • hypolipidemic agent would be an HMG CoA reductase inhibitor which includes, but is not limited to, mevastatin and related compounds as disclosed in U.S. Patent No. 3,983,140, lovastatin (mevinolin) and related compounds as disclosed in U.S. Patent No. 4,231,938, pravastatin and related compounds such as disclosed in U.S. Patent No. 4,346,227, simvastatin and related compounds as disclosed in U.S. Patent Nos. 4,448,784 and 4,450,171.
  • HMG CoA reductase inhibitors which may be employed herein include, but are not limited to, fluvastatin, disclosed in U.S. Patent No. 5,354,772, cerivastatin disclosed in U.S.
  • Patent Nos. 5,006,530 and 5, 177,080 atorvastatin disclosed in U.S. Patent Nos. 4,681,893, 5,273,995, 5,385,929 and 5,686,104, pitavastatin (Nissan/Sankyo's nisvastatin (NK- 104) or itavastatin), disclosed in U.S. Patent No. 5,011,930, Shionogi-Astra/Zeneca rosuvastatin (visastatin (ZD-4522)) disclosed in U.S. Patent No. 5,260,440, and related statin compounds disclosed in U.S. Patent No. 5,753,675, pyrazole analogs of mevalonolactone derivatives as disclosed in U.S. Patent No.
  • phosphinic acid compounds useful in inhibiting HMG CoA reductase suitable for use herein are disclosed in GB 2205837.
  • the squalene synthetase inhibitors suitable for use herein include, but are not limited to, ⁇ - ⁇ hos ⁇ hono-sulfonates disclosed in U.S. Patent No. 5,712,396, those disclosed by Biller et al., J. Med. Chem., 31:1869-1871 (1998) including isoprenoid (phosphinyl-methyl)phosphonates as well as other known squalene synthetase inhibitors, for example, as disclosed in U.S. Patent Nos.
  • squalene synthetase inhibitors suitable for use herein include the terpenoid pyrophosphates disclosed by Ortiz de Montellano, P. et al., J. Med. Chem., 20:243-249 (1977), the farnesyl diphosphate analog A and presqualene pyrophosphate (PSQ-PP) analogs as disclosed by Corey et al,, J. Am. Chem. Soc, 98:1291-1293 (1976), phosphinylphosphonates reported by McClard, R.
  • hypolipidemic agents suitable for use herein include, but are not limited to, fibric acid derivatives, such as fenofibrate, gemfibrozil, c ⁇ ofibrate, bezafibrate, ciprofibrate, clinofibrate and the like, probucol, and related compounds as disclosed in U.S. Patent No.
  • bile acid sequestrants such as cholestyramine, colestipol and DEAE-Sephadex (SECHOLEX®, policexide) and cholestagel (Sankyo/Geltex), as well as LIPOSTABIL® (Rhone-Poulenc), EISA!® E-5050 (an N-substituted ethanolamine derivative), imanixil (HOE-402), tetrahydrolipstatin (THL), istigmastanylphos- phorylcholine (SPC, Roche), aminocyclodextrin (Tanabe Seiyoku), Ajinomoto AJ- 814 (azulene derivative), melinamide (Sumitomo), Sandoz 58-035, American Cyanamid CL ⁇ 277,082 and CL-283,546 (disubstituted urea derivatives), nicotinic acid (niacin), aciprin, nicotinic acid (niacin
  • the hypolipidemic agent may be an upregulator of LDL receptor activity such as MD-700 (Taisho Pharmaceutical Co. Ltd) and LY295427 (Eli Lilly).
  • the hypolipidemic agent may be a cholesterol absorption inhibitor preferably Schering- Plough's SCH48461 (ezetimibe) as well as those disclosed in Atherosclerosis, 115:45-63 (1995) and/. Med. Chem., 41 :97% (1998).
  • the other lipid agent or lipid-modulating agent may be a cholesteryl transfer protein inhibitor (CETP) such as Pfizer's CP-529,414 as well as those disclosed in WO/0038722 and in EP 818448 (Bayer) and EP 992496, and Pharmacia's SC-744 and SC-795, as well as CETi-I and JTT-705.
  • CETP cholesteryl transfer protein inhibitor
  • the hypolipidemic agent may be an ileal NaVbile acid cotransporter inhibitor such as disclosed in Dings of the Future, 24:425-430 (1999).
  • the ATP citrate lyase inhibitor which may be employed in the combination of the invention may include, for example, those disclosed in U.S. Patent No. 5,447,954.
  • the other lipid agent also includes a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as an isoflavone such as genistein, daidzein, glycitein or equol, or phytosterols, phytostanol or tocotrienol as disclosed in WO 2000/015201; a beta-lactam cholesterol absorption inhibitor such as disclosed in EP 675714; an HDL upregulator such as an LXR agonist, a PPAR ⁇ -agonist and/or an FXR agonist; an LDL catabolism promoter such as disclosed in EP 1022272; a sodium-proton exchange inhibitor such as disclosed in DE 19622222; an LDL-receptor inducer or a steroidal glycoside such as disclosed in U.S.
  • a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as
  • Patent No. 5,698,527 and GB 2304106 an anti-oxidant such as beta-carotene, ascorbic acid, ⁇ -tocopherol or retinol as disclosed in WO 94/15592 as well as Vitamin C and an antihomocysteine agent such as folic acid, a folate, Vitamin B6, Vitamin B 12 and Vitamin E; isoniazid as disclosed in WO 97/35576; a cholesterol absorption inhibitor, an HMG-CoA synthase inhibitor, or a lanosterol demethylase inhibitor as disclosed in WO 97/48701; a PPAR ⁇ agonist for treating dyslipidemia; or a sterol regulating element binding protein-I (SREBP-I) as disclosed in WO 2000/050574, for example, a sphingolipid, such as ceramide, or neutral sphingomyelenase (N-SMase) or fragment thereof.
  • an anti-oxidant such as beta-carotene
  • Preferred hypolipidemic agents are pravastatin, lovastatin, simvastatin, atorvastatin, fiuvastatin, pitavastatin and rosuvastatin, as well as niacin and/or cholestagel.
  • the compounds of the present invention may be employed in combination with anti-hypertensive agents.
  • suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and/or T-type; e.g., diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g.
  • renin inhibitors ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril), AT-I receptor antagonists (e.g., losartan, irbesartan, valsartan), ET receptor antagonists (e.g.
  • Dual ET/AH antagonist e.g. , compounds disclosed in WO 00/01389
  • neutral endopeptidase (NEP) inhibitors neutral endopeptidase (NEP) inhibitors
  • vasopepsidase inhibitors dual NEP-ACE inhibitors
  • DGAT inhibitors could be useful in treating other diseases associated with obesity, including sleep disorders. Therefore, the compounds described in the present invention could be used in combination with therapeutics for treating sleep disorders.
  • Suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GABA receptor modulators; NMDA receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators.
  • melatonin analogs include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GABA receptor modulators; NMDA receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators.
  • suitable anti-inflammatory agents include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GABA receptor modulators; NMDA receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators.
  • Suitable antiinflammatory agents include prednisone, dexamethasone, cyclooxygenase inhibitors (Le., COX-I and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, naproxen, CELEBREX®, VIOXX®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CELLCEPT®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-I, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR 1384, including TNF-alpha inhibitors, such as tenidap, anti-TNF antibodies or soluble TNF receptor such as etanercept (ENBREL®), rap
  • j ZELNORM® and Maxi-K openers such as those disclosed in U.S. Patent No. 6,184,231 Bl).
  • the above other therapeutic agents when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art.
  • PDR Physicians' Desk Reference
  • Also embraced within this invention is a class of pharmaceutical compositions comprising the compound of Formula (I) or a stereoisomer or a pharmaceutically acceptable salt thereof in association having one or more non-toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier” materials) and, if desired, other active ingredients.
  • carrier non-toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants
  • the compounds of the present invention may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • the compounds and compositions of the present invention may, for example, be administered orally, mucosally, or parentally including intravascularly, intravenously, intraperitoneally, subcutaneously, intramuscularly intrasternally and infusion techniques, in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.
  • the pharmaceutical carrier may contain a mixture of mannitol or lactose and raicrocrystalline cellulose. The mixture may contain additional components such as a lubricating agent, e.g.
  • the pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals.
  • the compounds of the present invention can be administered to various mammalian species known to be subject to such maladies, e.g. , humans, in an effective amount up to 1 gram, preferably up to 200 mg, more preferably up to 100 mg in a regimen of single, two or four divided daily doses.
  • the compounds of the Formula (T) can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or inlrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or inlrasternal
  • the present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds can also be administered liposomally.
  • compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the compounds of the present invention can also be delivered through the oral cavity by sublingual and/or buccal administration.
  • Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used.
  • Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins.
  • fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins.
  • high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG).
  • Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g.
  • compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor.
  • compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • a suitable non-irritating excipient such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug.
  • exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene).
  • DJPEA diisopropylethylamine
  • HOBt 1-hydroxybenzotriazole
  • K 3 PO 4 potassium phosphate tribasic
  • NaHCO 3 sodium bicarbonate
  • Na 2 CO 3 sodium carbonate
  • Na 2 SO 4 sodium sulfate
  • Pd(dppf)Cl 2 [1,1 '-bis(diphenylphosphino)-ferrocene]dichloropalladium(II)
  • Compounds of the present invention may be prepared by procedures illustrated in the accompanying schemes.
  • the compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis.
  • the compounds of the present invention may be prepared by the methods described below, other with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art.
  • the reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformation being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformation proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention.
  • the compounds of the present invention can be prepared according to the general methods shown in the schemes below. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art.
  • PG is a suitable nitrogen protecting group, exemplified by benzyl, tert- butoxycarbonyl- [BOC], benzyloxycarbonyl- [CBZ], or 9-fiuorenylmethoxycarbonyl- [FMOC].
  • LG is a leaving group exemplified by halogen (Cl, Br, I) and sulfonates
  • OSO 2 -aryl e.g., -OSO 2 Ph or -OSO 2 PhCH 3
  • -OSO 2 -alkyl e.g., -OSO 2 CH 3 or -OSO 2 CF 3
  • Compounds of formula Ia' (with triazolopyridine core) can be prepared as described in Scheme 1.
  • Compounds of formula II are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula III can be prepared by reacting compounds of formula II with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature.
  • Compounds of formula V can be prepared by reacting compounds of formula III with a compound of formula IV in the presence of a coupling agent, such as N ⁇ [3 ⁇ (dimethylaminopropyl)-N'- ethylcarbodiimide.
  • Compounds of formula IV (where Z is an N-protecting group (PG) or a functional group (A) as defined in the claims) are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula VI can be prepared by reacting compounds of formula V with a dehydrating agent, such as a mixed solvent of ethanol and acetic acid at elevated temperature, or dichlorotriphenylphosphorane in the presence of a base, such as diisopropylethylamine.
  • a dehydrating agent such as a mixed solvent of ethanol and acetic acid at elevated temperature, or dichlorotriphenylphosphorane in the presence of a base, such as diisopropylethylamine.
  • Compounds of formula VII can be prepared by reacting compounds of formula VI with carbon monoxide and methanol in a pressured vessel at elevated temperature in the presence of a palladium catalyst and a ligand, such as palladium acetate and (i?)-(+)-l,2-bis(diphenylphosphino)propane.
  • Compounds of formula VIII can be prepared by saponification of compounds of formula VII in aqueous THF in the presence of a base, such as lithium hydroxide.
  • Compounds of formula X can be prepared by reaction of compounds of formula VIII with an amino acid ester IX' in the presence of a coupling agent, such as N-[3- (dimethylaminopropyl)-N'-ethylcarbodiimide.
  • a coupling agent such as N-[3- (dimethylaminopropyl)-N'-ethylcarbodiimide.
  • Compounds of formula XII where A is R 6 (CO) ⁇ can be prepared by acylation of compounds of formula XI with an acyl chloride or anhydride in the presence of a base, such as triethylamine.
  • Compounds of formula XII where A is R 6 O(CO)- can be prepared by reaction of compounds of formula XI with a chloroformate in the presence of a base, such as triethylamine.
  • Compounds of formula XII where A is R 6 N(CO)- can be prepared by reaction of compounds of formula XI with an isocyanate in the presence of a base, such as triethylamine.
  • Compounds of formula Ia' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula XII in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification, Scheme 2
  • compounds of formula Ia and Ia' can be prepared as described in Scheme 2 starting from compounds of formula XIlI, which are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula XIV can be prepared by reacting compounds of formula XIII with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature.
  • Compounds of formula XV can be prepared by reacting compounds of formula XIV with a compound of formula IV in the presence of a coupling agent, such as N-[3-(dimethylaminopropyl)-N'-ethylcarbodiimide, Compounds of formula XVI can be prepared by reaction of compounds of formula XV with a dehydrating agent, such phosphorus oxychloride at elevated, temperature, or dichlorotriphenylphosphorane in the presence of a base, such as diisopropylethylamine.
  • a dehydrating agent such phosphorus oxychloride at elevated, temperature, or dichlorotriphenylphosphorane
  • a base such as diisopropylethylamine.
  • Compounds of formula XVI where Z is an N-protecting group (PG) can be converted to compounds of formula XVII by the methods described in Greene, T.W.
  • Compounds of formula XVIII where A is R f '(CO)- can be prepared by acylation of compounds of formula XVII with an acyl chloride or anhydride in the presence of a base, such as triethylamine.
  • Compounds of formula XVIII where A is R 6 O(CO)- can be prepared by reaction of compounds of formula XVII with a chloroformate in the presence of a base, such as triethylamine.
  • Compounds of formula XVIII where A is R 6 N(CO)- can be prepared by reaction of compounds of formula XVII with an isocyanate in the presence of a base, such as triethylamine.
  • Compounds of formula XIX can be prepared by saponification of compounds of formula XVIII in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification.
  • Compounds of formula I can be prepared by reaction of compounds of formula XIX with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as JV- [3- (dimethylammopropyl)-N -ethylcarbodiimide.
  • Compounds of formula Ia' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula Ia (where B is -CO 2 R 5 and R s is an alkyl group) in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification.
  • a base such as lithium hydroxide
  • Compounds of formula Ic and Ic' can be prepared as described in Scheme 3.
  • Compounds of formula XX are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula XXI can be prepared by reacting compounds of formula XX with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature.
  • Compounds of formula XXII can be prepared by reacting compounds of formula XXI with a compound of formula IV in the presence of a coupling agent, such as N ⁇ [3- (dimethylaminopropyl)-N'-ethylcarbodiimide.
  • Compounds of formula ⁇ i can t ⁇ e prepared by reacting compounds of formula XXII with a dehydrating agent, such as phosphorus oxychloride at elevated temperature, or a phosphonium salt ⁇ e.g., triethylphosphine/carbon tetrachloride) in the presence of a base, such as diisopropylethylamine.
  • a dehydrating agent such as phosphorus oxychloride at elevated temperature
  • a phosphonium salt ⁇ e.g., triethylphosphine/carbon tetrachloride
  • Compounds of formula XXIV can be prepared by reacting compounds of formula XXIII with carbon monoxide and methanol in a pressured vessel at elevated temperature in the presence of a palladium catalyst and a ligand, such as palladium acetate and (i?)-(+)-l,2-bis(diphenylphosphino)propane.
  • a palladium catalyst and a ligand such as palladium acetate and (i?)-(+)-l,2-bis(diphenylphosphino)propane.
  • ⁇ where Z is an N-protecting group can be converted to compounds of formula XXV by the methods described in Greene, T. W. et al, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York (1991).
  • Compounds of formula ⁇ vi where A is R 6 (CO)- can be prepared by acylation of compounds of formula XXV with an acyl chloride or anhydride in the presence of a base, such as triethylamine.
  • Compounds of formula XXVI where A is R 0(CO)- can be prepared by reaction of compounds of formula XXV with a chloroformate in the presence of a base, such as triethylamine.
  • Compounds of formula XXVI where A is R 6 N(CO)- can be prepared by reaction of compounds of formula XXV with an isocyanate in the presence of a base, such as triethylamine.
  • Compounds of formula XXVII can be prepared by saponification of compounds of formula XXVI in aqueous THF in the presence of a base, such as lithium hydroxide.
  • Compounds of formula I can be prepared by reaction of compounds of formula XXVII with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as N-[3- (dimethylaminopropyl)-N'-ethylcarbodiimide.
  • Compounds of formula Ib and Ib' (with Iriazolopyridazine core) can be prepared as described in Scheme 4.
  • Compounds of formula XXVIII are either commercially available or available by means known to one skilled in the art.
  • Compounds of formula XXIX can be prepared by reacting compounds of formula ⁇ iu -vvitli anhydrous hydrazine in a polar solvent, such as ethanol at room temperature.
  • Compounds of formula xxx can be prepared by reacting compounds of formula XXIX with a compound of formula IV in the presence of a coupling agent, such as N-[3-(dimethylaniinopropyl)-N'- ethylcarbodiimide.
  • Compounds of formula XXXI can be prepared by reacting compounds of formula XXX with a dehydrating agent, such as triethylphosphine/carbon tetrachloride in the presence of a base (e.g., diisopropylethylamine).
  • a dehydrating agent such as triethylphosphine/carbon tetrachloride
  • a base e.g., diisopropylethylamine
  • Compounds of formula XXXI (where LG 2 is chlorine) can be converted to compounds of formula XXXII by dechlorination under hydrogenolysis conditions in the presence of a palladium catalyst.
  • Compounds of formula XXXI where Z is an N-protecting group can be converted to compounds of formula XXXIII by the methods described in Greene, T.W.
  • Compounds of formula XXXIV where A is R 6 N(CO)- can be prepared by reaction of compounds of formula XXXIII with an isocyanate in the presence of a base, such as triethylamine.
  • Compounds of formula XXXV can be prepared by treating compounds of formula XXXIV with trifluoroacetic acid.
  • Compounds of formula I can be prepared by reaction of compounds of formula XXXV with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as N-[3-(dimethylamino ⁇ ropyl)-N'-ethylcarbodiimide.
  • Compounds of formula Ib' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula Ib (where B is -COOR 5 and R 5 is an alkyl group) in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification.
  • a base such as lithium hydroxide
  • Method A Linear gradient of 10 to 100% solvent B over 2 min, with 1 min hold at 100% B;
  • Solvent A 0.1% trifluoroacetic acid, 10% water, 90 methanol.
  • Solvent B 0.1% trifluoroacetic acid, 90% methanol, 10% water.
  • Solvent A 0.1% trifluoroacetic acid, 90% water, 10% methanol.
  • Solvent B 0.1% trifluoroacetic acid, 90% methanol, 10% water.
  • Solvent A 0.1% trifluoroacetic acid, 90% water, 10% acetonitrile.
  • Solvent B 0.1% trifluoroacetic acid, 90% acetonitrile, 10% water.
  • Solvent A 1OmM ammonium acetate, 90% water, 10% acetonitrile.
  • Solvent B 1OmM ammonium acetate, 90% acetonitrile, 10% water.
  • Method E Linear gradient of 0 to 100% solvent B over 4 min, with 1 rnin hold at 100% B;
  • Solvent A 1OmM ammonium acetate, 90% water, 10% acetonitrile.
  • Solvent B 1OmM ammonium acetate, 90% acetonitrile, 10% water.
  • Solvent A 1OmM ammonium acetate, 90% water, 10% methanol.
  • Solvent B 1OmM ammonium acetate, 90% methanol, 10% water.
  • Carbon monoxide gas was charged into a pressure bottle containing a mixture of ID (300 mg, 0.77 mmol), Pd(OAc) 2 (69.2 mg, 0.31 mmol), dppp (127 mg, 0.31 mmol), Et 3 N (0.43 mL, 3.1 mmol), DMSO (5 mL) and MeOH (2.5 mL) until the pressure reached 30 psi.
  • the bottle was then sealed and heated at 80°C for 16 h.
  • Example 1 To a solution of Example 1 (50 mg, 0.14 mmol) in DMF (1 mL) was added L-valine ethyl ester hydrochloride salt (33 mg, 0.18 mmol), HOBt (28.4 mg, 0.21 mmol), EDC (40 mg, 0.21 mmol) and (JPr) 2 EtN (0.049 mL, 0.28 mmol). The resulting mixture was stirred at room temperature for 16 h, and then partitioned between water and EtOAc, The aqueous layer was extracted with EtOAc (2 x 20 mL), and the combined EtOAc extracts were washed with water, saturated aqueous NaCl, dried (Na 2 SO. ⁇ ) and concentrated under reduced pressure.
  • L-valine ethyl ester hydrochloride salt 33 mg, 0.18 mmol
  • HOBt 28.4 mg, 0.21 mmol
  • EDC 40 mg, 0.21 mmol
  • JPr 2 EtN
  • Example 2 [00123] To a solution of 2A (15 mg, 0.03 mmol) in THF (1 mL) was added 1 N aqueous NaOH solution (1 mL). The resulting mixture was stirred at room temperature for 16 h, and then acidified to pH 3-4 by dropwise addition of 1 N aqueous HCl. The mixture was extracted with EtOAc (3 x 10 mL), and the combined EtOAc extracts concentrated under reduced pressure. The crude product was purified using preparative HPLC (PHENOMENEX® Luna Axia, 5 ⁇ 30 x 100 mm using MeOH-H 2 O-TFA solvent system) to afford 8.2 mg (60%) of the title compound as a yellow solid.
  • preparative HPLC PHENOMENEX® Luna Axia, 5 ⁇ 30 x 100 mm using MeOH-H 2 O-TFA solvent system
  • Example 3 To a solution of 3B (105 mg, 0.185 mmol) in THF (2 mL) was added 2 N aqueous LiOH solution (2 mL). The resulting mixture was stirred at room temperature for 3 h. Analysis by HPLC indicated that the starting ester was consumed. The reaction mixture was cooled at 0°C, and acidified to pH 3 by dropwise addition of 1 N aqueous HCl. The mixture was extracted with EtOAc (2 x 20 mL) and the combined EtOAc extracts were concentrated under reduced pressure.
  • a suspension of 4B (13.4 g, 33.3 mmol) in POCl 3 (105 mL) was refluxed for 2.5 h. After cooling to room temperature, the reaction mixture was concentrated in vacuo to remove most of the POCI 3 . The residue was then dissolved in ethanol (100 mL), and slowly poured into water (350 mL) with stirring. After stirring for 30 min, the resulting mixture was concentrated in vacuo. To the residue was added 6N aqueous HCl (92 mL), and the mixture was heated at 6O°C for 3 h. After cooling to room temperature, the reaction mixture was neutralized with a NaOH aqueous solution to pH 5.
  • Example 4 A suspension of 4E (46 mg, 0.1 mmol), EDC (19.2 mg, 0.1 mmol) and HOBt (13.6 mg, 0.1 mmol) in anhydrous DCM (1 mL) was stirred at room temperature for 5 min, and then L- Valine methyl ester (12.5 mg, 0.095 mmol) was added. The mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was suspended in ethyl acetate (3 mL) and washed with saturated aqueous NaHCO 3 (1 x 0.8 mL), 1 N aqueous HCl (1 x 1 mL) and water (1 x 1 mL).
  • Examples 5 to 21 were prepared by coupling 3-(4-(3-(2 ⁇ (trifiuorornethoxy) phenyl) ureido)phenyl)-[l,2,4]triazolo[4,3- «]pyridine-8-carboxylic acid (4E) with various amino acid esters according to the general procedures described for Example 4 and listed in Table 1. Analytical data for the compounds in Table 1 are reported as follows: compound retention times were recorded using LC-MS conditions (Method E), and the molecular masses of the compounds were determined by MS (ES) by the formula m/z. TABLE 1
  • Example 23 [00139] A suspension of 23B (25.4 mg, 0.081 mmol), EDC (15.6 mg > 0.081 mmol) and HOBt (11 mg, 0.081 mmol) ) in anhydrous dichloromethane (1 mL) was stirred at room temperature for 5 min, and then L- valine methyl ester (11.8 mg, 0.09 mmol) was added. The resulting mixture was stirred at room temperature overnight and then concentrated in vacuo. The crude product was suspended in EtOAc (3 mL) and washed with saturated aqueous NaHCOj (1 x 0.8 mL), 1 N aqueous HCl (1 x 1 mL) and water (1 x 1 mL).
  • Example 25 [00145] A suspension of 3-(4-(6-chlorobenzo[ ⁇ /]thiazol-2-ylamino)phenyl)- [l,2,4]triazoIo[4,3- «]pyridme-8-carboxylic acid (33.7 mg, 0.08 tnmol), EDC (15.5 mg, 0.081 ramol) and HOBt (11 mg, 0.081 mmol) in a mixed solvent of DMF (0.5 niL) and CH 2 Cl 2 (0.5 mL) was stirred for 5 min.
  • Examples 26 to 43 were prepared by coupling 3-(4-(6- chlorobenzo[d r ]thiazol-2-y]amino)phenyl)-[l,2,4]triazolo[4 J 3- ⁇ ]pyridine-8-carboxylic acid (25D) with various amino acid esters according to the general procedure described in Example 25E and listed in Table 2. Analytical data for the compounds in Table 2 was reported as follows: compound retention times were recorded using LC- MS conditions (Method D), and the molecular mass of the compounds were determined by MS (ES) by the formula m/z.
  • Preparation 44A 4-(Thiazol-2-ylamino)benzoic acid [00147] To a suspension of tert-butyl 4-aminobenzoate (1.93 g, 10 mmol) and 2- bromothiazole (1.2 g, 7.3 mmol) in isopropanol (60 mL) was added 4 N HCl solution in 1,4-dioxane (0.5 mL, 2 mmol). The mixture was refluxed at 100°C for 50 h, and then allowed to cool to room temperature. The mixture was concentrated to one third of the initial volume. The resulting precipitate was isolated by filtration to yield the first crop of the title compound as a brown solid (915 mg). The mother liquor was dried in vacuo.
  • Example 44 A suspension of 3-(4-(thiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4,3- ⁇ ]pyridine-8-carboxylic acid (44D) (27 mg 5 0.08 mmol), EDC (15.5 mg ; 0.081 mraol) and HOBt (11 mg, 0.081 mmol) in a mixed solvent of DMF (0.5 rnL) and CH 2 Cl 2 (0.5 niL) was stirred for 5 min.
  • Examples 45 to 58 were prepared by coupling 3-(4-(thiazol-2- ylammo)phenyl)-[l,2,4]triazolo[4,3- ⁇ ]pyridine-8-carboxylic acid (44D) with various amino acid esters according to the procedures described in Example 44E and listed in Table 3. Analytical data for the compounds in Table 3 was reported as follows: compound retention times were recorded using LC-MS conditions (Method D), and the molecular mass of the compounds were determined by MS (ES) by the formula m/z. TABLE 3
  • Example 59 A suspension of 3-(4-(2-phenylacetamido)phenyl)-[l,2 > 4]triazolo[4 ! ,3- ⁇ ]pyridine-8-carboxylic acid (59A) (14 mg, 0.038 mmol), EDC (9 mg, 0.047 mmol) and HOBt (6.4 mg, 0.047 mmol) in a mixed solvent of DMF (0.5 mL) and DCM (0.5 mL) was stirred for 5 rain, and then L-valine tert-bntyl ester hydrochloride (8 mg, 0.038 mmol) and triethylamine (7.6 mg, 0.075 mmol) were added.
  • Carbon monoxide gas was charged into a pressure bottle containing a mixture of tert-butyl 4-(8-chloro-[1,2,4]triazolo[4,3- ⁇ ]pyrazin-3-yl)phenylcarbamate (61C) (714 mg, 2.065 mmol), Pd(OAc) 2 (185 mg, 0.826 mmol), dppp (341 mg, 0.826 mmol), Et 3 N (1.15 mL, 8.26 mmol), DMSO (12 mL) and MeOH (6 mL) until the pressure reached to 30 psi. The bottle was then sealed and heated at 75°C for 4 h.
  • Example 61 A suspension of 3-(4-(3-(2-(trifluoromethoxy)phenyl)ureido)phenyl)- [1,2,4] triazolo[4,3- ⁇ ]pyrazine ⁇ 8-carboxylic acid (61G) (35 mg, 0.076 mmol), EDC (15.4 mg, 0.08 mmol) and HOBt (11 mg, 0.08 mmol) in a mixed solvent of DMF (0.5 niL) and CH 2 CI 2 (1 raL) was stirred until it became a clear solution, and then L-valine tert-b ⁇ tyl ester hydrochloride salt (16.8 mg, 0.08 mmol) and triethylamine (16 mg, 0.16 mmol) were added.
  • 61G 3-(4-(3-(2-(trifluoromethoxy)phenyl)ureido)phenyl)- [1,2,4] triazolo[4,3- ⁇ ]pyrazine ⁇ 8-car
  • Example 63 A suspension of 3-(4-(6-chlorobenzo[W]thiazol-2-ylamino)phenyI)- [l,2,4]triazolo [4,3- ⁇ ]pyridine-8-carboxylic acid (63E) (60 mg, 0.14 mmol), EDC (27 mg, 0,14 mmol) and HOBt (19 mg, 0.14 mmol) in a mixed solvent of DMF (1 mL) and DCM (1 mL) was stirred until it became a clear solution, and then L-valine tert- butyl ester hydrochloride salt (30 mg, 0.12 mmol) and triethylamine (29 mg, 0.28 mmol) were added.
  • 6-chIoropyridazine ⁇ 4 ⁇ carboxylate To a solution of tert-butyl ⁇ -ch ⁇ oro-S-hydrazinylpyridazine ⁇ -carboxylate (66B) (800 mg, 3.27 mmol) in anhydrous DMF (5 mL) was added A-(tert- butoxycarbonyl- amine)benzoic acid (LO g, 4.25 mmol), EDC (940 mg, 4.91 mmol), HOBt (663mg, 4.91 mmol), followed by DIPEA (1.1 mL, 6.54 mmol). The reaction mixture was stirred at room temperature overnight.
  • A-(tert- butoxycarbonyl- amine)benzoic acid LO g, 4.25 mmol
  • EDC 940 mg, 4.91 mmol
  • HOBt 663mg, 4.91 mmol
  • DIPEA 1.1 mL, 6.54 mmol
  • DGATl enzyme assays were conducted using membranes isolated from Sf9 insect cells expressing the recombinant human DGATl cDNA. The assays were conducted in 384-well plates with total volume of 25 ⁇ l at 25°C. In each assay, 300 ng of recombinant human DGATl membrane was incubated with 25 ⁇ M of 2- monooleoylglycerol and 25 ⁇ M of [ 3 H]-stearoyl-CoA in 100 mM potassium phosphate (pH 7.4) for 30 min with various concentrations of compounds delivered in DMSO.
  • the assay was terminated by the addition of 30 ⁇ l of Stopping Solution [50 mM HEPES, 5 mg/mL Yittrium Oxide (YOX) Polylysine SPA beads, 3.33 mg/mL Fraction V BSA, 200 ⁇ M Mercuric chloride].
  • the signal was measured using LEADSEBKER SM for 5 minutes.
  • the zero level of enzyme activity was defined by the above assay using Sf9 cell membrane uninfected with baculovirus and the 100% level of DGATl enzyme activity was defined by human DGATl assay with the vehicle DMSO.
  • the IC 50 values of inhibitors were determined by Excel-fit. [00187] Compounds described herein were tested in the DGATl enzyme assay described immediately above. The following results were obtained.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Diabetes (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Child & Adolescent Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed are triazolopyridine compounds of Formula (I), including pharmaceutically acceptable salts thereof. Also, disclosed are methods of using the compound in the treatment of obesity, dyslipidemia, diabetes and atherosclerosis, and to pharmaceutical compositions comprising at least one compound of Formula (I) or a stereoisomer or pharmaceutically acceptable salt thereof.

Description

TRIAZOLO COMPOUNDS USEFUL AS DGATl INHIBITORS
BACKGROUND [0001] In mammals, there are two biochemical pathways for the synthesis of triacylglycerol: the monoacyl glycerol pathway, which occurs exclusively in the small intestine (Lehner, R. et al., Prog. Lipid Res,, 35:169-201 (1996)), and the glycerophosphate pathway, which takes place ubiquitously but most notably in the liver and in adipose tissue (Bell, R.M. et al., Annu. Rev. Biochem., 49:459-487 (1980)). The monoacylglycerol pathway initiates from acyl coenzyme A:monoacylglycerol acyltransferase (MGAT) (EC 2.3.1.22). Within minutes of its appearance from the digestion of dietary fat in the lumen of the small intestine, 2-monoacylglycerol is acylated by MGAT to form diacylglycerol. Diacylglycerol is further acylated by acyl coenzyme A: diacylglycerol acyltransferase (DGAT) (EC 2.3.1.20) to re-synthesize triacylglycerol, which is packaged into chylomicron lipoprotein particles that eventually are secreted into the lymph. In the glycerol-3-phosphate pathway, two fatty acyl coenzyme A molecules are added to glycerol-3-phosphate to form phosphatidate. These reactions are followed by the removal of the phosphate group by phosphatidate phosphohydrolase to generate diacylglycerol. Diacylglycerol is then further acylated by DGAT to form triacylglycerol. Collectively, DGAT lies at the final step of both triacylglycerol synthesis pathways. [0002] Two DGAT enzymes have been identified and have been designated as DGATl and DGAT2 (Cases, S. et al., Proc. Natl Acad. ScL USA, 95: 13018-13023 (1998)) (Oelkers, P. et al., J. Biol. Chem., 273:267 '65-2671 'I (1998)) (Cases, S. et al., J. Biol. Chem., 276:38870-38876 (2001)). Although they cany out identical enzymatic reactions, DGATl and DGAT2 are encoded by two different genes that bear little sequence homology. Functionally, these two enzymes might have different physiological importance in vivo. DGATl knockout mice exhibit resistance towards becoming obese when challenged with a high fat (Smith, SJ. et al., Nat. Genet., 25:87-90 (2000)). They are physically more active, possess a higher metabolic rate (Chen, H.C. et al., Trends Cardiovasc. Med., 10: 188-192 (2000)) and appear to have greater insulin sensitivity (Chen, H.C. et al., J. Clin. Invest, 109:1049-1055 (2002)). In contrast, DGAT2 knockout mice exhibit phenotypes such as Hpopenia and skin barrier abnormalities, resulting in death soon after birth (Stone, SJ. et al., J. Biol. Chem., 279: 1176741776 (2004)). [0003] U.S. Patent 7,300,932 B2 discloses fused bicyclic nitrogen-containing heterocyclic compounds that are useful for treating or preventing conditions and disorders associated with DGAT. As may be appreciated, there still remains a need for new compounds that are inhibitors of DGAT and are useful for the treatment of DGAT related conditions and disorders. [0004] Applicants have found triazolopyridine compounds that have activity as inhibitors of DGAT, in particular DGATl, and are thereby useful in therapy.
SUMMARY OF THE INVENTION [0005] The present invention is directed to compounds according to Formula (I):
Figure imgf000003_0001
including stereoisomers and pharmaceutically acceptable salts thereof, wherein: A is hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7;
L is -(CR1R2V; n is 1, 2, or 3;
B is -OR5, -C(O)OR5, -OC(O)R5, or -OC(O)OR3; one of X and Y is CH or N, and the other of X and Y is CH;
R1 and R2 are, independently at each occurrence, hydrogen, alkyl, cycloalkyl, aryl, and/or heterocyclyl, or one R1 and one R2 form a Cs-Cγcycloalkyl or 4- to 7- membered heterocyclyl ring having one or two heteroatoms;
R3 is hydrogen or alkyl, or R3 and R1 form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms;
R4 is hydrogen or alkyl;
R5 is hydrogen or alkyl;
R6 is alkyl, cycloalkyl, aryl, or heterocyclyl; and R7 is hydrogen or alkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein: each of said alkyl is substituted with 0-3 Ra; each of said cycloalkyl is substituted with 0-3 Ra; each of said aryl is substituted with 0-4 R ; each of said heterocyclyl is substituted with 0-4 Rb;
Ra is, independently at each occurrence, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, phenyl, imidazolyl, and/or C1- Qalkoxy;
Rb is, independently at each occurrence, Q^alkyl, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, and/or C,-C3alkoxy; and
Rc and Rd are, independently at each occurrence, H and/or CrC4alkyl, or Rc and Rd together with the nitrogen atom to which they are attached, form a 4- to 7- membered heterocyclyl ring with one or two heteroatoms.
[0006] Also described is a pharmaceutical composition comprising a compound of Formula (I) and a pharmaceutically acceptable carrier or diluent. [0007] Further described is method for treating a condition or disorder comprising administering to a patient in need thereof at least one compound of Formula (I); wherein said condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis.
DETAILED DESCRIPTION [0008] Listed below are definitions of various terms used to describe the present invention. These definitions apply to the terms as they are used throughout the specification (unless they are otherwise limited in specific instances) either individually or as part of a larger group. [0009] The term "alkyl" and "alk" refer to a straight or branched chain alkane (hydrocarbon) radical containing from 1 to 12 carbon atoms, preferably from 1 to 6 carbon atoms, and more preferably from 1 to 4 carbon atoms. Exemplary "alkyl" and/or "alk" groups include, but are not limited to, for example, methyl, ethyl, propyl, butyl, pentyl, hexyl, isohexyl, heptyl, octyl, nonyl, decyl, and dodecyl. [0010] The term "lower alkyl" refers to an "alkyl" and/or "alk" group containing from 1 to 4 carbon atoms and preferably from 1 to 2 carbon atoms. When a subscript is used with reference to an alkyl or other group, the subscript refers to the number of carbon atoms the group may contain. For example, the term "Co-C4alkyl" includes a bond and an alkyl group containing 1 to 4 carbon atoms, and the term "CrC4alkyl" refers to alkyl groups containing 1 to 4 carbon atoms. Exemplary lower alkyl groups include, but are not limited to, for example, methyl, ethyl, propyl including n-propyl and isopropyl, and butyl including n-butyl, isobutyl, and t-butyl. [0011 ] The "alkyl" and/or "aϊk" group can be optionally substituted with one or more substituents, preferably 1 to 3 substituents, at any available and substitutable position. Exemplary substituents include halogen (e.g., a single halo substituent or multiple halo substituents form, in the latter case, groups such as, for example, a perfluoroalkyl group or an alkyl group bearing -CCI3 or -CF3), hydroxyl, -NH2, -NH(alkyl), -CF3, -N(alkyl)2, cyano, Ci-C3alkoxy group, phenyl, imidazolyl, and halogenated alkoxy group such as -OCF3. [0012] The term "cycloalkyl" refers to a fully saturated hydrocarbon group containing from 1 to 4 rings and 3 to 8 carbon atoms per ring. Exemplary cycloalkyl groups include, but at not limited to, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. The cycloalkyl group can be optionally substituted with one or more substituents, preferably 1 to 3 substituents, at any available and substitutable point of attachment. Exemplary substituents include those groups recited for substituted alkyl. [0013] The term "aryl" refers to cyclic aromatic hydrocarbon groups having from 1 to 2 aromatic rings, such as, for example, phenyl, biphenyl, or naphthyl. When the aryl group contains two aromatic rings (e.g., bicyclic, etc.), the aromatic rings may be joined at a single point (e.g., biphenyl) or fused (e.g., naphthyl and phenanthrenyl). The aryl group can be optionally substituted with one or more substituents, preferably 1 to 4 substituents, at any available and substitutable ring position, or where valence allows on any rings fused or attached thereto. Exemplary substituents include alkyl and those groups recited for substituted alkyl. {0014] The term "heterocyclo", "heterocycle", "heterocyclyl" or "heterocyclic ring", as used herein, represents 3-, 4-, 5~, 6-, or 7-membered monocyclic or polycyclic or 7-, 8-, 9-, 10-, 11-, 12-, 13-, or 14-membered polycyclic heterocyclic ring that is saturated, partially unsaturated, or fully unsaturated, and that contains carbon atoms and i , 2, 3, or 4 heteroatoms independently selected from the group consisting of N, O, and S; and including any polycyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring. The nitrogen and sulfur heteroatoms may optionally be oxidized to -NO-, -SO-, or -SO2-. The heterocyclic ring may be attached to its pendant group at any heteroatora or carbon atom which results in the creation of a stable structure. The heterocycle, heterocyclic, or heterocyclo group can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl. When the term "heterocycle" is used, it is intended to include heteroaryl. The heterocyclo, heterocycle, heterocyclic, or heterocyclo group can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl. [0015] Examples of heterocycles include, but are not limited to, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzthiazolyϊ, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazalonyl, carbazolyl, 4aH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyϊ, decahydroquinolinyl, 2H,6H~l,5,2~dithiazinyl, dihyα!roraro[2,3-6]tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, imidazolopyridinyl, indolenyl, indolinyl, indolizinyl, indolyl, 3Η-indolyl, isatinoyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindoiyl, isoquinolinyl, isothiazolyl, isothiazolopyridinyl, isoxazolyl, isoxazolopyridinyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyI, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolopyridinyl, oxazolidinylperimidinyl, oxindolyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolopyridinyl, pyrazolyl, pyridazinyl, pyridooxazolyl, pyridoimidazolyl, pyridothiazolyl, pyridinyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2-pyrrolidonyl, 2H~pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrazolyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H- 1,2,5 -thiadiazinyl, 1,2,3-thiadiazolyl, 1,2,4-thiadiazolyI, 1,2,5-thiadiazolyl, 1,3,4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thiazolopyridinyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1 ,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, and xanthenyl. Also included are fused ring and spiro compounds containing, for example, the above heterocycles. [0016] Examples of 5- to 10-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl, imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, moφholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, triazolyi, benzimidazolyl, lH-indazoIyl, benzofuranyl, benzothiofuranyl, benztetrazolyl, benzotriazolyl, benzisoxazolyl, benzoxazolyl, oxindolyl, benzoxazolinyl, benzthiazolyl, benzisothiazolyl, isatinoyl, isoquinolinyl, octahydroisoquinolinyl , tetrahydroisoquinol inyl, tetrahydroquinolinyl , isoxazolopyridinyl, quinazolinyl, quinolinyl, isothiazolopyridinyl, thiazolopyridinyl, oxazolopyridinyl, imidazolopyridinyl, and pyrazolopyridinyl, [0017] Examples of 5- to 6-membered heterocycles include, but are not limited to, pyridinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, pyrazinyl, piperazinyl, piperidinyl , imidazolyl, imidazolidinyl, indolyl, tetrazolyl, isoxazolyl, morpholinyl, oxazolyl, oxadiazolyl, oxazolidinyl, tetrahydrofuranyl, thiadiazinyl, thiadiazolyl, thiazolyl, triazinyl, and triazolyi. [0018] As used herein, the terra "aromatic heterocyclic group" or "heteroaryl" is intended to mean fully unsaturated heterocyclyl rings, including monocyclic and polycyclic aromatic hydrocarbons having at least one heteroatom ring member such as sulfur, oxygen, or nitrogen. Heteroaryl groups include, without limitation, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, furyl, quinolyl, isoquinolyl, thienyl, imidazolyl, thiazolyl, indolyl, pyrrolyl, oxazolyl, benzofuryl, benzothienyl, benzthiazolyl, isoxazolyl, pyrazolyl, triazolyi, tetrazolyl indazolyl, indolmyl, 1,2,4- thiadiazolyl, isothiazolyl, purinyl, carbazolyl, benzimidazolyl, benzodioxolanyl, and benzodioxane. Heteroaryl groups are unsubstituted or substituted. Heteroaryl groups can be substituted at any available point of attachment with at least one substituent, preferably 1 to 4 substituents, selected from alkyl and those recited for substituted alkyl. [0019] The term "alkoxy" as employed herein alone or as part of another group includes an alkyl as defined above linked through an oxygen atom. [0020] The term "halogen" or "halo" as used herein alone or as part of another group refers to fluorine, chlorine, bromine, and iodine, with fluorine, chlorine, and bromine being preferred. [0021] The term "cyano," as used herein, refers to a -CN group. [0022] The term "methylene," as used herein, refers to a -CH2- group. [0023] The compounds of the present invention can be present as salts, which are also within the scope of this invention. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred. If the compounds of the present invention have, for example, at least one basic center, they can form acid addition salts. These are formed, for example, with strong inorganic acids, such as mineral acids, for example sulfuric acid, phosphoric acid or a hydrohalic acid, with organic carboxylic acids, such as alkanecarboxylic acids of 1 to 4 carbon atoms, for example acetic acid, which are unsubstituted or substituted, for example, by halogen as chloroacetic acid, such as saturated or unsaturated dicarboxylic acids, for example oxalic, malonic, succinic, nialeic, fumaric, phthalic or terephthalic acid, such as hydroxycarboxylic acids, for example ascorbic, glycolic, lactic, malic, tartaric or citric acid, such as amino acids, (for example aspartic or glutamic acid or lysine or arginine), or benzoic acid, or with organic sulfonic acids, such as (Ci-C4) alkyl or arylsulfonic acids which are unsubstituted or substituted, for example by halogen, for example methyl- or/?-toluene- sulfonic acid. Corresponding acid addition salts can also be formed having, if desired, an additionally present basic center. The compounds of Formula (ϊ) having at least one acid group (for example COOH) can also form salts with bases. Suitable salts with bases are, for example, metal salts, such as alkali metal or alkaline earth metal salts, for example sodium, potassium or magnesium salts, or salts with ammonia or an organic amine, such as morpholine, thiornorpholine, piperidine, pyrrolidine, a mono-, di-, or tri-lower alkylamine, for example ethyl, tert-butyl, diethyl, diisopropyl, triethyl, tributyl or dimethyl- propylamine, or a mono, di or trihydroxy lower alkylamine, for example mono, di or triethanolamine. Corresponding internal salts may furthermore be formed. Salts which are unsuitable for pharmaceutical uses but which can be employed, for example, for the isolation or purification of free compounds of Formula (I) or their pharmaceutically acceptable salts, are also included. [0024] Preferred salts of the compounds of Formula (I) which contain a basic group include monohydrochloride, hydrogensulfate, methanesulfonate, phosphate, nitrate or acetate. [0025] Preferred salts of the compounds of Formula (I) which contain an acid group include sodium, potassium and magnesium salts and pharmaceutically acceptable organic amines, [0026] The term "modulator" refers to a chemical compound with capacity to either enhance (e.g. , "agonist" activity) or partially enhance (e.g. , "partial agonist" activity) or inhibit (e.g., "antagonist" activity or "inverse agonist" activity) a functional property of biological activity or process (e.g., enzyme activity or receptor binding); such enhancement or inhibition may be contingent on the occurrence of a specific event, such as activation of a signal transduction pathway, and/or may be manifest only in particular cell types. [0027] The term "bioactive metabolite" as employed herein refers to any functional group contained in a compound of Formula (I) with an open valence for further substitution wherein such substitution can, upon biotransformation, generate a compound of Formula (I). Examples of such functional groups of bioactive metabolites include, but are not limited to, -OH, -NH2, or functional groups wherein the hydrogen can be replaced with a functional group such as -PO3H2 for example, which, upon biotransformation generates an -OH or -NH2 functional group of a compound of Formula (I). [0028] The term "prodrug" as employed herein includes functionalization of bioactive amine- or hydroxyl-containing compounds of Formula (I) to form alkyl-, acyl-, sulfonyl-, phosphoryl-, or carbohydrate-substituted derivatives. Such derivatives are formed by reacting compounds of Formula (I) with alkylating-, acylating-, sulfonylating-, or phosphorylating reagents employing procedures known to those skilled in the art. Alkyiation of amines of Formula (I) may result in, but is not limited to, derivatives that include spacer units to other prodrug moieties such as substituted alkyoxymethyl-, acyloxymethyl-, phosphoryloxymethyl-, or sulfonyloxymethyl- groups. Alkyiation of amines of Formula (I) may result in the generation of quarternary amine salts that act in vivo to provide the bioactive agent (i.e., the compound of Formula (I)). [0029] Preferred prodrugs consist of a compound of Formula (I) where a pendant hydroxyl is phosphorylated to generate a phosphate derivative. Such a prodrug may also include a spacer group between the compound of Formula (I) and the phosphate group, such as a methyleneoxy-group. Methods to generate such a prodrug from a compound of Formula (I) are known to those skilled in the art, and are listed in the references below. [0030] Preferred prodrugs also consist of a compound of Formula (I) where a pendant amine, such as a pyridine group, is alkylated with a group, such as methyl, to form a quarternary ammonium ion salt. Methods to generate such a prodrug from a compound of Formula (I) are known to those skilled in the art, and are listed in the references below. [0031 ] Any compound that can be converted in vivo to provide the bioactive agent (i.e., the compound of Formula (I)) is a prodrug within the scope and spirit of the invention. [0032] Various forms of prodrugs are well known in the art. A comprehensive description of prodrugs and prodrug derivatives are described in: a) The Practice of Medicinal Chemistry t Camille G. Wermuth et al., Ch. 31 (Academic Press, 1996); b) Design of Prodrugs, edited by H. Bundgaard (Elsevier, 1985); c) A Textbook of Drug Design and Development, P. Krogsgaard-Larson and H. Bundgaard, eds., Ch. 5, pp. 113-191 (Harwood Academic Publishers, 1991); d) Hydrolysis in Drug and Prodrug Metabolism, B. Testa and J.M. Mayer (Verlag Helvetica Chimica Acta AG, Zurich, Switzerland; Wiley- VCH, Weinheim,
Federal Republic of Germany, 2003); e) Ettmayer, P. et al., "Lessons Learned from Marketed and Investigational Prodrugs", J. Med. Chem., 47(10):2393-2404 (2004); and f) Davidsen, S.K.. et al,, "N-(Acyloxyalkyl)pyridinram Salts as Soluble Prodrugs of a Potent Platelet Activating Factor Antagonist", J. Med. Chem., 37(26):4423-4429 (1994).
[0033] The term "patient" as used herein encompasses all mammalian species including humans, cows, horses, dogs, and cats; and preferably, humans. [0034] The term "therapeutically effective" is intended to qualify the amount of each agent, which will treat or prevent a condition treatable by administration of a composition of the invention. That amount is the amount sufficient to exhibit a detectable therapeutic or preventative or ameliorative effect. The effect may include, for example, treatment or prevention of the conditions listed herein. The precise effective amount for a subject will depend upon the subject's size and health, the nature and extent of the condition being treated, recommendations of the treating physician, and the therapeutics or combination of therapeutics selected for administration. [0035] All stereoisomers of the compounds of the instant invention are contemplated, either in mixture or in pure or substantially pure form. The compounds of the present invention can have asymmetric centers at any of the carbon atoms including any one of the R substituents. Consequently, compounds of the present invention can exist in enantiomeric or diastereomeric forms or in mixtures thereof. The processes for preparation can utilize racemates, enantioraers or diastereoraers as starting materials. When diastereomeric or enantiomeric products are prepared, they can be separated by conventional methods for example, chromatographic techniques, chiral HPLC or fractional crystallization. [0036] In one embodiment, compounds of Formula (I) are provided where X is CH and Y is CH. The compounds of this embodiment have structures represented by Formula (Ia):
Figure imgf000012_0001
and stereoisomers or salts thereof, wherein: A, L, B, R3, and R4 are as defined hereinabove. The compounds of Formula (Ia) are triazolopyridine derivatives. [0037] In one embodiment, compounds of Formula (I) are provided where X is N and Y is CH. The compounds of this embodiment have structures represented by Formula (Ib):
Figure imgf000012_0002
including stereoisomers or salts thereof, wherein: A, L, Bf R3, and R4 are as defined hereinabove. The compounds of Formula (Ib) are triazolopyridazine derivatives, [0038] In one embodiment, compounds of Formula (I) are provided where X is CH and Y is N. The compounds of this embodiment have structures represented by ' Formula (Ic):
Figure imgf000012_0003
Including stereoisomers or salts thereof, wherein: A, L, B, R3, and R4 are as defined hereinabove. The compounds of Formula (Ic) are triazolopyrazine derivatives. [0039] In one embodiment, compounds of Formula (I) are provided wherein L is - (CR1R2)!,-, n is 1 or 2, and R1 and R2 are as defined hereinabove. Examples of suitable L groups include, but are not limited to, -CR1R2- in which n is 1 and -CHR!- in which n is 1 and R2 is hydrogen. [0040] In one embodiment, compounds of Formula (I) are provided wherein RJ and R2 are, independently at each occurrence, hydrogen, C1-Ce alkyl, C3-C7 cycloalkyl, aryl, and/or 1- or 2-ring heterocyclyl; and each alkyl is substituted with 0-3 Ra, each cycloalkyl is substituted with 0-3 Ra, each aryl is substituted with 0-4 Rb, and each heteroaryl is substituted with 0-4 Rb; and Ra and Rb are as defined hereinabove. Preferably, R1 is hydrogen, Q-Qalkyl, C3~C7cycloalkyl, or phenyl; and R2 is hydrogen or methyl. More preferably, R! is hydrogen, Q^alkyl, Cs-Cvcycloakyl, or phenyl; and R2 is hydrogen. For example, the present embodiment provides compounds of Formula (I) wherein R1 is hydrogen, C]-C4alkyl, hydroxyethyl, cyclohexyl, phenyl, or methyl substituted with phenyl, hydroxyphenyl or imidazolyl; and R2 is hydrogen. [0041] In one embodiment, compounds of Formula (I) are provided wherein one R1 and one R2 form a C3-C7cycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein said cycloalkyl ring is substituted with 0-3 Ra and said heterocyclyl ring is substituted with 0-4 Rb, and Ra and Rb are as defined hereinabove. Preferably, R1 and R2 form a Cs-Cγcycloalkyl, and more preferably, R1 and R2 form a C5-C7cycloalkyl. For example, this embodiment provides compounds of Formula (I) wherein R1 and R2 form a cyclohexyl group. [0042] In one embodiment, compounds of Formula (I) are provided wherein n is 2 and L is -CR1 R2CR1 R2-, and R1 attached to one carbon atom of L and R2 attached to the other carbon atom of L, form a C3-C7CyClOaIkVl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein said cycloalkyl ring is substituted with 0-3 Ra and said heterocyclyl ring is substituted with 0-4 Rb, and Ra and Rb are as defined hereinabove. In this embodiment, preferably L is -CHR1CHR2-. [0043] In one embodiment, compounds of Formula (I) are provided wherein R3 is hydrogen or Ci-Cβalkyl, wherein each alkyl is substituted with 0-3 Ra and Ra is as defined hereinabove. Preferably, R3 is hydrogen or Ci-C4alkyl, and more preferably hydrogen and Q-Cialkyl. Examples of suitable R3 groups include, but are not limited to, hydrogen and methyl. Still more preferably, R3 is hydrogen. [0044] In one embodiment, compounds of Formula (I) are provided wherein R3 and R1 form a 4~ to 7-membered heterocyclyl ring having one or two heteroatoms, wherein the heterocyclyl ring is substituted with 0-4 Rb. Examples of suitable heteroatoms include nitrogen, oxygen, and sulfur. The heterocyclyl ring is saturated or partially unsaturated. Preferably, the heterocyclyl ring is saturated. Examples of suitable heterocyclyl rings include 5- to 7-membered heterocyclyl rings such as imidazolyl, pyrrol idinyl, and piperiditiyl. [0045] In one embodiment, compounds of Formula (ϊ) are provided wherein R4 is hydrogen or Cj-Cealkyl, wherein each alkyl is substituted with 0-3 Ra and Ra is as defined hereinabove. Preferably, R4 is hydrogen or Ci-C4EIkVl, and more preferably hydrogen and
Figure imgf000014_0001
Examples of suitable R4 groups include, but are not limited to, hydrogen and methyl. Still more preferably, R4 is hydrogen, [0046] In one embodiment, compounds of Formula (I) are provided wherein B is -OR5, -C(O)OR5, -OC(O)R5, or -OC(O)OR5; R5 is hydrogen or Ci-Qalkyl; and wherein said alkyl is substituted with 0-3 Ra; and Ra is defined hereinabove. Preferably, B is -OR5 or -C(O)OR5; and more preferably, B is -OH, -C(O)OH, or -C(O)OCH2CH3. Preferably, R5 is hydrogen or Ci-C4alkyl. For example, this embodiment provides compounds of Formula (I) wherein B is -OH, -OCH3, -C(O)OH, -C(O)OCH3, and -C(O)OCH2CH3. [0047] In one embodiment, compounds of Formula (I) are provided wherein R6 is Ci-C&alkyl, CrCγcycloalkyl, aryl, or 1- or 2-ring heterocyclyl; and said alkyl is substituted with 0-3 Ra, said cycloalkyl is substituted with 0-4 Rb, said aryl is substituted with 0-4 R , and said heterocyclyl is substituted with 0-4 Rb; and Ra and R are as defined hereinabove. [0048] In one embodiment, compounds of Formula (I) or are provided wherein A is hydrogen, Cj-Cealkyl, C3-C7cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7; R6 is alkyl, cycloalkyl, aryl, or heterocyclyl; and R7 is hydrogen or alkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; and each of said alkyl is substituted with 0-3 Ra; each of said cycloalkyl is substituted with 0-3 Ra; each of said aryl is substituted with 0-4 Rb; and each of said heterocyclyl is substituted with 0-4 Rb; and Ra and Rb are as defined hereinabove. For example, the present embodiment provides compounds of Formula (I) in which A is a 1- or 2-ring heterocyclyl having 1 or 2 heteroatoms selected from nitrogen and sulfur. [0049] In one embodiment, compounds of Formula (I) are provided wherein: A is hydrogen, Q-Cealkyl, C3-C7cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7; R1 and R2 are, independently at each occurrence, hydrogen, CrQalkyl, Cj-C7cycloalkyl, aryl, and/or 1- or 2-ring heterocyclyl, or one R1 and one R2 form a Q-Cγcycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; R3 is hydrogen or Q-Cgalkyl, or R3 and R1 form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; R4 is hydrogen or Ci-Cgalkyl; R5 is hydrogen or d
Figure imgf000015_0001
-C^Ikyl; R6 is C3-C7cycloalkyl, aryl, or 1- or 2-ring heterocyclyl; R7 is hydrogen or Cj-Cealkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein: each alkyl is substituted with 0-3 Ra; each cycloalkyl is substituted with 0-3 Ra; each aryl is substituted with 0-4 Rb; and each heterocyclyl is substituted with 0-4 Rb. [0050] In one embodiment, compounds of Formula (I) are provided wherein: A is hydrogen, C]-C4alkyl, phenyl, 1- or 2-ring heterocyclyl having 1- or 2-heteroatoms selected from S and N, -C(O)R6, -C(O)OR6, or -C(O)NR6R7; R1 is hydrogen, Ci-Cgalkyl, CyC^cycloalkyl, or phenyl; R is hydrogen or methyl; or R and R form a Cs-Cycycloalkyl ring or 5- to ό-membered heterocyclyl ring having one or two heteroatoms; R3 is hydrogen or Ci-C4alkyl, or R3 and R1 form a 5- to 7-membered heterocyclyl ring having one or two heteroatoms; R4 is hydrogen or C|-C4alkyl; R5 is hydrogen or Ci-C4alkyl; R6 is Q^aϊkyl, or phenyl; and R7 is hydrogen or methyl; wherein: each alkyl is substituted with 0-3 Ra; each cycloalkyl is substituted with 0-3 Ra; each phenyl is substituted with 0-3 Rb; and each heterocyclyl is substituted with 0-3 Rb. [0051] In one embodiment, compounds of Formula (I) are provided wherein: A is hydrogen, -C(O)R6, -C(O)OR6, -C(O)NHR6, thiazolyl, or benzothiazolyl; B is -OH or -C(O)OR5; R1 is hydrogen, CrC4alkyl, C5-C7cycloalkyl, or phenyl; R2 is hydrogen; or R1 and R2 form a Cs-Qcycloalkyl; R3 is hydrogen or Q-Caalkyl, or R3 and R1 form a 5- to 6-merabered heterocyclyl ring having one heteroatom; R4 is hydrogen or Cj-Cϊalkyl; R5 is hydrogen or
Figure imgf000015_0002
and n is 1 or 2; and each alkyl is substituted with 0-2 Ra; each cycloalkyl is substituted with 0-2 Ra; each heterocyclyl is substituted with 0-2 Rb; each phenyl is substituted with 0-2 Rb; each thiazolyl is substituted with 0-2 Rb; and each benzothiazolyl is substituted with 0-2 Rb; Ra is, independently at each occurrence, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, phenyl, and/or imidazolyl; and Rb is, independently at each occurrence, Q-G÷alkyl, F5 CI, Br, -CF3, -OH, -OCH3, -OCF3, and/or -CN. [0052] In one embodiment, compounds of Formula (I) are provided wherein: A is hydrogen, -C(O)-CH2CH3, -C(O)-benzyl, -C(O)O-CH3, -C(O)O-(butyl), -C(O)NH- (trifluoromethylphenyl), -C(O)NH-(trifluoromethoxyphenyl), thiazolyl, or chlorobenzothiazolyl; B is -OH, -C(O)OH, or -C(O)OCH2CH3; R1 is hydrogen, Ci-C4alkyl, hydroxyelhyl, cyclohexyl, phenyl, or methyl substituted with phenyl, hydroxyphenyl, or imidazolyl; R2 is hydrogen; or R1 and R2 form a cyclopentyl ring; R3 is hydrogen, or R3 and R1 form a pyrrolidinyl ring; and R4 is hydrogen. [0053] In one embodiment, a compound of Formula (I) or a salt thereof is provided, wherein said compound is:
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
Figure imgf000027_0001
[0054] In mammals, there are two biochemical pathways for the synthesis of triacylglycerol: the monoacylglycerol pathway, which happens exclusively in the small intestine (tehner, R. et al.s Prog. Lipid Res,, 35:169-201 (1996)), and the glycerol-3 -phosphate pathway, which takes place ubiquitously but most notably in the liver and in adipose tissue (Bell, R.M. et al., Annu. Rev. Biochem,, 49:459-487 (1980)). The monoacylglycerol pathway initiates from acyl coenzyme A: monoacylglycerol acyl transferase (MGAT) (EC 2.3.1.22). Within minutes of its appearance from the digestion of dietary fat in the lumen of the small intestine, 2- monoacyl glycerol is acylated by MGAT to form diacylglycerol. Diacylglycerol is further acylated by acyl coenzyme A: diacylglycerol acyl transferase (DGAT) (EC 2.3.1.20) to re-synthesize triacylglycerol, which is packaged into chylomicron lipoprotein particles that eventually are secreted into the lymph. In the gIycerol-3- phosphate pathway, two fatty acyl coenzyme A molecules are added to glycerol-3- phosphate to form phosphatidate. These reactions are followed by the removal of the phosphate group by phosphatidate phosphohydrolase to generate diacylglycerol. Diacylglycerol is then further acylated by DGAT to form triacylglycerol. Collectively, DGAT lies at the final step of both triacylglycerol synthesis pathways. [0055] Two DGAT enzymes have been identified, which are designated as
DGATl and DGAT2 (Cases, S. et al., Proc. Natl Acad. ScL USA, 95:13018-13023 (1998)) (Oelkers, P. et al., J. Biol. Chem., 273:26765-26771 (1998)) (Cases, S. et al., J. Biol Chem., 276:38870-38876 (2001)). Although they carry out identical enzymatic reactions, DGATl and 2 are encoded by two different genes that bear little sequence homology. Functionally, these two enzymes might have different physiological importance in vivo. DGATl knockout mice exhibit resistance towards becoming obese when challenged with a high fat (Smith, SJ. et al,, Nat Genet, 25:87-90 (2000)). They are physically more active, possess a higher metabolic rate (Chen, H.C. et al., Trends Cardiovasc. Med., 10:188-192 (2000)) and appear to have greater insulin sensitivity (Chen, H.C. et al., J. Clin. Invest, 109: 1049-1055 (2002)). In contrast, DGAT2 knockout mice exhibit phenotypes such as lipopenia and skin barrier abnormalities, resulting in death soon after birth (Stone, SJ. et al., J. Biol Chem., 279:11767-11776 (2004)). [0056] The compounds of the present invention are useful as DGAT modulators, and include compounds which are, for example, activators or inhibitors of DGAT enzyme. Accordingly, the compounds of the present invention may be useful for the treatment or prevention of diseases and disorders associated with DGAT enzyme activities. Preferably, compounds of the present invention possess activity as inhibitors of DGAT enzyme activities, and may be used in the treatment of diseases or disorders associated with the activity of the DGAT enzyme. [0057] The compounds of the present invention or stereoisomers or pharmaceutically acceptable salts thereof can be administered to mammals, preferably humans, for the treatment of a variety of conditions and disorders, including, but not limited to metabolic as well as conditions associated with metabolic disorders, (e.g. , obesity, diabetes, arteriosclerosis, hypertension, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders, hyperlipidemic conditions, bulimia nervosa and compulsive eating disorders). These compounds could also be used for treatment of cataboHsm in connection with pulmonary dysfunction and ventilator dependency; treatment of cardiac dysfunction (e.g. , associated with valvular disease, myocardial infarction, cardiac hypertrophy or congestive heart failure); and improvement of the overall pulmonary function; transplant rejection; rheumatoid arthritis; multiple sclerosis; inflammatory bowel disease; lupus; graft vs. host disease; T-cell mediated hypersensitivity disease; psoriasis; asthma; Hashimoto's thyroiditis; Guillain-Barre syndrome; cancer; contact dermatitis; allergic rhinitis; and ischemic or reperfusion injury. [0058] The compounds of the present invention can possess both DGAT and ACAT inhibitory activities. ACAT inhibition is a known mechanism to provide hypolipidemic effects (which also has anti-atherosclerosis activity) such as disclosed in, Drugs of the Future, 24:9-15 (1999) (Avasimibe); Nicolosi et al., "The ACAT inhibitor, Cl-1011 is effective in the prevention and regression of aortic fatty streak area in hamsters", Atherosclerosis (Shannon, Irel.), 137(1):77~85 (1998); Ghiselli, G., "The pharmacological profile of FCE 27677: a novel ACAT inhibitor with potent hypolipidemic activity mediated by selective suppression of the hepatic secretion of ApoBlOO-contaming lipoprotein", Cardiovasc. DrugRev., 16(l): 16-30 (1998); Smith, C. et al., "RP 73163: a bioavailable alkylsulfmyl-diphenylimidazole ACAT inhibitor", Bioorg. Med. Chem. Lett, 6(l):47-50 (1996); Krause, B.R. et al., Chapter 6: "ACAT Inhibitors: Physiologic Mechanisms for Hypolipidemic and Anti- Atherosclerotic Activities in Experimental Animals", Inflammation: Mediators and Pathways, CRC Press, Inc., publ., Ruffolo, Jr., R.R. et al., eds., pp. 173-198 (1995); Sliskovic et al., "ACAT inhibitors: potential anti-atherosclerotic agents", Curr. Med. Chem., l(3):204-225 (1994); Stout et al., "Inhibitors of acyl-CoA:cholesterol O-acyl transferase (ACAT) as hypocholesterolemic agents. 6. The first water-soluble ACAT inhibitor with lipid-regulating activity. Inhibitors of acyl-CoA:cholesterol acyl transferase (ACAT). 7. Development of a series of substituted N-phenyI-N'-[(l- phenylcyclopentyl)-methyϊ]ureas with enhanced hypocholesterolemic activity", Chemtracts: Org. Chem., 8(6):359-362 (1995), or TS-962 (Taisho Pharmaceutical Co. Ltd.), as well as F-1394, CS-505, F-12511, HL-004, K-10085 and YIC-C8-434. [0059] The present invention relates to the use of a DGAT inhibitor in the treatment of appetitive or motivational disorders that regulate desires to consume sugars, carbohydrates, alcohol or drugs and more generally to regulate the consumption of ingredients with hedonic value. In the present description and in the claims, appetitive disorders are understood as meaning: disorders associated with a substance and especially abuse of a substance and/or dependency on a substance, disorders of eating behaviors, especially those liable to cause excess weight, irrespective of its origin, for example: bulimia nervosa, craving for sugars. The present invention further relates to the use of a DGAT inhibitor for the treatment of bulimia and obesity, including obesity associated with type II diabetes (non-insulin- dependent diabetes), or more generally any disease resulting in the patient becoming overweight. Obesity, as described herein, is defined by a body mass index (kg/m2) of at least 26. It may be due to any cause, whether genetic or environmental, including overeating and bulimia, polycystic ovary disease, craniopharyngeoma, Prader-Willi Syndrome, Frohlich's Syndrome, Type II diabetes, growth hormone deficiency, Turner's Syndrome and other pathological slates characterized by reduced metabolic activity or reduced energy expenditure. As used with reference to the utilities described herein, the term "treating" or "treatment" encompasses prevention, partial alleviation, or cure of the disease or disorder. Further, treatment of obesity is expected to prevent progression of medical covariants of obesity, such as arteriosclerosis. Type II diabetes, polycystic ovary disease, cardiovascular disease, osteoarthritis, dermatological disorders, hypertension, insulin resistance, hypercholesterolemia, hypertriglyceridemia, cholelithiasis and sleep disorders. [0060] As modulators of the DGAT enzyme, the compounds of the present invention are further useful for the treatment and prevention of respiratory diseases and disorders. Respiratory diseases for which DGAT modulators are useful include, but are not limited to, chronic pulmonary obstructive disorder, emphysema, asthma, and bronchitis. In addition, DGAT modulators block the activation of lung epithelial cells by moieties such as allergic agents, inflammatory cytokines or smoke, thereby limiting release of mucin, cytokines, and chemokines, or selectively inhibiting lung epithelial cell activation. [0061] DGAT is important in the regulation of TNF alpha of adipocytes. Compounds of the present invention are especially of value, for example, in treating obesity associated inflammatory diseases such as arthritis or inflammatory bowel disease. In particular, the present compounds are useful for treating autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses leading to kidney damage. [0062] In one embodiment, a method is provided for treatment of a condition or disorder comprising administering to a patient in need thereof at least one compound of Formula (I) wherein said condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis. Preferred compounds useful in the method of the present embodiment include compounds of Formula (I) wherein: [0063] A is hydrogen, Ci-Cβalkyl, C3-C7cycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7; L is -(CR1R2V; n is 1 , 2, or 3; B is -OR5, -C(O)OR5, -OC(O)R5, or -OC(O)OR5; R1 and R2 are, independently at each occurrence, hydrogen, Cj-Cβalkyl, C3-C7CyClOaIkVl, aryl, and/or 1- or 2-ring heterocyclyl, or one R1 and one R2 form a C3-C7cycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms; R3 is hydrogen or Ci-Cgalkyl, or R3 and R1 form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; R4 is hydrogen or CrQalkyl; R5 is hydrogen or Q-Cgalkyl; R6 is CrQaϊkyl, C3-C7cycloalkyl, aryl, or 1- or 2-ring heterocyclyl; R7 is hydrogen or Cj-Qalkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7- membered heterocyclyl ring having one or two heteroatoms; wherein: each alkyl is substituted with 0-3 Ra; each cycloalkyl is substituted with 0-3 Ra; each aryl is substituted with 0-4 Rb; and each heterocyclyl is substituted with 0-4 Rb; Ra is, independently at each occurrence, F, Cl, Br5 -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, phenyl, imidazolyl, and/or Ci-C3alkoxy; Rb is, independently at each occurrence, C1-C4JtIlCyI, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, and/or d-C3alkoxy; and R° and Rd are, independently at each occurrence, H and/or
Figure imgf000031_0001
Preferably, the method of this embodiment is used to treat obesity. Preferably, the patient is a human. Preferably, a therapeutically effective amount of the compound of Formula (I) or stereoisomer or a pharmaceutically acceptable salt thereof is administered in the method of this embodiment. [0064] In one embodiment, the use of a compound of Formula (I) in the manufacture of a medicament for the treatment of a condition or disorder is provided, wherein the condition or disorder is obesity, dyslipidemia, diabetes, or atherosclerosis. Preferably, the condition or disorder is obesity. [0065] In one embodiment, the use of a compound of Formula (I) in the manufacture of a medicament for the treatment of a condition or disorder is provided, wherein the condition or disorder is treatable by modulation of the DGATl enzyme. The modulation can be activation or inhibition of the DGATl enzyme. Preferably, the condition or disorder is treatable by inhibition of the DGATl enzyme. Conditions or disorder treatable by the DGATl enzyme include, but are not limited to, obesity, dyslipidemia, diabetes, or atherosclerosis. Preferably, the condition or disorder treated by inhibition of the DGATl enzyme is obesity. [0066] In one embodiment, a method is provided for treating a condition or disorder in a patient wherein the condition or disorder is dependent upon DGATl inhibition, comprising administering to the patient a compound of Formula (I), The method of this embodiment can be used to treat conditions or disorders including obesity, dyslipidemia, diabetes, or atherosclerosis. Preferably, the condition or disorder is obesity. Preferably, a therapeutically effective amount of the compound of Formula (I) is administered in the method of this embodiment. [0067] In one embodiment, the present invention provides a compound of the present invention for use in therapy. [0068] The present invention includes within its scope pharmaceutical compositions comprising, as an active ingredient, a therapeutically effective amount of at least one of the compounds of Formula (I) or a pharmaceutically acceptable salt thereof alone or in combination with a pharmaceutical carrier or diluent. Optionally, compounds of the present invention can be used alone, in combination with other suitable therapeutic agents useful in the treatment of the aforementioned disorders including: anti-obesity agents; anti-diabetic agents, appetite suppressants; agents used to treat eating disorders, cholesterol/lipid-lowering agents, HDL-raising agents, cognition enhancing agents, agents used to treat neurodegeneration, agents used to treat respiratory conditions, agents used to treat bowel disorders, anti-inflammatory agents; anti-anxiety agents; anti-depressants; anti-hypertensive agents; cardiac glycosides; and anti-tumor agents. Such other therapeutic agent(s) may be administered prior to, simultaneously with, or following the administration of the DGAT inhibitors in accordance with the invention. [0069] Examples of suitable anti-obesity agents for use in combination with the compounds of the present invention include melanocortin receptor (MC4R) agonists, melanin-concentrating hormone receptor (MCHR) antagonists, growth hormone secretagogue receptor (GHSR) antagonists, galanin receptor modulators, orexin antagonists, CCK agonists, GLP-I agonists, and other Pre-proglucagon-derived peptides; NPYl or NPY5 antagonist, NPY2 and NPY4 modulators, amylin receptor modulators, corticotropin releasing factor agonists, histamine receptor-3 (H3) modulators, aP2 inhibitors, PPAR gamma modulators, PPAR delta modulators, acetyl-CoA carboxylase (ACC) inhibitors, 11-β-HSD-l inhibitors, adinopectin receptor modulators; beta 3 adrenergic agonists, such as AJ9677 (Takeda/Dainippon), L750355 (Merck), or CP331648 (Pfizer) or other known beta 3 agonists as disclosed in U.S. Patent Nos. 5,541,204, 5,770,615, 5,491,134, 5,776,983 and 5,488,064, a thyroid receptor beta modulator, such as a thyroid receptor ligand as disclosed in WO 97/21993 (U. CaI SF), WO 99/00353 (KaroBio), a lipase inhibitor, such as orlistat or ATL-962 (Alizyme), serotonin receptor agonists, (e.g., BVT-933 (Biovitrum)), monoamine reuptake inhibitors or releasing agents, such as fenfluramine, dexfenflxtramine, fluvoxamine, fluoxetine, paroxetine, sertraline, chlorphentermine, cloforex, clortermine, picilorex, sibutramine, dexamphetamine, phentermine, phenylpropanolamine or mazindol, anorectic agents such as topiramate (Johnson & Johnson), CNTF (ciliary neurotrophic factor)/AXOKJNE® (Regeneron), BDNF (brain-derived neurotrophic factor), leptin and leptin receptor modulators, or cannabinoid- 1 (CBl) receptor antagonists, such as SR-141716 (Sanofi), MK-0364 (Merck), CP-945,598 (Pfizer) or SLV-319 (Solvay). [0070] Examples of suitable anti-diabetic agents for use in combination with the compounds of the present invention include: insulin secretagogues or insulin sensitizers, which may include biguanides, sulfonyl ureas, glucosidase inhibitors, aldose reductase inhibitors, PPAR γ agonists such as thiazoϊidinediones, PPAR α agonists (such as fibric acid derivatives), PPAR δ antagonists or agonists, PPAR ot/γ dual agonists, 11-β-HSD-l inhibitors, dipeptidyl peptidase IV (DPP4) inhibitors, SGLT2 inhibitors, glucokinase inhibitors, AMP kinase modulators, glycogen phosphorylase inhibitors, and/or meglitinides, as well as insulin, and/or glucagon-like peptide- 1 (GLP-I), GLP-I agonist, and/or a PTP-IB inhibitor (protein tyrosine phosphatase- IB inhibitor). [0071] The antidiabetic agent may be an oral antihyperglycemic agent preferably a biguanide such as metformin or phenformin or salts thereof, preferably metformin HCl. Where the antidiabetic agent is a biguanide, the compounds of the present invention will be employed in a weight ratio to biguanide within the range from about 0.001 : 1 to about 10:1 , preferably from about 0.01 : 1 to about 5:1. [0072] The antidiabetic agent may also preferably be a sulfonyl urea such as glyburide (also known as glibenclamide), glimepiride (disclosed in U.S. Patent No. 4,379,785), glipizide, gliclazide or chlorpropamide, other known sulfonylureas or other antihyperglycemic agents which act on the ATP-dependent channel of the beta- cells, with glyburide and glipizide being preferred, which may be administered in the same or in separate oral dosage forms. The oral antidiabetic agent may also be a glucosidase inhibitor such as acarbose (disclosed in U.S. Patent No. 4,904,769) or miglitol (disclosed in U.S. Patent No. 4,639,436), which may be administered in the same or in a separate oral dosage forms. [0073] The compounds of the present invention may be employed in combination with a PPAR γ agonist such as a thiazolidinedione oral anti-diabetic agent or other insulin sensitizers (which has an insulin sensitivity effect in NIDDM patients) such as rosiglitazone (SKB), pioglkazone (Takeda), Mitsubishi's MCC-555 (disclosed in U.S. Patent No. 5,594,016), Glaxo-Wellcome's GL-262570, englitazone (CP-68722, Pfizer) or darglitazone (CP-86325, Pfizer, isaglitazone (MIT/J&J), JTT-501 (JPNT/P&U), L-895645 (Merck), R-119702 (Sankyo/WL), NN-2344 (Dr. Reddy/NN), or YM-440 (Yamanouchi), preferably rosiglitazone and pioglitazone. [0074] The compounds of the present invention may be employed with a
PPARα/γ dual agonist such as MK-767/KRP-297 (Merck/Kyorin; as described in Yajirna, K. et al., Am. J. Physiol. Endocrinol. Metab., 284:E966~E971 (2003)), AZ- 242 (tesaglitazar; Astra-Zeneca; as described in Ljung, B. et al., J. Lipid Res., 43:1855-1863 (2002)); muraglitazar; or the compounds described in U.S. Patent No. 6,414,002. [0075] The compounds of the present invention may be employed in combination with anti-hyper lipidemia agents, or agents used to treat arteriosclerosis. An example of an hypolipidemic agent would be an HMG CoA reductase inhibitor which includes, but is not limited to, mevastatin and related compounds as disclosed in U.S. Patent No. 3,983,140, lovastatin (mevinolin) and related compounds as disclosed in U.S. Patent No. 4,231,938, pravastatin and related compounds such as disclosed in U.S. Patent No. 4,346,227, simvastatin and related compounds as disclosed in U.S. Patent Nos. 4,448,784 and 4,450,171. Other HMG CoA reductase inhibitors which may be employed herein include, but are not limited to, fluvastatin, disclosed in U.S. Patent No. 5,354,772, cerivastatin disclosed in U.S. Patent Nos. 5,006,530 and 5, 177,080, atorvastatin disclosed in U.S. Patent Nos. 4,681,893, 5,273,995, 5,385,929 and 5,686,104, pitavastatin (Nissan/Sankyo's nisvastatin (NK- 104) or itavastatin), disclosed in U.S. Patent No. 5,011,930, Shionogi-Astra/Zeneca rosuvastatin (visastatin (ZD-4522)) disclosed in U.S. Patent No. 5,260,440, and related statin compounds disclosed in U.S. Patent No. 5,753,675, pyrazole analogs of mevalonolactone derivatives as disclosed in U.S. Patent No. 4,613,610, indene analogs of mevalonolactone derivatives as disclosed in PCT application WO 86/03488, 6-[2-(substituted-pyrrol-l-yl)-alkyl)pyran-2-ones and derivatives thereof as disclosed in U.S. Patent No. 4,647,576, Searle's SC-45355 (a 3-substituted pentanedioic acid derivative) dichloroacetate, imidazole analogs of mevalonolactone as disclosed in PCT application WO 86/07054, 3-carboxy-2-hydroxy-propane- phosphonic acid derivatives as disclosed in French Patent No. 2,596,393, 2,3- disubstituted pyrrole, furan and thiophene derivatives as disclosed in European Patent Application No. 0221025, naphthyl analogs of mevalonolactone as disclosed in U.S. Patent No. 4,686,237, octahydronaphthalenes such as disclosed in U.S. Patent No. 4,499,289, keto analogs of mevinolin (lovastatin) as disclosed in European Patent Application No.0,142, 146 A2, and quinoline and pyridine derivatives disclosed in U.S. Patent Nos. 5,506,219 and 5,691,322. In addition, phosphinic acid compounds useful in inhibiting HMG CoA reductase suitable for use herein are disclosed in GB 2205837. [0076] The squalene synthetase inhibitors suitable for use herein include, but are not limited to, α-ρhosρhono-sulfonates disclosed in U.S. Patent No. 5,712,396, those disclosed by Biller et al., J. Med. Chem., 31:1869-1871 (1998) including isoprenoid (phosphinyl-methyl)phosphonates as well as other known squalene synthetase inhibitors, for example, as disclosed in U.S. Patent Nos. 4,871,721 and 4,924,024 and in Biller, S.A. et al., Current Pharmaceutical Design, 2: 1 -40 (1996). [0077] In addition, other squalene synthetase inhibitors suitable for use herein include the terpenoid pyrophosphates disclosed by Ortiz de Montellano, P. et al., J. Med. Chem., 20:243-249 (1977), the farnesyl diphosphate analog A and presqualene pyrophosphate (PSQ-PP) analogs as disclosed by Corey et al,, J. Am. Chem. Soc, 98:1291-1293 (1976), phosphinylphosphonates reported by McClard, R. W1 et al., /. Am. Chem. Soc, 109:5544 (1987) and cyclopropanes reported by Capson, T.L., PhD dissertation, June, 1987, Dept. Med. Chem. U of Utah, Abstract, Table of Contents, pp. 16, 17, 40-43, 48-51, Summary. [0078] Other hypolipidemic agents suitable for use herein include, but are not limited to, fibric acid derivatives, such as fenofibrate, gemfibrozil, cϊofibrate, bezafibrate, ciprofibrate, clinofibrate and the like, probucol, and related compounds as disclosed in U.S. Patent No. 3,674,836, probucol and gemfibrozil being preferred, bile acid sequestrants such as cholestyramine, colestipol and DEAE-Sephadex (SECHOLEX®, policexide) and cholestagel (Sankyo/Geltex), as well as LIPOSTABIL® (Rhone-Poulenc), EISA!® E-5050 (an N-substituted ethanolamine derivative), imanixil (HOE-402), tetrahydrolipstatin (THL), istigmastanylphos- phorylcholine (SPC, Roche), aminocyclodextrin (Tanabe Seiyoku), Ajinomoto AJ- 814 (azulene derivative), melinamide (Sumitomo), Sandoz 58-035, American Cyanamid CL~277,082 and CL-283,546 (disubstituted urea derivatives), nicotinic acid (niacin), acipimox, acifran, neomycin, p-aminosalicylic acid, aspirin, poly(diallylmethylamine) derivatives such as disclosed in U.S. Patent No. 4,759,923, quaternary amine poly(diallyldimethylammonium chloride) and ionenes such as disclosed in U.S. Patent No. 4,027,009, and other known serum cholesterol lowering agents. [0079] The hypolipidemic agent may be an upregulator of LDL receptor activity such as MD-700 (Taisho Pharmaceutical Co. Ltd) and LY295427 (Eli Lilly). The hypolipidemic agent may be a cholesterol absorption inhibitor preferably Schering- Plough's SCH48461 (ezetimibe) as well as those disclosed in Atherosclerosis, 115:45-63 (1995) and/. Med. Chem., 41 :97% (1998). [0080] The other lipid agent or lipid-modulating agent may be a cholesteryl transfer protein inhibitor (CETP) such as Pfizer's CP-529,414 as well as those disclosed in WO/0038722 and in EP 818448 (Bayer) and EP 992496, and Pharmacia's SC-744 and SC-795, as well as CETi-I and JTT-705. [0081] The hypolipidemic agent may be an ileal NaVbile acid cotransporter inhibitor such as disclosed in Dings of the Future, 24:425-430 (1999). The ATP citrate lyase inhibitor which may be employed in the combination of the invention may include, for example, those disclosed in U.S. Patent No. 5,447,954. [0082] The other lipid agent also includes a phytoestrogen compound such as disclosed in WO 00/30665 including isolated soy bean protein, soy protein concentrate or soy flour as well as an isoflavone such as genistein, daidzein, glycitein or equol, or phytosterols, phytostanol or tocotrienol as disclosed in WO 2000/015201; a beta-lactam cholesterol absorption inhibitor such as disclosed in EP 675714; an HDL upregulator such as an LXR agonist, a PPAR α-agonist and/or an FXR agonist; an LDL catabolism promoter such as disclosed in EP 1022272; a sodium-proton exchange inhibitor such as disclosed in DE 19622222; an LDL-receptor inducer or a steroidal glycoside such as disclosed in U.S. Patent No. 5,698,527 and GB 2304106; an anti-oxidant such as beta-carotene, ascorbic acid, α-tocopherol or retinol as disclosed in WO 94/15592 as well as Vitamin C and an antihomocysteine agent such as folic acid, a folate, Vitamin B6, Vitamin B 12 and Vitamin E; isoniazid as disclosed in WO 97/35576; a cholesterol absorption inhibitor, an HMG-CoA synthase inhibitor, or a lanosterol demethylase inhibitor as disclosed in WO 97/48701; a PPAR δ agonist for treating dyslipidemia; or a sterol regulating element binding protein-I (SREBP-I) as disclosed in WO 2000/050574, for example, a sphingolipid, such as ceramide, or neutral sphingomyelenase (N-SMase) or fragment thereof. Preferred hypolipidemic agents are pravastatin, lovastatin, simvastatin, atorvastatin, fiuvastatin, pitavastatin and rosuvastatin, as well as niacin and/or cholestagel. [0083] The compounds of the present invention may be employed in combination with anti-hypertensive agents. Examples of suitable anti-hypertensive agents for use in combination with the compounds of the present invention include beta adrenergic blockers, calcium channel blockers (L-type and/or T-type; e.g., diltiazem, verapamil, nifedipine, amlodipine and mybefradil), diuretics (e.g.y chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, hydroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzthiazide, ethacrynic acid tricrynafen, chlorthalidone, furosemide, musolimine, bumetanide, triamtrenene, amiloride, spironolactone), renin inhibitors, ACE inhibitors (e.g., captopril, zofenopril, fosinopril, enalapril, ceranopril, cilazopril, delapril, pentopril, quinapril, ramipril, lisinopril), AT-I receptor antagonists (e.g., losartan, irbesartan, valsartan), ET receptor antagonists (e.g., sitaxsentan, atrsentan and compounds disclosed in U.S. Patent Nos. 5,612,359 and 6,043,265), Dual ET/AH antagonist (e.g. , compounds disclosed in WO 00/01389), neutral endopeptidase (NEP) inhibitors, vasopepsidase inhibitors (dual NEP-ACE inhibitors) (e.g., omapatriϊat and gemopatrilat), and nitrates. [0084] DGAT inhibitors could be useful in treating other diseases associated with obesity, including sleep disorders. Therefore, the compounds described in the present invention could be used in combination with therapeutics for treating sleep disorders. Examples of suitable therapies for treatment of sleeping disorders for use in combination with the compounds of the present invention include melatonin analogs, melatonin receptor antagonists, ML 1 B agonists, GABA receptor modulators; NMDA receptor modulators, histamine-3 (H3) receptor modulators, dopamine agonists and orexin receptor modulators. [0085] The compounds described in the present invention could be used in combination with suitable anti-inflammatory agents. Examples of suitable antiinflammatory agents for use in combination with the compounds of the present invention include prednisone, dexamethasone, cyclooxygenase inhibitors (Le., COX-I and/or COX-2 inhibitors such as NSAIDs, aspirin, indomethacin, ibuprofen, piroxicam, naproxen, CELEBREX®, VIOXX®), CTLA4-Ig agonists/antagonists, CD40 ligand antagonists, IMPDH inhibitors, such as mycophenolate (CELLCEPT®), integrin antagonists, alpha-4 beta-7 integrin antagonists, cell adhesion inhibitors, interferon gamma antagonists, ICAM-I, tumor necrosis factor (TNF) antagonists (e.g., infliximab, OR 1384, including TNF-alpha inhibitors, such as tenidap, anti-TNF antibodies or soluble TNF receptor such as etanercept (ENBREL®), rapamycin (sirolimus or RAPAMUNE®) and leflunomide (Arava)), prostaglandin synthesis inhibitors, budesonide, clofazimine, CNI- 1493, CD4 antagonists (e.g., priliximab), p38 mitogen-activated protein kinase inhibitors, protein tyrosine kinase (PTK) inhibitors, IKK inhibitors, and therapies for the treatment of irritable bowel syndrome (e.g. j ZELNORM® and Maxi-K openers such as those disclosed in U.S. Patent No. 6,184,231 Bl). [0086] The above other therapeutic agents, when employed in combination with the compounds of the present invention, may be used, for example, in those amounts indicated in the Physicians' Desk Reference (PDR) or as otherwise determined by one of ordinary skill in the art. [0087] Also embraced within this invention is a class of pharmaceutical compositions comprising the compound of Formula (I) or a stereoisomer or a pharmaceutically acceptable salt thereof in association having one or more non-toxic, pharmaceutically-acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier" materials) and, if desired, other active ingredients. The compounds of the present invention may be administered by any suitable route, preferably in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. The compounds and compositions of the present invention may, for example, be administered orally, mucosally, or parentally including intravascularly, intravenously, intraperitoneally, subcutaneously, intramuscularly intrasternally and infusion techniques, in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. For example, the pharmaceutical carrier may contain a mixture of mannitol or lactose and raicrocrystalline cellulose. The mixture may contain additional components such as a lubricating agent, e.g. , magnesium stearate and a disintegrating agent such as crospovidone. The carrier mixture may be filled into a gelatin capsule or compressed as a tablet. [0088] The pharmaceutically active compounds of this invention can be processed in accordance with conventional methods of pharmacy to produce medicinal agents for administration to patients, including humans and other mammals. [0089] The compounds of the present invention can be administered to various mammalian species known to be subject to such maladies, e.g. , humans, in an effective amount up to 1 gram, preferably up to 200 mg, more preferably up to 100 mg in a regimen of single, two or four divided daily doses. [0090] The compounds of the Formula (T) can be administered for any of the uses described herein by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular, or inlrasternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally, including administration to the nasal membranes, such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds can, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release can be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds can also be administered liposomally. [0091] Exemplary compositions for oral administration include suspensions which can contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which can contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The compounds of the present invention can also be delivered through the oral cavity by sublingual and/or buccal administration. Molded tablets, compressed tablets or freeze-dried tablets are exemplary forms which may be used. Exemplary compositions include those formulating the present compound(s) with fast dissolving diluents such as mannitol, lactose, sucrose and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (avicel) or polyethylene glycols (PEG). Such formulations can also include an excipient to aid mucosal adhesion such as hydroxy propyl cellulose (HPC), hydroxy propyl methyl cellulose (HPMC), sodium carboxy methyl cellulose (SCMC), maleic anhydride copolymer (e.g. , Gantrez), and agents to control release such as polyacrylic copolymer (e.g., Carbopol 934). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use. [0092] Exemplary compositions for nasal aerosol or inhalation administration include solutions in saline which can contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, and/or other solubilizing or dispersing agents such as those known in the art. [0093] Exemplary compositions for parenteral administration include injectable solutions or suspensions which can contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid, or Cremaphor. [0094] Exemplary compositions for rectal administration include suppositories which can contain, for example, a suitable non-irritating excipient, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures, but liquefy and/or dissolve in the rectal cavity to release the drug. [0095] Exemplary compositions for topical administration include a topical carrier such as Plastibase (mineral oil gelled with polyethylene). [0096] It will be understood that the specific dose level and frequency of dosage for any particular subject can 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 species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. [0097] The present invention may be embodied in other specific forms without departing from the spirit or essential attributes thereof. This invention encompasses all combinations of preferred aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment or embodiments to describe additional more preferred embodiments. It is also to be understood that each individual element of the preferred embodiments is its own independent preferred embodiment. Furthermore, any element of an embodiment is meant to be combined with any and all other elements from any embodiment to describe an additional embodiment. [0098] The compounds of the present invention can be prepared as shown in the following reaction schemes and description thereof, as well as relevant published literature procedures that may be used by one skilled in the art. Exemplary reagents and procedures for these reactions appear hereinafter and in the working Examples.
ABBREVIATIONS [0099] The following abbreviations are employed in the Schemes, Examples and elsewhere herein:
AcOH = acetic acid DCM - dichloromethane
DJPEA = diisopropylethylamine
DMF = N,N-dxmethylformamide
DMSO = dimethyl sulfoxide
Dppp = (/?)-(+)- l,2-bis(diphenylphosphino)propane EDC = N-(3-dimthylaminopropyl)-N'-ethylcarbodiimide
EtOAc = ethyl acetate
EtOH = ethanol
Et3N = triethylamine
Et2O = diethyl ether HCl = hydrogen chloride
HOBt = 1-hydroxybenzotriazole
HPLC or LC = high performance liquid chromatography
K3PO4 = potassium phosphate tribasic
K2CO3 = potassium carbonate LiOH = lithium hydroxide
MeOH = methanol
MS or Mass Spec = mass spectrometry
NaCl = sodium chloride
NaHCO3 = sodium bicarbonate Na2CO3 = sodium carbonate
Na2SO4 = sodium sulfate
NaOH = sodium hydroxide Pd(OAc)2 = palladium(II) acetate
PI13PCI2 = triphenylphosphine dichloride
PG = protecting group
POCl3 - phosphorus oxychloride Pd(dppf)Cl2 = [1,1 '-bis(diphenylphosphino)-ferrocene]dichloropalladium(II)
TFA = trifluoroacetic acid
THF = tetrahydrofuran min = minute(s) h = hour(s) L = liter mL = milliliter μL = microliter g = gram(s) mg = milligram(s) mol = moles mrnol = millimole(s) nM = nanomolar [00100] Compounds of the present invention may be prepared by procedures illustrated in the accompanying schemes.
METHODS OF PREPARATION [00101] The compounds of the present invention can be prepared in a number of ways known to one skilled in the art of organic synthesis. The compounds of the present invention may be prepared by the methods described below, other with synthetic methods known in the art of synthetic organic chemistry, or by variations thereon as appreciated by those skilled in the art. The reactions are performed in a solvent or solvent mixture appropriate to the reagents and materials employed and suitable for the transformation being effected. It will be understood by those skilled in the art of organic synthesis that the functionality present on the molecule should be consistent with the transformation proposed. This will sometimes require a judgment to modify the order of the synthetic steps or to select one particular process scheme over another in order to obtain a desired compound of the invention. [00102] The compounds of the present invention can be prepared according to the general methods shown in the schemes below. Solvents, temperatures, pressures, and other reaction conditions may readily be selected by one of ordinary skill in the art.
Starting materials are commercially available or can be readily prepared by one of ordinary skill in the art using known methods. For the scheme and compounds described below, A, B, L, R3, and R4 are as described for a compound of Formula (I). [00103 J The following are the definitions of symbols used throughout Schemes 1,
2, 3 and 4:
PG is a suitable nitrogen protecting group, exemplified by benzyl, tert- butoxycarbonyl- [BOC], benzyloxycarbonyl- [CBZ], or 9-fiuorenylmethoxycarbonyl- [FMOC].
LG is a leaving group exemplified by halogen (Cl, Br, I) and sulfonates
("OSO2-aryl (e.g., -OSO2Ph or -OSO2PhCH3), or -OSO2-alkyl (e.g., -OSO2CH3 or -OSO2CF3)).
Scheme 1
Figure imgf000045_0001
[00104] Compounds of formula Ia' (with triazolopyridine core) can be prepared as described in Scheme 1. [00105] Compounds of formula II are either commercially available or available by means known to one skilled in the art. Compounds of formula III can be prepared by reacting compounds of formula II with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature. Compounds of formula V can be prepared by reacting compounds of formula III with a compound of formula IV in the presence of a coupling agent, such as N~[3~(dimethylaminopropyl)-N'- ethylcarbodiimide. Compounds of formula IV (where Z is an N-protecting group (PG) or a functional group (A) as defined in the claims) are either commercially available or available by means known to one skilled in the art. Compounds of formula VI can be prepared by reacting compounds of formula V with a dehydrating agent, such as a mixed solvent of ethanol and acetic acid at elevated temperature, or dichlorotriphenylphosphorane in the presence of a base, such as diisopropylethylamine. Compounds of formula VII can be prepared by reacting compounds of formula VI with carbon monoxide and methanol in a pressured vessel at elevated temperature in the presence of a palladium catalyst and a ligand, such as palladium acetate and (i?)-(+)-l,2-bis(diphenylphosphino)propane. Compounds of formula VIII can be prepared by saponification of compounds of formula VII in aqueous THF in the presence of a base, such as lithium hydroxide. Compounds of formula X can be prepared by reaction of compounds of formula VIII with an amino acid ester IX' in the presence of a coupling agent, such as N-[3- (dimethylaminopropyl)-N'-ethylcarbodiimide. Compounds of formula X where Z is an N-protecting group (PG) can be converted to compounds of formula XI by the methods described in Greene, T.W. et al,, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York (1991). Compounds of formula XII where A is R6(CO)~ can be prepared by acylation of compounds of formula XI with an acyl chloride or anhydride in the presence of a base, such as triethylamine. Compounds of formula XII where A is R6O(CO)- can be prepared by reaction of compounds of formula XI with a chloroformate in the presence of a base, such as triethylamine. Compounds of formula XII where A is R6N(CO)- can be prepared by reaction of compounds of formula XI with an isocyanate in the presence of a base, such as triethylamine. Compounds of formula Ia' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula XII in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification, Scheme 2
Figure imgf000047_0001
[Θ0106] Alternatively, compounds of formula Ia and Ia' (with triazolopyridine core) can be prepared as described in Scheme 2 starting from compounds of formula XIlI, which are either commercially available or available by means known to one skilled in the art. [00107] Compounds of formula XIV can be prepared by reacting compounds of formula XIII with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature. Compounds of formula XV can be prepared by reacting compounds of formula XIV with a compound of formula IV in the presence of a coupling agent, such as N-[3-(dimethylaminopropyl)-N'-ethylcarbodiimide, Compounds of formula XVI can be prepared by reaction of compounds of formula XV with a dehydrating agent, such phosphorus oxychloride at elevated, temperature, or dichlorotriphenylphosphorane in the presence of a base, such as diisopropylethylamine. Compounds of formula XVI where Z is an N-protecting group (PG) can be converted to compounds of formula XVII by the methods described in Greene, T.W. et al., Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York (1991). Compounds of formula XVIII where A is Rf'(CO)- can be prepared by acylation of compounds of formula XVII with an acyl chloride or anhydride in the presence of a base, such as triethylamine. Compounds of formula XVIII where A is R6O(CO)- can be prepared by reaction of compounds of formula XVII with a chloroformate in the presence of a base, such as triethylamine.
Compounds of formula XVIII where A is R6N(CO)- can be prepared by reaction of compounds of formula XVII with an isocyanate in the presence of a base, such as triethylamine. Compounds of formula XIX can be prepared by saponification of compounds of formula XVIII in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification. Compounds of formula I can be prepared by reaction of compounds of formula XIX with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as JV- [3- (dimethylammopropyl)-N -ethylcarbodiimide. Compounds of formula Ia' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula Ia (where B is -CO2R5 and Rs is an alkyl group) in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification.
Scheme 3
Figure imgf000049_0001
[00108] Compounds of formula Ic and Ic' (with triazolopyrazine core) can be prepared as described in Scheme 3. [00109] Compounds of formula XX are either commercially available or available by means known to one skilled in the art. Compounds of formula XXI can be prepared by reacting compounds of formula XX with anhydrous hydrazine in a polar solvent, such as 1,4-dioxane or pyridine at elevated temperature. Compounds of formula XXII can be prepared by reacting compounds of formula XXI with a compound of formula IV in the presence of a coupling agent, such as N~[3- (dimethylaminopropyl)-N'-ethylcarbodiimide. Compounds of formula χχπi cane prepared by reacting compounds of formula XXII with a dehydrating agent, such as phosphorus oxychloride at elevated temperature, or a phosphonium salt {e.g., triethylphosphine/carbon tetrachloride) in the presence of a base, such as diisopropylethylamine. Compounds of formula XXIV can be prepared by reacting compounds of formula XXIII with carbon monoxide and methanol in a pressured vessel at elevated temperature in the presence of a palladium catalyst and a ligand, such as palladium acetate and (i?)-(+)-l,2-bis(diphenylphosphino)propane. Compounds of formula χχ|γ where Z is an N-protecting group can be converted to compounds of formula XXV by the methods described in Greene, T. W. et al, Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York (1991). Compounds of formula χχvi where A is R6(CO)- can be prepared by acylation of compounds of formula XXV with an acyl chloride or anhydride in the presence of a base, such as triethylamine. Compounds of formula XXVI where A is R 0(CO)- can be prepared by reaction of compounds of formula XXV with a chloroformate in the presence of a base, such as triethylamine. Compounds of formula XXVI where A is R6N(CO)- can be prepared by reaction of compounds of formula XXV with an isocyanate in the presence of a base, such as triethylamine. Compounds of formula XXVII can be prepared by saponification of compounds of formula XXVI in aqueous THF in the presence of a base, such as lithium hydroxide. Compounds of formula I can be prepared by reaction of compounds of formula XXVII with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as N-[3- (dimethylaminopropyl)-N'-ethylcarbodiimide. Compounds of formula Ic' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula Ic (where B is -COOR5 and R5 is an alkyl group) in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification. Scheme 4
Figure imgf000051_0001
[00110] Compounds of formula Ib and Ib' (with Iriazolopyridazine core) can be prepared as described in Scheme 4. [00111] Compounds of formula XXVIII are either commercially available or available by means known to one skilled in the art. Compounds of formula XXIX can be prepared by reacting compounds of formula χχγiu -vvitli anhydrous hydrazine in a polar solvent, such as ethanol at room temperature. Compounds of formula xxx can be prepared by reacting compounds of formula XXIX with a compound of formula IV in the presence of a coupling agent, such as N-[3-(dimethylaniinopropyl)-N'- ethylcarbodiimide. Compounds of formula XXXI can be prepared by reacting compounds of formula XXX with a dehydrating agent, such as triethylphosphine/carbon tetrachloride in the presence of a base (e.g., diisopropylethylamine). Compounds of formula XXXI (where LG2 is chlorine) can be converted to compounds of formula XXXII by dechlorination under hydrogenolysis conditions in the presence of a palladium catalyst. Compounds of formula XXXI where Z is an N-protecting group can be converted to compounds of formula XXXIII by the methods described in Greene, T.W. et al., Protecting Groups in Organic Synthesis, John Wiley & Sons, Inc, New York (1991). Compounds of formula χχχiy where A is R6(CO)- can be prepared by acylation of compounds of formula XXXIII with an acyl chloride or anhydride in the presence of a base, such as triethylamine. Compounds of formula XXXIV where A is R6O(CO)- can be prepared by reaction of compounds of formula XXXIII with a chloroformate in the presence of a base, such as triethylamine. Compounds of formula XXXIV where A is R6N(CO)- can be prepared by reaction of compounds of formula XXXIII with an isocyanate in the presence of a base, such as triethylamine. Compounds of formula XXXV can be prepared by treating compounds of formula XXXIV with trifluoroacetic acid. Compounds of formula I can be prepared by reaction of compounds of formula XXXV with an amino acid ester or an aminoalcohol IX in the presence of a coupling agent, such as N-[3-(dimethylaminoρropyl)-N'-ethylcarbodiimide. Compounds of formula Ib' (where B is a carboxylic acid) can be prepared by saponification of compounds of formula Ib (where B is -COOR5 and R5 is an alkyl group) in aqueous THF in the presence of a base, such as lithium hydroxide, followed by acidification.
EXAMPLES [00112] The invention is further defined in the following Examples. It should be understood that the Examples are given by way of illustration only. From the above discussion and the Examples, one skilled in the art can ascertain the essential characteristics of the invention, and without departing from the spirit and scope thereof, can make various changes and modifications to adapt the invention to various uses and conditions. As a result, the invention is not limited to the illustrative examples set forth herein below, but rather defined by the claims appended thereto.
GENERAL [00113] The following methods were used in the working Examples, except where noted otherwise.
Analytical HPLC and HPLC/MS Methods Employed in
Characterization of Examples [00114] Reverse phase analytical HPLC was performed on Shimadzu LClOAS systems and reverse phase analytical HPLC/MS on Shimadzu LClOAS systems coupled with Waters ZMD Mass Spectrometers using the following methods:
Method A. Linear gradient of 10 to 100% solvent B over 2 min, with 1 min hold at 100% B;
UV visualization at 220 nm
Column: XTERRA® MS-C18> 2.1 X 50 mm
Flow rate: 1.0 ml/min
Solvent A: 0.1% trifluoroacetic acid, 10% water, 90 methanol. Solvent B: 0.1% trifluoroacetic acid, 90% methanol, 10% water.
Method B. Linear gradient of 0 to 100% solvent B over 4 min, with 1 min hold at
100% B;
UV visualization at 220 nm Column: PHENOMENEX® Luna C 18 4.6 x 50 mm Flow rate: 4 ml/min
Solvent A: 0.1% trifluoroacetic acid, 90% water, 10% methanol. Solvent B: 0.1% trifluoroacetic acid, 90% methanol, 10% water.
Method C. Linear gradient of 0 to 100% solvent B over 4 min, with 1 min hold at
100% B; UV visualization at 220 nro Column: PHENOMENEX® Luna Cl 8 4.6 x 50 mm Flow rate: 4 ml/min
Solvent A: 0.1% trifluoroacetic acid, 90% water, 10% acetonitrile. Solvent B: 0.1% trifluoroacetic acid, 90% acetonitrile, 10% water.
Method D. Linear gradient of 0 to 100% solvent B over 4 min, with 1 min hold at
100% B;
UV visualization at 220 nm
Column: PHENOMENEX® Luna Cl 8 4.6 x 50 mm Flow rate: 4 ml/min
Solvent A: 1OmM ammonium acetate, 90% water, 10% acetonitrile. Solvent B: 1OmM ammonium acetate, 90% acetonitrile, 10% water.
Method E. Linear gradient of 0 to 100% solvent B over 4 min, with 1 rnin hold at 100% B;
UV visualization at 220 nm
Column: Xbridge C 18 4.6 x 50 mm
Flow rate: 4 ml/min
Solvent A: 1OmM ammonium acetate, 90% water, 10% acetonitrile. Solvent B: 1OmM ammonium acetate, 90% acetonitrile, 10% water.
Method F. Linear gradient of 0 to 100% solvent B over 4 min, with 1 min hold at
100% B;
UV visualization at 220 nm Column: PHENOMENEX® Luna C18 4.6 x 50 mm Flow rate: 4 ml/min
Solvent A: 1OmM ammonium acetate, 90% water, 10% methanol. Solvent B: 1OmM ammonium acetate, 90% methanol, 10% water.
NMR Employed in Characterization of Examples [00115] 1H NMR spectra were obtained with Broker or JEOL Fourier transform spectrometers operating at frequencies as follows: 1H NMR: 400 MHz (Bruker or JEOL) or 500MHz (JEOL). 13C NMR: 100 MHz (Bruker or JEOL). Spectra data are reported in the format: chemical shift (multiplicity, coupling constants, number of hydrogens). Chemical shifts are specified in ppm downfield of a tetramethylsilane internal standard (δ units, tetramethylsilane = 0 ppm) and/or referenced to solvent peaks, which in 1H NMR spectra appear at 2.49 ppm for CDaHSOCD3, 3.30 ppm for CD2HOD5 and 7.24 ppm for CHCl3, and which in 13C NMR spectra appear at 39.7 ppm for CD3SOCD3, 49.0 ppm for CD3OD, and 77.0 ppm for CDCl3. AU 13C NMR spectra were proton decoupled.
PREPARATIVE EXAMPLE 1
3-(4-(terM3utoxycarbonylamino)phenyl)-[ 1 ,2,4]triazolo [4,3-α]pyridine-8-carboxylic acid
Figure imgf000055_0001
Preparation IA: 3~Bromo-2-hydrazinylpyridine
Figure imgf000055_0002
[00116] To a stirring solution of 3-brorno-2-chloropyridine (8.56 g, 45 mmol) in pyridine (40 mL) was added anhydrous hydrazine (4.0 g, 125 mmol). The resulting mixture was stirred at 6O°C for 24 h. Analysis by HPLC indicated that about 50% of starting material was consumed. Additional hydrazine (4.0 g, 125 mmol) was added and the reaction mixture was stirred at 8O°C for another 16 h (total 40 h). After cooling to room temperature, the resulting white suspension was filtered. The collected solid was washed with water (2 x 50 mL), and dried in a 50°C vacuum oven for 16 h to afford 5.13 g (61%) of the title compound as a white solid. HPLC/MS (Method C): retention time = 0.38 min, [M+H]+ = 188.0. Preparation IB: tot-Butyl 4-(2-(3-bromopyridin-2~y])hydrazinecarbonyl) phenylcarbamate
Figure imgf000056_0001
[00117] A mixture of 4~(fe/t--butoxycarbonylarnino)benzoic acid (5.05 g, 21.3 mmol), HOBt (2.9 g, 21.3 mmol) and EDC (4.07 g, 21.3 mmol) in CH2Cl2 (100 mL) was stirred at room temperature until becoming a clear solution, IA (4.0 g, 21.3 mmol) was then added. The reaction mixture was stirred at room temperature for 2 h, and then concentrated under reduced pressure. The obtained residue was stirred with EtOAc (15 mL) and water (150 mL) for 1 h. The resulting suspension was filtered. The collected solid was rinsed with water (100 mL) and EtOAc (5 mL), dried in a 50°C vacuum oven to afford 8.75 g (100%) of the title compound as a white solid. HPLC/MS (Method B): retention time - 2.32 min, [M+Hf = 407.1.
Preparation 1C: 4-(8-Bromo-[l;2,4]triazolo[433-α]pyridin-3-yI)aniline
Figure imgf000056_0002
[00118] A solution of IB (8.7 g, 21.3 mmol) in a mixed solvent of AcOH (15 mL) and EtOH (30 mL) was heated in a microwave reactor at 160°C for 1.5 h (The reaction was carried out in three individual microwave vials). The precipitate in the reaction vials was collected by filtration. The collected solid was rinsed with EtOH5 dried in a 50°C vacuum oven to yield 1.56 g of the title compound as a light brown solid. The mother liquor was heated in a microwave reactor at 185°C for 0.5 h, and then concentrated under reduced pressure. The obtained residue was dissolved in EtOAc, washed carefully with saturated aqueous NaHCO3, water, saturated aqueous NaCl, dried (Na2S O4) and concentrated under reduced pressure to give a crude product, which was purified by silica gel chromatography eluting with a gradient of EtOAc (50-100%) in hexanes to afford additional 1.18 g (total 2.74 g, 44%) of the title compound. HPLC/MS (Method A): retention time = 1.23 min, [MH-H]+ - 289.1.
Preparation ID: tert-Butyl 4-(8-bromo-[l,2J4]triazolo[4,3-α]pyridin-3-yl) phenylcarbamate
Figure imgf000057_0001
[00119] To a stirred solution of 1C (0.7 g, 2.42 mmol) in THF (10 niL) at room temperature was added άi-tert-bntyl dicarbonate (0.555 g, 2.54 mmol). The resulting mixture was stirred at 80°C for 16 h. Analysis by HPLC indicated that the starting amine was not consumed. Additional di-føt-butyl dicarbonate (0.278 g, 1.27 mmol) and THF (5 mL) were added. The reaction mixture was stirred at 80°C for another 24 h. After cooling to room temperature, the reaction mixture was diluted with EtOAc (200 mL), washed with water, saturated aqueous NaCl, dried (Na2SO4) and concentrated under reduced pressure. The crude product was purified using a silica gel cartridge (40 g) eϊuting with a gradient of EtOAc (30-100%) in hexanes to afford 0.307 g (54%) of the title compound as an off-white solid. HPLC/MS (Method A): retention time = 1.66 min, [M+H]+ = 389.3.
Preparation IE: Methyl 3-(4-(ferf-butoxycarbonylamino)phenyl)"[lJ2,4]triazolo[4,3- α]pyridine-8-carboxylate
Figure imgf000057_0002
[00120] Carbon monoxide gas was charged into a pressure bottle containing a mixture of ID (300 mg, 0.77 mmol), Pd(OAc)2 (69.2 mg, 0.31 mmol), dppp (127 mg, 0.31 mmol), Et3N (0.43 mL, 3.1 mmol), DMSO (5 mL) and MeOH (2.5 mL) until the pressure reached 30 psi. The bottle was then sealed and heated at 80°C for 16 h.
After cooling to room temperature, the reaction was diluted with EtOAc, washed with water, saturated aqueous NaCl, dried (Na2SO4) and concentrated under reduced pressure. The crude product was purified using a silica gel cartridge (40 g) eluting with a gradient of EtOAc (50-100%) in hexanes to afford 205 mg (72%) of the title compound as an off-white solid. HPLC/MS (Method A): retention time = 1.46 min, [M+H]+ = 369.4.
Example 1 [00121] To a solution of IE (150 mg, 0.44 mmol) in THF (2 mL) was added 2 M aqueous LiOH solution (2 mL). The resulting mixture was stirred at room temperature for 16 h. The reaction mixture was acidified to pH 3-4 by dropwise addition of 1 N aqueous HCl. The resulting precipitate was collected by filtration, washed with water, and dried in a 50°C vacuum oven to afford 108 mg (75%) of the title compound as an off-white solid. HPLC/MS (Method B): retention time = 1.38 min, [M+H]+ = 355.3. 1H NMR (CD3OD, 400 MHz): δ 8.68 (d J= 8.7 Hz, IH), 8.26 (d, J= 6.0 Hz, IH), 7.80 (d, J= 8.8 Hz, 2H), 7.72 (d, J= 8.8 Hz, 2H), 7.23 (t, J= 6.9 Hz, IH), 1.55 (s, 9H).
EXAMPLE 2
(S)-2-(3-(4-( tert-Butoxycarbonylamino)phenyl)-[l,2,4]triazolo[4,3-α]pyridme-8- carboxamido)-3-methylbutanoic acid
Figure imgf000058_0001
Preparation 2A: (S)-Etfayl 2-(3-(4-(tert-butoxycarbonylamino)phenyl)-[l,2,4]triazolo [4,3-α]pyridine-8-carboxamido)-3-methylbutanoate
Figure imgf000058_0002
[00122] To a solution of Example 1 (50 mg, 0.14 mmol) in DMF (1 mL) was added L-valine ethyl ester hydrochloride salt (33 mg, 0.18 mmol), HOBt (28.4 mg, 0.21 mmol), EDC (40 mg, 0.21 mmol) and (JPr)2EtN (0.049 mL, 0.28 mmol). The resulting mixture was stirred at room temperature for 16 h, and then partitioned between water and EtOAc, The aqueous layer was extracted with EtOAc (2 x 20 mL), and the combined EtOAc extracts were washed with water, saturated aqueous NaCl, dried (Na2SO.^) and concentrated under reduced pressure. The crude product was purified using a silica gel cartridge (12 g) eluting with a gradient of EtOAc (30- 100%) in hexanes to afford 65 mg (97%) of the title compound as an off-white foam. HPLC/MS (Method A): retention time - 1.93 mm, [M+Hf = 482.5.
Example 2 [00123] To a solution of 2A (15 mg, 0.03 mmol) in THF (1 mL) was added 1 N aqueous NaOH solution (1 mL). The resulting mixture was stirred at room temperature for 16 h, and then acidified to pH 3-4 by dropwise addition of 1 N aqueous HCl. The mixture was extracted with EtOAc (3 x 10 mL), and the combined EtOAc extracts concentrated under reduced pressure. The crude product was purified using preparative HPLC (PHENOMENEX® Luna Axia, 5μ 30 x 100 mm using MeOH-H2O-TFA solvent system) to afford 8.2 mg (60%) of the title compound as a yellow solid. HPLC/MS (method A): retention time = 1.79 min, [M+H]+ = 454.5. 1H NMR (CD3OD, 400 MHz): δ 8.68 (d, /= 8.7 Hz, IH), 8.26 (d, J- 6.0 Hz, IH), 7.80 (d, J= 8.8 Hz, 2H), 7.72 (d, J= 8.8 Hz, 2H), 7.23 (t, J = 6.9 Hz, IH), 4.70 (d, J = 4.4 Hz, IH), 2.37-2.47 (m, IH), 1.55 (s, 9H), 1.15 (d, J - 7.2 Hz, 3H), 1.12 (d, J= 7.2 Hz, 3H).
EXAMPLE 3
(5)-3-Methyl-2~(3-(4-(3-(2-(trifluoromethyl)phenyl)ureido)phenyl)- [l,2,4]triazolo[4,3-β]pyridine-8-carboxamido)butanoic acid
Figure imgf000059_0001
Preparation 3A: (S)-Ethyl 2"(3~(4-aminophenyl)-[l,2,4]tria2θlo[4f3~α]pyridine-8- carboxamido)-3-methylbutanoate
Figure imgf000060_0001
[00124] To a solution of 2A (105 rag, 0.22 mmol) in CH2Cl2 (1.5 niL) was added trifluoroacetic acid (1.5 mL). After stirring at room temperature for 1 h, the reaction mixture was concentrated under reduced pressure. The crude product was taken into EtOAc, then washed with saturated aqueous NaHCO3, saturated aqueous NaCl, dried (Na2SO4) and concentrated under reduced pressure to afford 81 mg (96%) of the title compound as a yellow foam. HPLC/MS (Method B): retention time = 2.57 min, [MH-H]+ = 381.5.
Preparation 3B: (S)-Ethyl 3-methyl-2-(3-(4-(3-(2-(trifluoromethyl)phenyl)ureido) phenyl)~[ 1 ,2,4]triazolo[4,3-α]pyridine-8-carboxamido)butanoate
Figure imgf000060_0002
[00125 J A solution of 3 A (81 mg, 0.20 mmol) and 2-trifluoromethylphenyI isocyanate (59 mg, 0.32 mmol) in THF (2 mL) was stirred at 60°C for 1 h. Analysis by HPLC indicated- that the starting amine was not consumed. Additional 2- trifluoromethyl phenyl isocyanate (20 mg, 0.107 mmol) was added and the reaction stirred at 60°C for another 0.5 h. After cooling to room temperature, the reaction was concentrated under reduced pressure. The crude product was purified using a silica gel cartridge (12 g) eluting with a gradient of EtOAc (0-100%) in hexanes to afford 103 mg (87%) of the title compound as a white solid. HPLC/MS (Method A): retention time = 1.94 min, [M+H]+ = 569.6.
Example 3 [00126] To a solution of 3B (105 mg, 0.185 mmol) in THF (2 mL) was added 2 N aqueous LiOH solution (2 mL). The resulting mixture was stirred at room temperature for 3 h. Analysis by HPLC indicated that the starting ester was consumed. The reaction mixture was cooled at 0°C, and acidified to pH 3 by dropwise addition of 1 N aqueous HCl. The mixture was extracted with EtOAc (2 x 20 mL) and the combined EtOAc extracts were concentrated under reduced pressure. The crude product was purified using preparative HPLC (PHENOMENEX® Luna Axia, 5μ 3O x 100 ram, using MeOH-H2O-TFA solvent system) to afford 71 mg (71%) of the title compound as a white solid. HPLC/MS (Method A): retention time = 1.85 min, [M+Hf = 541.4. 1H NMR (CD3OD, 400 MHz): δ 8.73 (d5 J = 7.2 Hz, IH), 8.31 (d, J- 6.6 Hz, IH), 7.97 (d, J- 7.2 Hz, IH), 7.86 (d, J- 8.8 Hz, 2H), 7.79 (d, J= 8.8 Hz, 2H), 7.56-7.72 (m, 2H), 7.22-7.37 (m, 2H), 4.70 (d, J= 4.8 Hz, IH), 2.37-2.48 (m, IH), 1.15 (d, J= 7.2 Hz, 3H), 1.12 (d, J = 7.2 Hz, 3H).
EXAMPLE 4
(5)-3-Melhyl-2-(3-(4-(3-(2-(trifluoromethoxy)phenyl) ureido)phenyl)- [1 ,2,4]triazolo[4,3~α]pyπdine-8-carboxamido)butanoic acid
Figure imgf000061_0001
Preparation 4A: Ethyl-2-hydrazinylnicotinate
Figure imgf000061_0002
[00127] A solution of ethyl 2-chloronicotinate (10.0 g, 53.9 mmol) and anhydrous hydrazine (2.04 g, 63.8 mmol) in anhydrous 1,4-dioxane (200 mL) was stirred at 60°C for 2 h. Analysis by LC/MS indicated the starting material was not consumed. Additional amount hydrazine (2.04 g, 63.8 mmol) was added and the reaction mixture was stirred at 60°C overnight. After cooling to room temperature, the reaction mixture was concentrated in vacuo. The obtained residue was triturated with MeOH (35 niL), the insoluble white solid was removed by filtration and discarded. The methanol solution was concentrated in vacuo to yield a fluffy yellow solid which was triturated in diethylether to yield the title compound as an orange solid (5.4 g, 55%). HPLC/MS (Method D): retention time - 1.46 min, [M+H]+ = 182.3.
Preparation 4B: Ethyl 2~(2~(4-(fer^butoxycarbonylarnino)benzoyl)hydrazinyI) nicotinate
Figure imgf000062_0001
[00128] A suspension of 4-(terϊ-butoxycarbonylamino)benzoic acid ( 10.9 g, 45.8 mmol), HOBt (6.25 g, 46 mmoϊ) and EDC (8.74 g, 46 ramol) in anhydrous DCM (200 mL) was stirred at room temperature for 10 min, and then a solution of 4A (9.5 g, 52.4 mmol) in anhydrous DCM (120 m.L) was added. The reaction mixture was stirred at room temperature for 2,5 h, and then concentrated in vacuo, The obtained residue was suspended in EtOAc (300 mL) and water (200 mL) and stirred at room temperature for 0.5 h. The insoluble material was isolated by filtration, rinsed with water and a small amount of EtOAc, and then air dried to yield the title compound as a white solid (15.3 g, 73%). HPLC/MS (Method F): retention time = 3,22 min, [M+H]+ = 401.2.
Preparation 4C: Ethyl 3-(4-aminophenyl)-[l,2,4]triazolot4J3-ύ;]pyridine-8- carboxylate
Figure imgf000062_0002
[00129] A suspension of 4B (13.4 g, 33.3 mmol) in POCl3 (105 mL) was refluxed for 2.5 h. After cooling to room temperature, the reaction mixture was concentrated in vacuo to remove most of the POCI3. The residue was then dissolved in ethanol (100 mL), and slowly poured into water (350 mL) with stirring. After stirring for 30 min, the resulting mixture was concentrated in vacuo. To the residue was added 6N aqueous HCl (92 mL), and the mixture was heated at 6O°C for 3 h. After cooling to room temperature, the reaction mixture was neutralized with a NaOH aqueous solution to pH = 5. The precipitate was isolated by filtration, rinsed with water, and air-dried to yield the title compound as a golden-brown solid (7,27 g, 77%), HPLC/MS (Method F): retention time = 2.01 min, [M+H]+ - 283.1.
Preparation 4D: Ethyl 3-(4-(3-(2-(trifluoromethoxy)phenyl)ureido)phenyϊ)-[ 1,2,4] triazolo[4,3-α]pyridine-8-carboxylate
Figure imgf000063_0001
[00130] A solution of 4C (845 mg, 3 mmol) and o- tiiQuromethoxyphenylisocyanate (650 mg, 3.2 mmol) in anhydrous THF (9 mL) was stirred at 70°C for 2 h. LC/MS analysis indicated the starting amine was not consumed. Additional o-trifluororaethoxyphenyl-isocyanate (300 mg, 1.48 mmol) was added, and the mixture stirred at 70°C for another 3.5 h. The mixture was allowed to cool to room temperature. The resulting precipitate was collected by filtration and rinsed with THF several times to yield the title compound as a yellow solid (1.12 g, 77%). HPLC/MS (Method D): retention time = 2.35 min, [M+H]+ = 486.2.
Preparation 4E: 3-(4-(3-(2-(Trifluoromethoxy)phenyl)ureido)phenyl)-[l ,2,4]triazolo [4,3-α]pyridme-8-carboxylic acid
Figure imgf000063_0002
[00131] To a solution of 4D (1.1 g, 2.25 mmol) in THF (30 mL) was added IN aqueous LiOH solution (4.5 mL, 4.5 mmol). The reaction mixture was stirred at room temperature for 2 h, and then neutralized with 1 N aqueous HCl. The mixture was concentrated in vacuo. The obtained residue was suspended in ethyl acetate (10 mL) and water (15 mL) and stirred for 5 min. The insoluble material was isolated by filtration and air-dried to yield the title compound (893 mg, 87%) as a brown solid. HPLC/MS (Method D): retention time = 1.64 min, [M+Hf - 458.2.
Example 4 [00132] A suspension of 4E (46 mg, 0.1 mmol), EDC (19.2 mg, 0.1 mmol) and HOBt (13.6 mg, 0.1 mmol) in anhydrous DCM (1 mL) was stirred at room temperature for 5 min, and then L- Valine methyl ester (12.5 mg, 0.095 mmol) was added. The mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was suspended in ethyl acetate (3 mL) and washed with saturated aqueous NaHCO3 (1 x 0.8 mL), 1 N aqueous HCl (1 x 1 mL) and water (1 x 1 mL). The solvent was removed in vacuo to yield the product as an ester intermediate. The ester was dissolved in THF (1.5 mL) and 1 N aqueous LiOH solution (0.35 mL, 0.35 mmol) was added. The mixture was stirred at room temperature for 4 h, neutralized with 1 N aqueous HCl and concentrated in vacuo. The product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield the title compound. HPLC/MS (Method E): retention time = 1.73 min, [M+H]+ = 557.1.
EXAMPLES 5 TO 21 [00133] Examples 5 to 21 were prepared by coupling 3-(4-(3-(2~(trifiuorornethoxy) phenyl) ureido)phenyl)-[l,2,4]triazolo[4,3-«]pyridine-8-carboxylic acid (4E) with various amino acid esters according to the general procedures described for Example 4 and listed in Table 1. Analytical data for the compounds in Table 1 are reported as follows: compound retention times were recorded using LC-MS conditions (Method E), and the molecular masses of the compounds were determined by MS (ES) by the formula m/z. TABLE 1
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0002
EXAMPLE 22
(Sy- 1 -(3 -(4-(Methoxycarbonylamino)phenyl)- [ 1 , 2 ,4 ]tr iazolo [4,3 -α]pyridine- carbonyl)pyrrolidme-2-carboxylic acid
Figure imgf000067_0001
Preparation 22A: Ethyl 3-(4-methoxycarbonylamino)phenyl)-[l,2,4]triazoIo[4J3-<3] pyridine-8 -carboxylate
Figure imgf000068_0001
[00134] To a solution of 4C (0.845 g, 3 mmol) in a mixed solvent of pyridine (10 mL) and dichloromethane (10 mL) at 0°C was added dropwise methylchlorofomiate (0.57 g, 6 mmol). The resulting mixture was stirred at 0°C for 10 min, then at room temperature for 50 min. The reaction mixture was concentrated in vacuo. The solid residue was triturated in EtOAc (15 mL) and water (15 mL) to yield a yellow solid as the first crop of the product. The solution phase was separated into layers. The organic layer was washed with water (1 x 5 mL), dried over anhydrous Na2SO4> and concentrated in vacuo to yield the second crop of product. The combined product was further triturated in MeOH to afford the title compound as a yellow solid (0.55 g, 54%). HPLC/MS (Method D): retention time = 1.54 min, [M+H]"1 - 341.2
Preparation 22B: 3-(4-(Methoxycarbonylamino)phenyl)-[ 1 ,2,4]triazolo[4,3» α]pyridine~8-carboxylic acid
Figure imgf000068_0002
[00135] To a suspension of 22A (0.55 g, 1.61 mmol) in methanol (20 mL) was added 1 N aqueous NaOH solution (3 mL, 3 mmol). The mixture was stirred at room temperature for 2.5 h, and then concentrated in vacuo. The residue was suspended in water (20 mL) and acidified with 1 N aqueous HCl to pH 5. The resulting precipitate was isolated by filtration, rinsed with water several times, and air dried to yield the title compound as a brown solid (0.42 g, 84%). HPLC/MS (Method F): retention time = 1.34 min, [MH-H]+= 313.1.
Example 22 [00136] A suspension of22B (31 mg, 0.1 mmol), EDC (19 mg, 0.1 mmol) and
HOBt (13.6 mg, 0.1 mmol) in anhydrous dichloromethane (1 mL) was stirred at room temperature for 5 min, and then L-proline methyl ester (12.3 mg, 0.095 mmol) was added. The resulting mixture was stirred at room temperature overnight, and then concentrated in vacuo. The obtained residue was suspended in ethyl acetate (3 mi) and washed with saturated aqueous NaHCO3 (1 x 0.8 mL), 1 N aqueous HCl (1 x 1 ITiL) and water (1 x 1 mL). The solvent was removed in vacuo to yield the product as an ester intermediate. The ester was dissolved in THF (1.5 mL) and then 1 N aqueous LiOH solution (0.35 mL, 0.35 mmol) was added. The mixture was stirred at room temperature for 4 h, acidified with 1 N aqueous HCl and dried in vacuo. The product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield the title compound (8.5 mg, 20%). HPLC/MS (Method E): retention time = 1.13 min, [M+H]+ = 412.06. 1H NMR (500 MHz, DMSO-d6): 5 ppm 10.02 (s, IH), 8.56 - 8.68 (m, IH), 7.83 (d, J- 8.80 Hz5 2H), 7.72 (d, J= 8.80 Hz, 2H), 7.36 - 7.51 (in, IH), 7.03 - 7.12 (m, IH), 4.40-4.50 (m, IH), 3.71 (s, 3H), 3.48-3.55 (m, IH), 3.40-3.45 (m, IH), 2.28 - 2.35 (m, IH), 1.92 - 2.02 (m, IH), 1.82 - 1.92 (m, 2H).
EXAMPLE 23
(5)-3-Methyl-2-(3-(4-propionamidophenyl)-[l,2,4]triazolo[4,3-α]pyridine-8- carboxamido)butanoic acid
Figure imgf000069_0001
Preparation 23A: Ethyl 3-(4-propionamidophenyl)-[l,2J4]triazolo[4,3-a]pyridine-8- carboxylate
Figure imgf000069_0002
[00137] To a suspension of 4C (0.85 g, 3.01 mmol) in anhydrous dichloromethane (25 mL) was added triethylamine (0.4 g, 4 mmol), followed by dropwise addition of propionylchloride (0.285 g, 3.1 mmol). The resulting mixture was stirred at room temperature for 40 min. Analytical HPLC showed the starting amine was not consumed. Additional propionylchloride (0.15 g, 1.63 mmol) was added and the reaction was stirred at room temperature for 30 min. The mixture was diluted with dichloromethane (10 mL), washed with water (Ix), saturated aqueous NaBCC^ (3x) and saturated aqueous NaCl (Ix), dried over anhydrous Na2SO4, and concentrated in vacuo to yield a yellow solid. The product was triturated in MeOH to afford the title compound as a light brown solid (0.76 g, 75%). HPLC/MS (Method C): retention time - 1.36, [M+Hf = 339.5.
Preparation 23B: 3-(4-Propionamidophenyl)-[lr2,4]triazolo[4,3-a]pyridine-8- carboxylic acid
Figure imgf000070_0001
[00138] To a suspension of 23A (0.76 g, 2.25 mmol) in methanol (38 mL) was added 1 N aqueous NaOH solution (5 mL, 5 mmol). The mixture was stirred at room temperature for 2 h, and then concentrated in vacuo. The crude product was dissolved in water (40 mL) and acidified with 1 N aqueous HCl solution. The resulting precipitate was isolated by filtration, rinsed with water, and air dried to afford the title compound as a brown solid (0.50 g, 72%). HPLC/MS (Method F): retention time =
Figure imgf000070_0002
Example 23 [00139] A suspension of 23B (25.4 mg, 0.081 mmol), EDC (15.6 mg> 0.081 mmol) and HOBt (11 mg, 0.081 mmol) ) in anhydrous dichloromethane (1 mL) was stirred at room temperature for 5 min, and then L- valine methyl ester (11.8 mg, 0.09 mmol) was added. The resulting mixture was stirred at room temperature overnight and then concentrated in vacuo. The crude product was suspended in EtOAc (3 mL) and washed with saturated aqueous NaHCOj (1 x 0.8 mL), 1 N aqueous HCl (1 x 1 mL) and water (1 x 1 mL). The solvent was removed in vacuo to yield the product as an ester intermediate. The ester was dissolved in THF (1.5 mL) and then 1 N aqueous LiOH solution (0.35 mL, 0.35 mmol) was added. The mixture was stirred at room temperature for 4 h, acidified with 1 N aqueous HCl and dried in vacuo. The product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system Io afford the title compound (12 mg, 29%). HPLC/MS (Method E): retention time = 1.10 min, [M+H]+ = 410.09. 1H NMR (500 MHz, DMSO-dό): δ ppm 10.20 (s, 1 H), 10.03 (d, J = 8.25 Hz, IH), 8.75 (d, J= 7.15 Hz, IH), 8.15 (d, J= 7.15 Hz, IH), 7.80 - 7.91 (m, 4H), 7.20 (t, J = 6.87 Hz, IH), 4.57 (dd, J= 8.25, 4.40 Hz, IH), 2.38 (q, J =7.33 Hz, 2H), 2.31 (dd, J= 11.55, 7.15 Hz, IH), 1.11 (I, J= 7.42 Hz, 3H), 1.04 (d, J= 6.60 Hz, 3H)5 1.01 (d, J- 6.60 Hz, 3H).
EXAMPLE 24 (1S)-3-(4-HydiOxyphenyl)-2-(3-(4-propionamidophenyl)-[ 1 ,2,4]triazolo[4,3~a]pyridine-
8-carboxamido)propanoic acid
Figure imgf000071_0001
[00140] The title compound was prepared from 23B and L-tyrosine methyl ester as described for Example 23C. HPLC/MS (Method E): retention time = 1.01 min, [M+H]+ = 474.06.
EXAMPLE 25
(S)-2-(3-(4-(6-Chlorobenzo[J]thiazol-2-ylamino)phenyl>[lJ2!14]triazolo[4J3- a]pyridine-8-carboxamido)-3-methylbutanoic acid
Figure imgf000071_0002
Preparation 25A: 4-(6-Chlorobenzo[<φhiazol-2-ylamino)benzoic acid
Figure imgf000071_0003
[00141] To a suspension of tert-butyl 4-aminobenzoate (6.36 g, 32.9 mmol) and 2,6-dichlorobenzothiazole (5 g, 24.5 mmol) in isopropanol (150 mL) was added 4 N HCl solution in 1 ,4-dioxane (1.5 mL, 6 mraol). The mixture was refluxed at 100°C for 7 h, and then allowed to cool to room temperature. The resulting precipitate was isolated by filtration to yield the first crop of the title compound as a white solid (3.13 g). The mother liquor was concentrated in vacuo and then stirred in a mixed solvent of TFA (20 mL) and dichloromethane (20 mL) overnight. The solvent was removed in vacuo, the obtained residue triturated with isopropanol/ethyl acetate. The isolated white solid was further stirred in a mixed solvent of isopropanol (12 roL) and ethyl acetate (8 mL) for 0.5 h, then isolated by filtration to yield a second crop of the title compound (3.5 g) (total 6.63 g, 85%). HPLC/MS (Method C): retention time = 2.51 min, [M+H]+ - 305.3.
Preparation 25B: Ethyl 2-(2-(4-(6-chlorobenzo[J]thiazol-2-ylamino)benzoyl) hydrazinyl)nicotinate
Figure imgf000072_0001
[00142] A suspension of 4-(6-chlorobenzo[ύ?]thiazoI-2-ylamino)benzoic acid (117 mg, 0.38 mmol), EDC (88 mg, 0.46 mmol) and HOBt (63 mg, 0.46 mmol) in a mixed solvent of DMF (1 mL) and CH2CI2 (1 mL) was stirred for 5 min, and then ethyl 2- hydrazinylnicotinate (4A) (70 mg, 0.38 mmol) was added, followed by triethylamine (78 mg, 0.77 mmol). The mixture was stirred at room temperature for 2 h. Analysis by LC/MS indicated the starting material was not consumed. Additional EDC (20 mg, 0.1 mmol) and HOBt (15 nag, 0.11 mmol) were added, and the mixture stirred for another 2 h before quenching with water (3 mL). The resulting precipitate was isolated by filtration and triturated with MeOH to yield the title compound as a fluffy white solid (94 mg). The methanol solution was concentrated in vacuo to collect additional product (50 mg) (total 144 mg, 87% yield). HPLC/MS (Method C): retention time = 2.23 min, [M+H]+ = 468.4. Preparation 25C: Ethyl 3-(4-(6-chlorobenzo[#hiazol~2-yIamino)phenyl)-[ 1,2,4] triazolo[4,3-#jpyridirie-8-carboxylate
Figure imgf000073_0001
[00143] To a solution of ethyl 2-(2-(4-(6-chlorobenzo[<|thiazoI-2- ylamϊno)benzoyl) hydrazinyl)benzoate (142 ing, 0.3 mmol) in a mixed solvent of CCl4 (0,6 mL), THF (0.8 mL) and CH2Cl2 (1.2 mL) cooled to 0°C was added diisopropylethylamine (392 mg, 3 mmol) under argon atmosphere and then triethylphosphine (179 mg, 1,52 mmol). The mixture was stirred at O°C for 45 min, and then quenched with water (2.5 mL). The resulting precipitate was isolated by filtration and air dried to yield the title compound as a brown solid (108 mg). The filtrate was concentrated in vacuo to yield additional product (15 mg) (total 123 mg, 90% yield). HPLC/MS (Method C): retention time = 2.21 min, [M+Hf = 450.3.
Preparation 25D: 3-(4-(6-Chlorobenzo[t/]thiazol-2-ylamino)phenyl)- [l ,2,4]triazolo[4,3-fl]pyridine-8-carboxylic acid
Figure imgf000073_0002
[00144] To a suspension of ethyl 3-(4-(6-chlorobenzo[J]thiazol-2- ylammo)phenyl)-[l,2,4]triazolo[4,3-α]pyridine-8-carboxylate (935 mg, 2.08 mmol) in THF (20 mL) was added 1 N aqueous LiOH solution (4 mL, 4 rnmol). The mixture was stirred at room temperature overnight, concentrated to one third of the initial volume, and then acidified with 1 N aqueous HCl. To the resulting precipitate was added water (15 mL) and CH2Cl2 (5 mL). After stirring for 15 min, the precipitate was isolated by filtration, rinsed with water and CH2Cl2, and then air dried to yield the title compound as a brown solid (790 mg, 90%). HPLC/MS (Method C): retention time = 1.92 min, [M+H]+ = 422.3.
Example 25 [00145] A suspension of 3-(4-(6-chlorobenzo[</]thiazol-2-ylamino)phenyl)- [l,2,4]triazoIo[4,3-«]pyridme-8-carboxylic acid (33.7 mg, 0.08 tnmol), EDC (15.5 mg, 0.081 ramol) and HOBt (11 mg, 0.081 mmol) in a mixed solvent of DMF (0.5 niL) and CH2Cl2 (0.5 mL) was stirred for 5 min. Then to this mixture was added in L- Valine methyl ester hydrochloride salt (13.4 mg, 0.08 mmol) in CH2CI2 (0.5 mL) and diisopropylethylamine (20.7 mg, 0.16 mmol). The resulting mixture was stirred at room temperature overnight. The solvent was removed in vacuo. The mixture was suspended in 2: 1 EtOAc/THF and washed with water (2 x 1 mL), The solvent was removed in vacuo. The obtained ester intermediate was suspended in THF (1 mL) and then 1 N aqueous LiOH solution (0.3 mL, 0.3 mmol) was added. The mixture was stirred at room temperature overnight, acidified with 1 N aqueous HCl. The solvent was removed in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to afford the title compound (18.3 mg, 36%). HPLC/MS (Method D): retention time = 1.91 min, [M+Hf = 521.13. 1H NMR (DMSO-D6, 500 MHz): δ 10.96 (s, IH), 10.04 (d, /= 8.25 Hz, 1H),8.8O (d, J = 6.05 Hz, IH), 8.17 (d, J= 6.05 Hz, IH), 8.06 (d, J= 8.80 Hz, 2H), 8.00 (d, J= 2.20 Hz, IH), 7.94 (d, J= 8.80 Hz, 2H), 7.65 (d, J- 8.80 Hz, IH), 7.38 (dd, J= 8.80, 2.20 Hz, IH), 7.21 (t, J= 6.87 Hz, IH), 4.58 (dd, J= 8.25, 4.40 Hz, IH), 2.24 - 2.39 (m, IH). 1.03 (dd, J= 16.50, 6.60 Hz, 6H).
EXAMPLES 26 TO 43 [00146] Examples 26 to 43 were prepared by coupling 3-(4-(6- chlorobenzo[dr]thiazol-2-y]amino)phenyl)-[l,2,4]triazolo[4J3-α]pyridine-8-carboxylic acid (25D) with various amino acid esters according to the general procedure described in Example 25E and listed in Table 2. Analytical data for the compounds in Table 2 was reported as follows: compound retention times were recorded using LC- MS conditions (Method D), and the molecular mass of the compounds were determined by MS (ES) by the formula m/z.
TABLE 2
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0002
EXAMPLE 44
(5)-3-Methyl-2-(3-(4-(thiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4,3~<2]pyridine- carboxamido)butanoic acid
Figure imgf000080_0001
Preparation 44A: 4-(Thiazol-2-ylamino)benzoic acid
Figure imgf000081_0001
[00147] To a suspension of tert-butyl 4-aminobenzoate (1.93 g, 10 mmol) and 2- bromothiazole (1.2 g, 7.3 mmol) in isopropanol (60 mL) was added 4 N HCl solution in 1,4-dioxane (0.5 mL, 2 mmol). The mixture was refluxed at 100°C for 50 h, and then allowed to cool to room temperature. The mixture was concentrated to one third of the initial volume. The resulting precipitate was isolated by filtration to yield the first crop of the title compound as a brown solid (915 mg). The mother liquor was dried in vacuo. The obtained residue was dissolved in ethyl acetate (50 mL), washed with water (2 x 15 mL), dried over anhydrous Na2SO4, and concentrated in vacuo. The residue was triturated with isopropanol (8 mL) to yield the second crop of the title compound (341 mg) (total 1.25 g, 78 % yield). HPLC/MS (Method D): retention time = 0.57 mm, [M+Hf = 221.3.
Preparation 44B: Ethyl 2-(2-(4-(thiazol~2-ylamino)benzoyl)hydrazmyl)benzoate
Figure imgf000081_0002
[00148] A suspension of 4-(thiazol-2-ylammo)benzoic acid (44A) (3.27g, 14.7 mmol), EDC (2.85 g, 14.9 mmol) and HOBt (2.02 g, 14.9 mmol) in a mixed solvent of DMF (40 mL) and CH2CI2 (80 mL) was stirred for 5 min, and then to this mixture was added in ethyl 2-hydrazinylnicotinate (4A) (2.7 g, 14.9 mmol) and triethylamine (2.25 g, 22,35 mmol). The mixture was stirred at room temperature for 3.5 h.
Analysis by LC/MS indicated the starting material was not consumed. Additional amount of EDC (250 mg, 1.3 mmol) and ethyl 2-hydrazinylnicotinate (4A) (200 mg, 1.1 mmol) were added and the reaction mixture stirred overnight. The reaction mixture was concentrated in vacuo to one third of the initial volume, and then diluted with water (200 mL) and ethyl acetate (100 mL). The insoluble material was isolated by filtration to yield the title compound as a light brown solid (1.14 g). The filtrate was separated into organic and aqueous layers. The organic layer was washed with water, and then concentrated in vacuo. The obtained residue was triturated with methanol to yield the second crop of the title compound (135 g). (total 2.49 g, 44% yield). HPLC/MS (Method C): retention time = 1.11 min, [MH-H]+ = 384.2.
Preparation 44C: Ethyl 3-(4-(thiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4,3- α]pyridine-8-carboxylate
Figure imgf000082_0001
[00149] A suspension of ethyl 2-(2-(4-(thiazol-2-ylamino)benzoyl)hydrazinyl) benzoate (44B) (1.22 g, 3.18 nxtnol) in POCI3 (7.5 mL) was heated in a sealed tube at 110°C for 3 h. After cooling to room temperature, the reaction mixture was concentrated in vacuo to remove most of the POCl3. The residue was dissolved in MeOH (5 mL) and water (15 mL) and neutralized with IN aqueous NaOH solution to pH ~5-6. The resulting precipitate was isolated by filtration, rinsed with water and methanol, and then air dried to yield the title compound as a brown solid (1.13 g, 97%). HPLC/MS (Method F): retention time = 2.75 min, [M+H]+ = 366.2.
Preparation 44D: 3-(4-(Thiazol-2-ylamino)phenyl)-[l ,2,4]triazolo[4,3-α]pyridine-8- carboxylic acid
Figure imgf000082_0002
[00150] To a solution of ethyl 3-(4<thiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4}3- α]pyridme-8-carboxylate (44C) (1.2 g, 3.3 mmol) in THF (25 mL) was added 1 N aqueous LiOH solution (6 mL, 6 mmol). The mixture was stirred at room temperature overnight., acidified with 1 N aqueous HCl and then concentrated in vacuo. The obtained residue was stirred in a mixed solvent of water (20 mL) and methanol (5 mL) for 5 min. The resulting precipitate was isolated by filtration, rinsed with water and methanol. The solid material was then triturated with a mixed solvent of isopropanol (20 mL), methanol (5 mL) and water (10 mL) to afford the title compound as a dark green solid (1.10 g, 100%). HPLC/MS (Method D): retention time = 1.09 min, [M+H]+ = 338.2.
Example 44 [00151] A suspension of 3-(4-(thiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4,3- α]pyridine-8-carboxylic acid (44D) (27 mg5 0.08 mmol), EDC (15.5 mg; 0.081 mraol) and HOBt (11 mg, 0.081 mmol) in a mixed solvent of DMF (0.5 rnL) and CH2Cl2 (0.5 niL) was stirred for 5 min. To this mixture was then added L- Valine methyl ester hydrochloride salt (13.4 mg, 0.08 mmol) in CH2Cl2 (0.5 mL) and diisopropylethylamine (20.7 mg, 0.16 mraol). The mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was suspended in 2: 1 EtOAc/THF and washed with water (2 x 1 mL). The solvent was removed in vacuo. The obtained ester intermediate was suspended in THF (1 mL) and then 1 N aqueous LiOH solution (0.3 mL, 0.3 mmol) was added. The mixture was stirred at room temperature overnight, and then acidified with 1 N aqueous HCI. The solvent was removed in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to afford the title compound (18.5 mg, 42%). HPLC/MS (Method D): retention time = 1.39 min, [M+Hf = 437.2. 1H NMR (500 MHz, DMSO-d6) δlθ.53 (s, IH), 9.99 (d, J= 8.25 Hz, IH), 8.70 (d, J= 7.15 Hz, IH), 8.09 (d, J = 7.15 Hz, IH)5 7.83 - 7.86 (m, 2H), 7.79 - 7.82 (m, 2H), 7.27 (d, J= 3.30 Hz, IH), 7.14 (t, J= 6.87 Hz, IH), 6.95 (d, J= 3.85 Hz, IH), 4.51 (dd, J- 8.25, 4.40 Hz5 IH), 2.18 - 2.32 (m, IH), 0.97 (dd, J- 16.22, 6.87 Hz, 6H).
EXAMPLES 45 TO 58 [00152] Examples 45 to 58 were prepared by coupling 3-(4-(thiazol-2- ylammo)phenyl)-[l,2,4]triazolo[4,3-β]pyridine-8-carboxylic acid (44D) with various amino acid esters according to the procedures described in Example 44E and listed in Table 3. Analytical data for the compounds in Table 3 was reported as follows: compound retention times were recorded using LC-MS conditions (Method D), and the molecular mass of the compounds were determined by MS (ES) by the formula m/z. TABLE 3
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0002
EXAMPLE 59
(S)-3-MethyI-2-(3-(4-(2-phenylacelamido)phenyl)-[1,2,4]triazolo[4,3-α]pyridine- carboxamido)butanoic acid
Figure imgf000086_0001
Preparation 59A: 3-(4-(2-Phenylacetamido)phenyl)-[l,2,4]tτiazolo[4,3~α]pyridine-8~ carboxylic acid
Figure imgf000087_0001
[00153] To a solution of ethyl 3-(4-aminophenyl>[ 1 ,2,4]triazolo[4,3-α]pyridine-8- carboxylate (4C) (30 mg, 0.106 mmol) and 2-phenylacetyl chloride (16.34 mg, 0.106 mmol) in dichloromethane (1 mL) was added triethylamine (11.8 mg, 0.117 mmol). The reaction mixture was stirred at room temperature for 30 min, and then concentrated in vacuo. The obtained residue was stirred in mixed solvents of EtOAc (4 mL) and H2O (2 mL) at room temperature. The resulting suspension was filtered to yield the first crop of the product as a brown solid. The filtrate was separated and the isolated organic layer concentrated in vacuo to yield the second crop of the product (total 40 mg, 94% yield as an ester intermediate). [00154] To a solution of the ester intermediate in THF (2 mL) was added 2 N aqueous LiOH (0.2 mL, 0.4 mmol). The resulting mixture was stirred at room temperature for 3 h, and then acidified with 1 N aqueous HCl and concentrated in vacuo. The obtained residue was then triturated with H2O and a small amount of CH2Cl2 to yield the title compound as a light brown solid (31 mg, 78%). HPLC/MS (Method C): retention time = 1.38, [M+Hf - 373.4. 1H NMR (500 MHz, DMSO-d6): δ 10.59 (s, IH), 8.85 (d, J= 7.15 Hz, IH), 8.24 (d, J= 6.60 Hz, IH), 7.87 - 7.95 (m, 2H), 7.81 - 7.86 (m, 2H), 7.31 - 7.39 (m, 4H), 7.22 - 7.30 (m, 2H), 3.71 (s, 2H).
Example 59 [00155] A suspension of 3-(4-(2-phenylacetamido)phenyl)-[l,2>4]triazolo[4!,3- α]pyridine-8-carboxylic acid (59A) (14 mg, 0.038 mmol), EDC (9 mg, 0.047 mmol) and HOBt (6.4 mg, 0.047 mmol) in a mixed solvent of DMF (0.5 mL) and DCM (0.5 mL) was stirred for 5 rain, and then L-valine tert-bntyl ester hydrochloride (8 mg, 0.038 mmol) and triethylamine (7.6 mg, 0.075 mmol) were added. The mixture was stirred at room temperature overnight, and then concentrated in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield an ester intermediate, which was stirred in a mixed solvent of TFA (1 mL) and DCM (1 mL) at room temperature for 2.5 h. The solvent was removed by vacuo to yield the title compound as a brown solid (10.2 mg, 57%). HPLC/MS (Method C): retention time - 1.99, (M+H]+ = 472.5. 1H NMR (500 MHz, DMSOd6): δ 10.51 (s, IH), 10.02 (d, J= 8.25 Hz, IH), 8.74 (d, J= 7.15 Hz, IH), 8.15 (d, J= 6.05 Hz, IH), 7.81 - 7.94 (m, 4H), 7.30 - 7.42 (m, 4H), 7.26 (t, J = 6.60 Hz, IH), 7.20 (t, J- 7.15 Hz, IH), 4.57 (dd, J = 8.25, 4.40 Hz, IH), 3.71 (s, 2H), 2.24 - 2.39 (m, IH), 1.02 (dd, J= 16.22, 6.87 Hz, 6H).
EXAMPLE 60
(5)-iV~(l-Hydroxybutan-2--yl)-3-(4-(2-phenylacetaniido) phenyl)-[l,2,4]triazolo[4>3- α]pyridme-8-carboxamide
Figure imgf000088_0001
[00156] A suspension of 3-(4-(2-phenylacetamido)phenyl)-[l,2,4]triazolo[4,3- a]pyridine-8-carboxylic acid (59A) (14 mg, 0.038 mmol), EDC (9 mg, 0.047 mmol) and HOBt (6.4 mg, 0.047 mmol) in a mixed solvent of DMF (0.5 mL) and DCM (0.5 mL) was stirred for 5 min, and then (S)-2-aminobutan-l~ol hydrochloride salt (5.6 mg, 0.045 mmol) and triethylamine (7.6 mg, 0.075 mmol) were added. The mixture was stirred at room temperature overnight. The solvent was removed in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield the title compound (8.3 mg, 50%). HPLC/MS (Method C): retention time = 1.75, [M+Hf - 444.5. 1H NMR (500 MHz, DMSO-(I6): δ 1.0.50 (s, IH), 9.58 (d, J = 8.25 Hz, IH), 8.71 (d, J = 7.15 Hz, IH), 8.14 (d, J= 6.05 Hz, IH), 7.80 - 7.92 (m, 4H), 7.30 - 7.43 (m, 4H), 7.26 (t, J- 6.60 Hz, IH), 7.19 (t, J= 6.87 Hz, IH), 3.93 - 4.07 (m, IH), 3.71 (s, 2H), 3.54 - 3.63 (m, IH), 3.50 (dd, J= 11.0 Hz, 4.95 Hz, IH), 1.68 - 1.78 (m, IH), 1.54 - 1.65 (m, IH), 0.87 - 1.06 (m, 3H). EXAMPLE 61
(5)"3-Methyl-2-(3-(4-(3-(2-(lrifluoromethoxy)phenyl) ureido)phenyI)" [1 ,2,4]triazolo[4,3-a]pyrazine-8-carboxamido)butanoic acid
Figure imgf000089_0001
Preparation 6 IA: 2-Chloro-3~hydrazinylpyrazine
Figure imgf000089_0002
[00157] A mixture of 2,3-dichloropyrazine (6.26 g, 42.0 mmol) and anhydrous hydrazine (5 mL, 159 mmol) in pyridine (22 mL, 272 mmol) was stirred at 6O°C for 2.5 h. After cooling Io room temperature, the reaction mixture was concentrated in vacuo. The obtained residue was triturated with water. The resulting slurry was filtered, and the collected solid air dried to yield the first crop of the title compound (3.88 g) as a white solid. The filtrate was concentrated in vacuo, the obtained residue was treated by repeating above process to afford additional amount of the product (1.73 g). (total 5.61 g, 92% yield). HPLC/MS (Method C): retention time - 0.30 min> [M+Hf = 144.8. 1H NMR (500 MHz, DMSOd6): δ 8.27 (s, IH), 8.05 (s, IH), 7.56 (s, IH), 4.32 (s, 2H); 13C NMR (500 MHz, DMSO-d6) δ 152.62, 140.67, 132.60, 130.01.
Preparation 6 IB: tert-Butyl 4-(2-(3-chloropyrazin-2-yl)hydrazinecarbonyl) phenyl carbamate
Figure imgf000089_0003
[00158] A suspension of 4-(terϊ-butoxycarbonylamino)benzoic acid (1.642 g, 6.92 mmol), EDC (1.33 g, 6.92 iranol) and HOBt (935 mg, 6.92 mmol) in a mixed solvent of DMF (10 mL) and dichloromethane (10 mL) was stirred until it became a clear solution, and then 2-chloro-3-hydrazinylpyrazine (61A) (1.0 g, 6,92) was added followed by triethylamine (1.4 g, 13.82 mmol). The resulting mixture was stirred at room temperature overnight. The solvent was removed in vacuo. The obtained residue was stirred with water (25 mL) and ethyl acetate (40 mL). The resulting suspension was filtered, and the collected solid dried in a vacuum oven to yield the title compound as a brown solid (2.395 g, 95%). HPLC/MS (Method C)-: retention time = 1.89 min, [M+H]+ = 364.0. 1H NMR (500 MHz, DMSO-(J6): δ 10.35 (s, IH), 9.68 (s, IH), 9.08 (s, IH), 8.08 (d, J = 2.20 Hz, IH), 7.83 (d, J= 8.80 Hz, 2H), 7.77 (d, J = 2.75 Hz, IH), 7.55 (d, J= 8.80 Hz, 2 H), 1.48 (s, 9H).
Preparation 61C: tert-Butyl 4-(8-chloro-[l,2,4]triazolo[4,3-α]pyrazin-3-yl) phenylcarbamate
Figure imgf000090_0001
[00159] To a solution of tert-butyl 4-(2-(3~chloropyrazin-2 yl)hydrazinecarbonyl) phenyl carbamate (61B) (1.96 g, 5.39 mmol) in a mixed solvent of CCU (11 mL), THF(16 mL) and CH2Cl2 (22 mL) cooled to O°C under argon was added DlPEA (6.96 g, 53.9 mmol), followed by dropwise addition of triethylphosphine (3.18 g, 26.9 mmol). The reaction mixture was stirred at O°C for 1 h and then quenched with addition of water (55 mL). The product was extracted with ethyl acetate (3 x). The combined organics were washed with brine, dried over anhydrous Na2 SO4, and then concentrated in vacuo to yield a brown solid. This crude product was triturated with CH2CHVEtOH (4 mL/8 mL). The resulting suspension was filtered to yield the first crop of the title compound as a white solid (790 mg).The filtrate was concentrated and the obtained residue purified using a silica gel cartridge, eluting with EtOAc/CH2Cl2 to yield additional amount of the product as an off-white solid (550 mg) (total amount: 1.34 g, 72% yield). HPLC/MS (Method C): retention time = 2.06 min, [M+H]+ = 346.1. 1H NMR (500 MHz, DMSO-Cl6): δ 9.75 (s, IH), 8.62 (d, J= 4.95 Hz, IH), 7.84 (d, J = 8.80 Hz, 2H), 7.68 - 7.77 (m, 3H), 1.47 (s, 9H). 13NMR (500 MHz, DMSO-d6): 152.86, 148.80, 143.76, 142.16, 142.32, 129.06, 128.72, 118.87, 118.46, 117.84, 79.84, 28.28.
Preparation 61D: Methyl 3-(4-( tert-butoxycarbonylamino)phenyl)-[l,2,4]triazolo[4,3-α]pyrazine-8 -carboxyl ate
Figure imgf000091_0001
[00160] Carbon monoxide gas was charged into a pressure bottle containing a mixture of tert-butyl 4-(8-chloro-[1,2,4]triazolo[4,3-α]pyrazin-3-yl)phenylcarbamate (61C) (714 mg, 2.065 mmol), Pd(OAc)2 (185 mg, 0.826 mmol), dppp (341 mg, 0.826 mmol), Et3N (1.15 mL, 8.26 mmol), DMSO (12 mL) and MeOH (6 mL) until the pressure reached to 30 psi. The bottle was then sealed and heated at 75°C for 4 h. After cooling to room temperature, the reaction was diluted with EtOAc, washed with water (3 x), saturated aqueous NaCl, dried (Na2SO4) and concentrated under reduced pressure. The crude product was purified using a silica gel cartridge eluting with a gradient of EtOAc in hexanes to afford 103 mg (17%) of the title compound. HPLC/MS (Method C): retention time = 1.72 min, [M+H]+ = 370.4; 1H NMR (500 MHz, DMSO-d6): δ 9.75 (s, IH), 8.78 (d, J = 4.95 Hz, IH), 8.04 (d, J = 4.95 Hz, IH), 7.86 (d, J = 8.80 Hz, 2H), 7.73 (d, J= 8.80 Hz, 2H), 4.02 (s, 3H), 1.47 (s, 9H).
Preparation 61E: Methyl 3-(4-aminophenyl)-[1,2,4]triazolo[4,3-α]pyrazine-8- carboxylate
Figure imgf000091_0002
[00161] To a solution of methyl 3-(4-( tert-butoxycarbonylamino)phenyl)- [1,2,4]triazolo [4,3-α]pyrazine-8-carboxylate (61D) (130 mg, 0.35 mmol) in CH2Cl2 (1 mL) was added TFA (1 mL). The resulting mixture was stirred at room temperature for 2 h, and then concentrated in vacuo. The obtained residue was stripped with CH2Cl2 and dried in high vacuum to yield the title compound as brown oil (94 mg). HPLC/MS (Method D): retention time - 0.39 min, [M+H]+ = 270.5.
Preparation 61F: Methyl 3-(4-(3-(2-(trifluoromethoxy)phenyl)ureido)phenyl)- [l,2,4]triazolo[4,3-α]pyrazine-8-carboxylate
Figure imgf000092_0001
[00162] To a solution of methyl 3-(4-aminophenyl)-[l,2,4]triazolo[4,3-α]pyrazine- 8-carboxylate (61E) (94 mg, 0.35 mmol) in anhydrous THF (1.5 mL) was added 2- trifluoromethylphenyl isocyanate (71 mg, 0.35 mmol). The mixture was stirred at 7O°C overnight. After cooling to room temperature, the mixture was concentrated. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield the title compound (32 mg, 19%). HPLC/MS (Method D): retention time = 2.09 min, [M+Hf = 473.3; 1H NMR (500 MHz, DMSOd6): δ 9.64 (s, IH), 8.81 (d, /= 4.95 Hz, IH), 8.59 (s, IH), 8.27 (d, J= 7.15 Hz, IH), 8.06 (d, J- 4.95 Hz, IH), 7.91 (d, J= 8.80 Hz, 2H), 7.75 (d, J= 8.80 Hz, 2H), 7.33 - 7.43 (m, 2H), 7.09 - 7.16 (m, IH), 4.03 (s, 3H).
Preparation 61G: 3-(4-(3-(2-(Trifluoromethoxy)phenyl)ureido)phenyl)-[l,2,4] triazolo[4,3→ϊ]pyrazine-8-carboxylic acjd
Figure imgf000092_0002
[00163] To a solution of methyl 3~(4~(3-(2-(trifluoromethoxy)phenyl)ureido) phenyI)-[l,2,4]triazoIo[4,3-α]pyrazine-8-carboxylate (61F) (72 mg, 0.152 mmol) in THF (3 mL), was added potassium trimethylsilanolate (78 mg, 0.61 mmol). The mixture was stirred at room temperature for 70 mm, and then acidified with 1 N aqueous HCl (0.62 mL). The mixture was diluted with EtOAc, washed with water (3 x), brine, and dried over Na2SO4. The solvent was removed in vacuo to yield the title compound as a brown solid (70 mg, 100%). HPLC/MS (Method C): retention time = 1.83 min, [M+H]+ = 459.4.
Example 61 [00164] A suspension of 3-(4-(3-(2-(trifluoromethoxy)phenyl)ureido)phenyl)- [1,2,4] triazolo[4,3-α]pyrazine~8-carboxylic acid (61G) (35 mg, 0.076 mmol), EDC (15.4 mg, 0.08 mmol) and HOBt (11 mg, 0.08 mmol) in a mixed solvent of DMF (0.5 niL) and CH2CI2 (1 raL) was stirred until it became a clear solution, and then L-valine tert-bυtyl ester hydrochloride salt (16.8 mg, 0.08 mmol) and triethylamine (16 mg, 0.16 mmol) were added. The mixture was stirred at room temperature for 2 h, and then concentrated in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield a brown solid (31 mg, 66%) as a methyl ester intermediate. HPLC/MS (Method C): retention time = 3.02 min, [M+H]+ - 614.6. [00165] A solution of the ester intermediate (31 mg, 0.05 mmol) in a mixed solvent of TFA (1 niL) and CH2CI2 (1 mL) was stirred at room temperature for 4 h, and then concentrated in vacuo to yield the title compound as a brown solid. HPLC/MS (Method C): retention time = 2.27 min, [MH-H]+ = 558.5. 1H NMR (500 MHz3 DMF- d7): δ 9.78 (s, ΪH), 9.75 (d, J= 8.25 Hz, IH), 8.91 (d, J- 4.95 Hz, IH), 8.72 (s, IH), 8.43 (d, J = 6.60 Hz, IH), 8.18 (d, J- 4.95 Hz, IH), 8.00 (m, 2H), 7.87 (d, J= 8.80 Hz, 2H), 7.43 (m, 2H), 7.09 - 7.24 (m, IH), 4.74 (dd, J= 8.52, 4.67 Hz, IH), 2.42 (d, J= 4.95 Hz, IH), 1.11 (dd, J- 7.15, 4.95 Hz, 6H).
EXAMPLE 62 (5)-l-(3-(4-(3-(2»(Trifluoromethoxy)phenyl)ureido) phenyl)-[lJ2J4]triazolo[4,3- a]pyrazine-8-carbonyl)pyrrolidine-2-carboxylic acid
Figure imgf000093_0001
[00166] The title compound was prepared from 3-(4-(3-(2- (trifluoromethoxy)phenyl) ureido)phenyl)-[ 1 ,2,4] triazolo[4,3-a]pyrazine-8-carboxylic acid (61G) and L-proline tert-bvtiyl ester by analogous procedure described in Example 61H. HPLC/MS (Method C): retention time = 1.91 - 2.01 min (broad rotomer), [M+H]+ = 556.4.
EXAMPLE 63 (5)-2-(3-(4-(6-ChlorobenzoMthiazol-2-ylamino)phenyl)-[l,2,4]triazolo[4,3- fl]pyrazine-8-carboxamido)-3 -methylbutanoic acid
Figure imgf000094_0001
Preparation 63 A: 4-(6-Chlorobenzo[J]thiazol-2-ylamino)benzoic acid
Figure imgf000094_0002
[00167] To a suspension of tert-bntyl 4-aminobenzoate (6.36 g, 32.9 mmol) and 2,6-dichlorobenzothiazole (5 g, 24.5 mmol) in isopropanol (150 mL) was added 4 N HCl in 1 ,4-dioxane (4 mL, 16 mmol). The mixture was refluxed at 100°C for 7 h, and then allowed to cool to room temperature. White precipitates formed during cooling were isolated by filtration to yield the first crop of the title compound as a white solid (3.13 g). The filtrate was concentrated in vacuo and then stirred in a mixed solvent of TFA (20 mL) and CH2Cl2 (20 mL) overnight. The solvent was removed in vacuo, and the residue was triturated with isopropanol/ethyl acetate. The isolated white solid was stirred in a mixed solvent of isopropanol (12 mL) and ethyl acetate (S mL) for 0.5 h and collected by filtration to yield the second crop of the title compound (3.5 g) (total 6.63 g, 85 % yield). HPLC/MS (Method C): retention time =
Figure imgf000094_0003
2.51 min, 305.3. 1H NMR (500 MHz, DMSOd6) δ 11.08 (bs, IH)5 7.98 (d, J= 2.20 Hz, IH)5 7.94 (m, 2H), 7.88 (m, 2H), 7.64 (d, J= 8.25 Hz, 3H), 7.36 (dd, J= 8.80, 2.20 Hz, IH). Preparation 63B: 4-(6-Chlorobeiizo[(f|thiazol-2-ylamino)-N'-(3-chloropyrazin-2- yl)benzohydrazide
Figure imgf000095_0001
[00168] A suspension of 4-(6-chlorobenzo[</jthiazol-2-ylamino)benzoic acid (63A) (6.62 g, 21.72 mmol), EDC (4.37 g, 22.81 mmol) and HOBt (3.08 mg, 22.81 mmol) in a mixed solvent of DMF (25 mL) and CH2C^ (50 mL) was stirred for 5 min, and then 2-chloro-3-hydrazinylpyrazine (61A) (3.14 g, 21.72 mmol) and triethylamine (4.4 g, 43.4 mmol) were added. The mixture was stirred at room temperature over the weekend. Additional EDC (1.5 g, 7.85 mmol) and HOBt (1.15 g, 8.45 mmol) were added in and the reaction mixture stirred for another 2 h before concentrated in vacuo. The obtained residue was partitioned between EtOAc (250 mL) and water (200 mL), and the separated EtOAc layer washed with water (2 x 200 mL), and concentrated in vacuo. The crude product was triturated with water (50 mL) and EtOAc (20 mL) and isolated by filtration, rinsed with water and then air dried to yield the title compound as a dark brown solid (6.17 g). The second crop of the product was isolated from the concentrated mother liquor using the same trituration process (0.76 g) (total 6.93 g, 74% yield). HPLC/MS (Method C): retention time = 2.43 min, [M+H]+ = 431.1. 1H NMR (500 MHz, DMSO-d6) δ 10.90 (s, IH), 10.41 (s, IH), 9.12 (s, IH), 8.09 (d, J = 2.20 Hz, IH), 7.99 (d, J= 2.20 Hz, IH), 7.91 - 7.97 (m, 2H), 7.85 - 7.90 (m, 2H)5 7.78 (d, J= 2.75 Hz5 IH), 7.65 (d, J= 8.80 Hz, IH), 7.37 (dd, J= 8.8O5 2.20 Hz, IH).
Preparation 63C: 6-Chloro-N-(4-(8-chloro-[l ,2,4]triazolo[4,3~α]pyrazin~3~ yl)phenyl)benzo[</]thiazol-2-amine
Figure imgf000095_0002
[00169] To a solution of 4-(6-chlorobenzo[cf]thiazol-2-ylamino)-N'-(3- chloropyrazin~2-yl)benzohydrazide (63B) (6.17 g, 14.31 mmol) in a mixed solvent of CCl4 (60 mL), THF (80 mL) and CH2Cl2 (120 mL) cooled to O°C under argon was added DlPEA (18.49 g, 143 mmol), followed by dropwise addition of triethylphosphine (8.45 g, 71.5 mmol). After addition, the reaction mixture was stirred at O°C for 1 h, and then quenched by addition of water (100 mL). The precipitate was isolated by filtration and air dried to yield the first crop of the title compound as a dark brown solid (3.92 g). The filtrate was extracted with ethyl acetate (2 x). The combined organics were washed with water (1 x 100 mL), dried over anhydrous Na2SO4 and concentrated in vacuo. The obtained residue was triturated with CH3OH/CH2C12 (35 mL/5 mL) to yield the second crop of the title compound (0.95 g) (total 4.87 g, 82% yield). HPLC/MS (Method C): retention time = 2.61 min5 [MH-H]+ = 413.3. 1H NMR (500 MHz, DMSO-d6): δ 10.98 (s, IH), 8.68 (d, J = 4.95 Hz, IH), 8.05 (d, J= 8.25 Hz, 2H), 7.98 (s, IH)5 7.96 (d, J- 8.25 Hz, 2H), 7.77 (d, /= 4.95 Hz, IH), 7.63 (d, J= 8.25 Hz5 IH)5 7.37 (d, J= 8.80 Hz, IH).
Preparation 63D: Methyl 3-(4-(6-chlorobenzo[<fjthiazol~2-ylamino)phenyl)- [ 1 ,2,4]triazolo[4,3-α]pyrazme-8-carboxylate
Figure imgf000096_0001
[00170] Carbon monoxide gas was charged into a pressure bottle containing a mixture of 6-chloro-N-(4-(8-chlorθ"[l,254]triazolo[453-α]pyrazin-3- yl)phenyl)benzo[tf]thiazol-2-amine (63C) (3.0 g, 7.26 mmol), Pd(OAc)2 (650 mg, 2.90 mmol), dppp (1.2Og, 2.90 mmol), Et3N (2.94 g, 29.0 mmol), DMSO (40 mL) and MeOH (20 mL) until the pressure reached to 30 psi. The bottle was then sealed and heated at 70°C for 2 h. The reaction mixture was cooled to room temperature, and the precipitate was collected by filtration to yield the title compound as a greenish solid (1.25 g, 40%). HPLC/MS (Method C): retention time = 2.22 min, [M+H]+= 437.4. 1H NMR (SOO MHz, DMSO-d6): θ 10.96 (s, IH), 8.84 (d, J = 4.95 Hz, IH), 8.02 - 8.14 (m, 3H), 7.94 - 8.01 (m, 3H), 7.64 (d, J = 8.25 Hz, IH), 7.38 (dd, /= 8.80, 2.20 Hz, IH), 4.03 (s, 3H).
Preparation 63E: 3~(4~(6-Chlorobenzo[uTjthiazol-2-ylamino)pherjyl)-[l,2,4] triazolo[4,3-ώ]pyrazme-8-carboxylic acid
Figure imgf000097_0001
[00171] To a solution of methyl 3-{4-(6-chlorobenzo[d]thiazol-2-ylamino)phenyl)- [l,2,4]triazoio[4,3-α]pyrazine-8-carboxylate (63D) (1.127 g, 2.58 mmol) in THF (150 mL) was added potassium trimethylsilanolate (881 mg, 6,87 mmol). The mixture was stirred at room temperature for 1 h. The insoluble material was collected by filtration and discarded. The filtrate was diluted with water (80 mL) and concentrated in vacuo to remove most of the THF. The remaining aqueous material was neutralized by addition of saturated aqueous NH4Cl solution. The resulting precipitate was filtered, rinsed with water and methanol, and air dried to yield the title compound as a brown solid (1.025 g, 94%). HPLC/MS (Method C): retention time = 1.98 min, [M+Hf = 423.4. 1H NMR (500 MHz, DMSOd6): δ 8.40 (d, J= 4.40 Hz5 IH), 8.03 (d, J = 8.25 Hz, 2H), 7.99 (s, IH), 7.94 (d, J= 8.25 Hz, 2H), 7.71 (d, J= 4.95 Hz, IH)5 7.64 (d, J = 8.80 Hz, IH), 7.37 (d, J- 8.25 Hz, IH).
Example 63 [00172] A suspension of 3-(4-(6-chlorobenzo[W]thiazol-2-ylamino)phenyI)- [l,2,4]triazolo [4,3-α]pyridine-8-carboxylic acid (63E) (60 mg, 0.14 mmol), EDC (27 mg, 0,14 mmol) and HOBt (19 mg, 0.14 mmol) in a mixed solvent of DMF (1 mL) and DCM (1 mL) was stirred until it became a clear solution, and then L-valine tert- butyl ester hydrochloride salt (30 mg, 0.12 mmol) and triethylamine (29 mg, 0.28 mmol) were added. The mixture was stirred at room temperature for 2 h. Analysis by HPLC indicated the starting acid was not consumed. Additional amount of EDC (15 mg), HOBt (10 mg) and L-valine rerf-butyl ester hydrochloride salt (10 mg) were added and the reaction mixture stirred for another 1 h. The reaction was concentrated in vacuo. The crude product was purified by preparative HPLC using CH3CN/H2O/TFA solvent system to yield the product (a tert-butyl ester intermediate, 10 mg) as a brown solid. HPLC purity: 95%; LCMS: [M+H]f = 578.5. [00173] A solution of the tert-butyl ester intermediate (10 mg) in a mixed solvent of TFA (0.5 mL) and DCM (0.5 mL) was stirred at room temperature for 4 h. The reaction mixture was concentrated in vacuo to yield the title compound as a brown solid (7.7 mg). HPLC/MS (Method C): retention time = 2.42 min, [M+Hf= 522.4. 1H NMR (500 MHz, DMF-d7): δ 9.78 (d, J- 8.25 Hz, IH), 8.97 (d, J = 4.95 Hz, IH), 8.20 - 8.24 (m, 2H), 8.11 (d, J = 8.80 Hz, 2H), 8.06 (d, J- 2.20 Hz, IH), 7.70 - 7.85 (m, IH), 7.47 (dd, J= 8.80, 2.20 Hz, IH), 4.77 (dd, J= 8.52, 4.67 Hz, IH), 2.36 -
2.49 (ra, IH), 1.06 - 1.31 (m, 6H).
EXAMPLE 64 (5)-l-(3-(4-(6-Chlorobenzo[Othiazol-2-ylamrao)phenyl)-[lJ2,4]triazolo[4,3- α]pyrazine~8~carbonyl)pyrrolidine-2-carboxylic acid
Figure imgf000098_0001
[00174] The title compound was prepared from 3-(4-(6-chlorobenzo[rf]thiazol-2- ylamino)phenyl)-[ 1,2,4] triazolo[4,3-β]pyrazine-8-carboxylic acid (63E) and L- proline tert-butyl ester by analogous procedure described in Example 63F. HPLC/MS (Method C): retention time = 2.03 - 2.16 min (broad rotomer), [M+H]+ = 520.4. !H NMR (500 MHz, DMSOd6): δ 10.97 (s, IH), 8.66 - 8.80 (m, IH), 8.03 - 8.10 (m, 2H)1 7.94 - 8.02 (m, 4H), 7.65 (d, J= 8.80 Hz, IH), 7.38 (dd, J = 8.25, 2.20 Hz, IH),
4.50 - 4.66 (m, IH), 3.42 - 3.78 (m, 2H), 2.19 - 2.39 (m, IH), 1.81 - 2.05 (in, 3H).
EXAMPLE 65 (S)-3 -(4-(6-Chlorobenzo [J]thiazol-2-ylamino)phenyl)-N-( 1 -hydroxybutan-2-yl)- [ 1 ,2,4]triazolo[4,3-«]pyrazine-8-carboxamide
Figure imgf000099_0001
[00175] A suspension of 3-(4-(6-chlorobenzo[rf]thiazol-2-ylamino)phenyl)- [l,2,4]triazolo [4,3-αjpyridine-8-carboxylic acid (63E) (43 mg, 0.1 mmol), EDC (24.4 mg, 0.127 mmol) and HOBt ( 17.2 mg, 0.127 mmol) in DMF ( 1 mL) was stirred for 5 min, and then (S)-2-aminobutan-l-ol, hydrochloride salt (13 mg, 0.104 mmol) and triethylamine (20.6 mg, 0.203 mmol) were added. The mixture was stirred at room temperature overnight. To the reaction mixture was added anhydrous DCM (1 mL), followed by additional EDC (25 mg) and HOBt (17 mg). The resulting mixture was then stirred for additional 2 h before concentrated in vacuo. The crude product was purified by preparative HPLC using MeOH/H2O/TFA solvent system to yield the title compound (3 mg). HPLC/MS (Method C): retention time = 2.14 rnin (broad rotomer), [M+H]+ - 494.1. 1H NMR (500 MHz, DMSO~d6): δ 10.97 (s, IH), 8.98 (d, J= 8.25 Bz, IH), 8.77 (d, /= 4.95 Hz, IH), 8.02 - 8.09 (m, 3H), 7.93 - 8.01 (m, 3H), 7.64 (d, J= 8.80 Hz, IH), 7.38 (dd, / = 8.80, 2.20 Hz, IH)5 3.94 - 4.03 (m, IH), 3.57 (dd, J= 10.72, 4.67 Hz, IH), 3.48 (dd, J= 11.00, 5.50 Hz, IH), 1.48 - 1.61 (m, IH). 0.89 - 1.02 (m, 3H).
EXAMPLE 66 (5)-3-Methyl-2-(3-(4-(3-(2-(tτifluoromethyl)phenyl) ureido)phenyl)- [l,2j4]triazolo[4,3-δ]pyridazine-8-carboxamido)butanoic acid
Figure imgf000099_0002
Preparation 66A: tert-Butyl 3,6-dichloropyridazine-4-carboxylate
Figure imgf000100_0001
[00176] A mixture of 3, 6-dichloropyridazine-4-carboxylic acid (5.79 g, 30 mmol), tert-butsnol (2.45 g, 33 mmol), 2-chloro-l-methylpyridinium iodide (6.12 g, 24 mmol), and tributylamine (8.9 g, 48 mmol) in dichloromethane (52 mL) was stirred at refluxing under N2 for 3h. After cooling to room temperature, the reaction mixture was concentrated under reduced pressure. The residue was purified using a silica gel cartridge (330 g) which was pretreated with 1% Et3N in hexanes for 10 min at 10 mL/min, eluting with a gradient of EtOAc (0-50%) in hexanes to afford 6.1 g (81%) of the title compound as a white solid. HPLC/MS (Method B): retention time = 2.95
Figure imgf000100_0002
.
Preparation 66B: tert-Butyl 6-chloro~3~hydrazmyϊpyridazine-4-carboxylate
Figure imgf000100_0003
[00177] To a solution of tert~buϊy\ 3,6-dichloropyridazine-4-carboxylate (66A) (3.9 g, 15.6 mmol) in EtOH (20 mL) was added anhydrous hydrazine (2 mL). The reaction mixture was stirred at room temperature for 2 h, and then concentrated under reduced pressure. The crude product was triturated with 80% EtOH in H2O to afford 1.58g
(40%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 1.77 min, [M+H-56]f - 189.3.
Preparation 66C: tert-Butyl 3-(2-(4-(terf-butoxycarbonylamino)benzoyl) hydrazinyl)-
6-chIoropyridazine~4~carboxylate
Figure imgf000101_0001
[00178] To a solution of tert-butyl ό-chϊoro-S-hydrazinylpyridazine^-carboxylate (66B) (800 mg, 3.27 mmol) in anhydrous DMF (5 mL) was added A-(tert- butoxycarbonyl- amine)benzoic acid (LO g, 4.25 mmol), EDC (940 mg, 4.91 mmol), HOBt (663mg, 4.91 mmol), followed by DIPEA (1.1 mL, 6.54 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was partitioned between EtOAc and water, the separated EtOAc layer washed with water, saturated aqueous NaHCOj5 saturated aqueous NaCl (2 x), dried (anhydrous NaZSO4), and concentrated. The obtained residue was dried in high vacuum for 1.5 h to afford 1.49g (98%) of the title compound. HPLC/MS (Method B): retention time - 3.40 min, [M+Hf = 464.3.
Preparation 66D: tert-Butyl 3-(4-(ter/-butoxycarbonylarnino)phenyϊ)-6-chloro- [l,2,4]triazolo[4,3-Z>]pyridazine-8-carboxylate
Figure imgf000101_0002
[00179J To a solution of tert-butyl 3-(2-(4-(terϊ-butoxycarbonylamino)benzoyl) hydrazinyl)-6-chloropyridazine-4-carboxylate (66C) (1.49 g, 3.20 mmol) in a mixed solvent of THF (10 mL) and CCl4 (5 mL) at O°C under argon was added Et3P (3.35 mL, 19.2 mmol), followed by DIPEA (3.35 mL, 19.2 mmol). The reaction mixture was stirred at 0°C for 10 min. Analysis by LC/MS indicated the starting material was consumed. The reaction mixture was partitioned between EtOAc and water, the separated EtOAc layer washed with water, saturated aqueous NaHCO3, saturated aqueous NaCl (2 x), dried (anhydrous Na2 SO4), and concentrated. The crude residue was purified using a silica gel cartridge (40 g) which was pretreated with 1% Et3N in hexanes for 10 min at 10 mL/min, eluting with a gradient of EtOAc (0-50%) in hexanes to afford 1.02 g (71%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 3.78 min, [M+H]+ = 446.3.
Preparation 66E: f erf-Butyl 3-(4-(tert-butoxycarbonylarm'no) phenyl)- [ 1 ,2,4]triazolo[4,3-&]pyridazine-8-carboxylate
Figure imgf000102_0001
[00180] A solution of terϊ-butyl 3-(4-(tert-butoxycarbonylamJno)phenyl)-6-chloro- [l,2,4]triazolo[4,3-£]pyridazine-8-carboxylate (66D) (300 mg, 0.67 mmol) in a mixed solvent of EtOH (4 mL) and MeOH (1 mL) was bubbled through argon for 5 mins and then NaOAc (66.2 mg, 0.81 mmol) was added, followed by 5% Pd/C (50 mg). The vessel was evacuated and flashed with H2 gas three times and stirred under H2 (1 atm) for 1 h. Analysis by LC/MS indicated the starting material was consumed. The reaction mixture was filtered, and the filtrate concentrated under reduced pressure.
The obtained crude residue was purified using a silica gel cartridge (40 g) eluting with a gradient of EtOAc (0-100%) in hexanes to afford 150 mg (54%) of the title compound as a yellow foam. HPLC/MS (Method B): retention time = 3.02 min, [M+H)+ = 412.3.
Preparation 66F: tert-Butyl 3-(4-ammophenyl)-[l,2,4]triazolo[4,3-6]pyridazine-8- carboxylate
Figure imgf000102_0002
[00181] To a solution of tert-butyl 3-(4-(tert-butoxycarbonylamino)phenyl)-[l ,2,4] triazolo[4,3-έ]pyridazine-8-carboxylate (66E) (100 mg, 0.24 mmol) in CH2Cl2 (4 mL) was added iodotrimethylsilane (86 μL, 0.6 mmol) dropwise. After addition, the reaction mixture was stirred at room temperature for 10 min. Analysis by LC/MS indicated the starting material was consumed. The reaction mixture was partitioned between CH2Cl2 and water, then the separated CH2CI2 layer was washed with water, saturated aqueous NaHCO3, saturated aqueous NaCl, dried (anhydrous Na2SO4), and concentrated. The obtained crude residue was purified using a silica gel cartridge (12 g) eluting with a gradient of EtOAc (50- 100%) in hexanes to afford 60 rag (79%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 2.03 rain, [MH-H]+ = 312.3.
Preparation 66G: tøt-Butyl 3-(4-(3-(2-(trifluoromethyl)phenyl)ureido)phenyl)- [1 ,2,4]triazolo[4,3-&]pyridazine-8-carboxylate
Figure imgf000103_0001
[00182] To a suspension of tert-buty\ 3-(4-aminophenyl)-[ 1 ,2,4]triazolo[4,3- &]pyridazine-8-carboxylate (66F) (60 mg, 0.19 mmol) in THF (3 mL) was added 4- (trifluoromethyl)- phenyl isocyanate (43.3 mg, 0,23 mmol). The reaction mixture was stirred at 6O°C for 2 h before allowed to cool to room temperature. The reaction mixture was partitioned between EtOAc and water, then the separated EtOAc layer was washed with saturated aqueous NaCl, dried (anhydrous Na2SO4), and concentrated. The obtained crude residue was purified using a silica gel cartridge (12 g) eluting with a gradient of EtOAc (50-100%) in hexanes to afford 62 mg (65%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 3.52 min, [M+H]+ = 499.4.
Preparation 66H: 3-(4"(3-(2~(Trifluoromethyl)phenyl)ureido)phenyl)-[l,2,4] triazolo[4,3-&]pyridazme~8~carboxylic acid
Figure imgf000103_0002
[00183] A solution of tert-butyl 3-(4-(3-(2-(trifluoromethyI)phenyl)ureido)phenyl)- [l,2,4]triazolo[4,3-i?]pyridazine-8-carboxylate (66G) (58 mg, 0.12 mmol) in a mixed solvent of TFA (0.5 mL) and CH2CI2 (0.5 mL) was stirred at room temperature for 3 h. Analysis by LC/MS indicated the starting material was consumed. The reaction was concentrated under reduced pressure, chased with CH2CI2 (2 x) and dried in high vacuum for 2 h to afford 51 mg of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 2. S 1 min> [M+H]+ = 443.3.
Preparation 661: (S)-tert-Buty\ 3-methyl-2-(3-(4-(3-(2-(trifluoromethyl)phenyl) ureido)phenyl)-[l,254]triazolo[4>3-6]pyridazine-8-carboxamido)butanoate
Figure imgf000104_0001
[00184] To a solution of 3"(4-(3-(2-(trifluororaethyl)phenyl)ureido)phenyl)-[ 1 ,2,4] triazolo[4,3-έ]pyridazine-8-carboxylic acid (66H) (38 mg, 0.086 mmol) in anhydrous DMF (1 mL) was added L~valine terf-butyl ester hydrochloride (23.4 mg, 0.11 mmol), EDC (24.7 mg, 0.13 mmol), HOBt (17 mg, 0.13 mmol), followed by DIPEA (45 μL, 0.26 mmol). The reaction mixture was stirred at room temperature for 3 h. Analysis by LC/MS indicated the starting material was consumed. The reaction mixture was partitioned between EtOAc and water, then the separated EtOAc layer was washed with water, saturated aqueous NaHCO3, saturated aqueous NaCl, dried (anhydrous Na2SO4), and concentrated. The obtained crude residue was purified using a silica gel cartridge (12 g) eluting with a gradient of EtOAc (0-100%) in hexanes to afford 33 mg (63%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 4.04 min, [M+H]+ = 598.4.
Example 66 [00185] A solution of (S>tert-butyl 3-methyl-2-(3-(4-(3-(2-(trifluoromethyl) phenyl)ureido)phenyl)-[ 1 ,2,4]triazolo[4,3-&]pyridazine-8-carboxamido)butanoate
(661) (32 mg, 0.053 mmol) in a mixed solvent of TFA (0.5 mL) and CH2Cl2 (0.5 mL) was stirred at room temperature for 2 h. Analysis by LC/MS indicated the starting eater was consumed. The reaction was concentrated under reduced pressure. Water (5 mL) was added to the residue, and the resulting yellow precipitate was collected by filtration, washed with water and dried in a 50°C vacuum oven to afford 20 mg (70%) of the title compound as a yellow solid. HPLC/MS (Method B): retention time = 3.34 min, [M+Hf = 542.4. 1H NMR (500 MHz, DMSOd6): δ 13.13 (s, IH), 9.77 (d, J = 7.72 Hz, IH), 9.71 (s, IH), 8.92 (d, J= 3.84 Hz, IH), 8.33 (d, J = 7.68 Hz, 2H), 8.21 (s3 IH), 7.95 (d, J= 7.68 Hz, IH), 7.87 (d, J= 3.32 Hz, IH), 7.61 - 7.82 (m, 4H), 7.24 - 7.34 (m, IH), 4.52-4.65 (m, IH), 2.23 - 2.39 (m, IH), 1.02 (q, J= 7.12 Hz, 6H).
BIOLOGICAL EVALUATION DGATl Enzyme Assay [00186] DGATl enzyme assays were conducted using membranes isolated from Sf9 insect cells expressing the recombinant human DGATl cDNA. The assays were conducted in 384-well plates with total volume of 25 μl at 25°C. In each assay, 300 ng of recombinant human DGATl membrane was incubated with 25 μM of 2- monooleoylglycerol and 25 μM of [3H]-stearoyl-CoA in 100 mM potassium phosphate (pH 7.4) for 30 min with various concentrations of compounds delivered in DMSO. The assay was terminated by the addition of 30 μl of Stopping Solution [50 mM HEPES, 5 mg/mL Yittrium Oxide (YOX) Polylysine SPA beads, 3.33 mg/mL Fraction V BSA, 200 μM Mercuric chloride]. The signal was measured using LEADSEBKERSM for 5 minutes. To calculate the degree of inhibition, the zero level of enzyme activity (blank) was defined by the above assay using Sf9 cell membrane uninfected with baculovirus and the 100% level of DGATl enzyme activity was defined by human DGATl assay with the vehicle DMSO. The IC50 values of inhibitors were determined by Excel-fit. [00187] Compounds described herein were tested in the DGATl enzyme assay described immediately above. The following results were obtained.
TABLE 1
Figure imgf000106_0001

Claims

CLAIMSWhat is claimed is:
1. A compound according to Formula (I):
Figure imgf000107_0001
or a pharmaceutically acceptable salt thereof: wherein:
A is hydrogen, alkyl, cycloalkyl, aryl, heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7;
L is -(CR1R2V; n is I, 2, or 3;
B is -OR5, -C(O)OR5, -OC(O)R5, or -OC(O)OR5; one of X and Y is CH or N, and the other of X and Y is CH; R1 and R2 are, independently at each occurrence, hydrogen, alkyl, cycloalkyl, aryl, and/or heterocyclyl, or one R1 and one R2 form a C3-Cγcycloalkyl or 4- to 7- membered heterocyclyl ring having one or two heteroatoms;
R3 is hydrogen or alkyl, or R3 and R1 form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; R4 is hydrogen or alkyl;
R5 is hydrogen or alkyl;
R6 is alkyl, cycloalkyl, aryl, or heterocyclyl; and
R7 is hydrogen or alkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein: each of said alkyl is substituted with 0-3 Ra; each of said cycloalkyl is substituted with 0-3 Ra; each of said aryl is substituted with 0-4 Rb; and each of said heterocyclyl is substituted with 0-4 Rb;
Ra is, independently at each occurrence, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, phenyl, imidazolyl, and/or CrC3alkoxy;
Rb is, independently at each occurrence, Ci-C4alkyl, F, Cl, Br, -CF3, -OH, -OCH3, -OCF3, -CN, -NRcRd, and/or Ci-C3alkoxy; and
Rc and Rd are, independently at each occurrence, H and/or Cj~C4alkyl, or Rc and Rd together with the nitrogen atom to which they are attached, form a 4- to 7- membered heterocyclyl ring with one or two heteroatoms.
2. The compound according to claim 1 wherein:
A is hydrogen, CrCsalkyL Cj-Cycycloalkyl, aryl, 1- or 2-ring heterocyclyl, -C(O)R6, -C(O)OR6, or -C(O)NR6R7;
R1 and R2 are, independently at each occurrence, hydrogen, Ci-Cealkyl, Cj-C7cycloalkyl, aryl, and/or 1- or 2-ring heterocyclyl, or one R1 and one R2 form a C3-C7cycloalkyl or 4- to 7-membered heterocyclyl ring having one or two heteroatoms;
R3 is hydrogen or Ci-Cealkyl, or R3 and R1 form a 4- to 7~membered heterocyclyl ring having one or two heteroatoms;
R4 is hydrogen or Ci-Q;alkyl; R5 is hydrogen or CrC6alkyl;
K6 is Ci-C^alkyl, C3-C7cycloalkyl, aryl, or 1- or 2-ring heterocyclyl; R7 is hydrogen or CrCβalkyl, or R7 and R6 together with the nitrogen atom to which they are attached form a 4- to 7-membered heterocyclyl ring having one or two heteroatoms; wherein: each alkyl is substituted with 0-3 Ra; each cycloalkyl is substituted with 0-3 Ra; each aryl is substituted with 0-4 Rb; and each heterocyclyl is substituted with 0-4 Rb.
3. The compound according to claim 2 wherein: A is hydrogen, Q^aϊkyl, phenyl, 1- or 2-ring heterocyclyl having 1- or 2-heteroaloms selected from S and N, -C(O)R6, -C(O)OR6, or -C(O)NR6R7;
R1 is hydrogen, Ci-Qalkyl, C3-C7cycloalkyl, or phenyl;
R2 is hydrogen or methyl; or R1 and R2 form a C5-C7cycloalkyl ring or 5- to 6-membered heterocyclyl ring having one or two heteroatoms;
R3 is hydrogen or C]-C4alkyl, or R3 and R1 form a 5- to 7-membered heterocyclyl ring having 1 or 2 heteroatoms;
R is hydrogen or R5 is hydrogen or
Figure imgf000109_0001
R6 is C]-C4alkyl, or phenyl; and
R7 is hydrogen or methyl; wherein: each alkyl is substituted with 0-3 Ra; each cycloalkyl is substituted with 0-3 Ra; each phenyl is substituted with 0-3 Rb; and each heterocyclyl is substituted with 0-3 Rb.
4, The compound according to claim 3 wherein: A is hydrogen, -C(O)R6, -C(O)OR6, -C(O)NHR6, thiazolyl, or benzothiazolyl;
B is -OH or -C(O)OR5;
R! is hydrogen,
Figure imgf000109_0002
C5-C7cycloalkyl, or phenyl;
R2 is hydrogen; or R1 and R2 form a Cs-C7cycloalkyl; R3 is hydrogen or C1-C^aIkVl, or R3 and R! form a 5- to 6-membered heterocyclyl ring having one heteroatom;
R4 is hydrogen or Ct-Cjalkyl;
R5 is hydrogen or
Figure imgf000109_0003
and n is 1 or 2; wherein: each alkyl is substituted with 0-2 Ra; each cycloalkyl is substituted with 0-2 Ra; each heterocyclyl is substituted with 0-2 Rb; each phenyl is substituted with 0-2 Rb; each thiazolyl is substituted with 0-2 Rb; and each benzothiazolyl is substituted with 0-2 Rb;
Ra is, independently at each occurrence, F, Cl, Br, -CF3, -OH, -OCH3, --OCF3, phenyl, and/or imidazolyl; and
Rb is, independently at each occurrence, d^alkyl, F, Cl, Br, -CF3, -OH, "OCH3, -OCF3, and/or -CN.
5. The compound according to claim 4 wherein:
A is hydrogen, -C(O)-CH2CH3, -C(O)-benzyl, -C(O)O-CH3, -C(O)O-(butyl), -C(O)NH-(trifluoromethylphenyl), -C(O)NH-(trifluorornethoxyphenyl), thiazolyl, or chlorobenzo thiazolyl ;
B is -OH, -C(O)OH, or -C(O)OCH2CH3; R! is hydrogen, C;-C4alkyl, hydroxyethyl, cyclohexyl, phenyl, or methyl substituted with phenyl, hydroxyphenyl, or imidazolyl; R2 is hydrogen; or R1 and R2 form a cyclopentyl ring; R3 is hydrogen, or R3 and R1 form a pyrroiidinyl ring; and R4 is hydrogen.
6. The compound according to claim 1 wherein said compound is:
Figure imgf000110_0001
Figure imgf000111_0001
-no-
Figure imgf000112_0001
Figure imgf000113_0001
Figure imgf000114_0001
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
or a pharmaceutically acceptable salt of any of said compounds (1) through (661).
7. A pharmaceutical composition comprising a compound according claim 1 together with a pharmaceutically acceptable carrier.
8. A method for treatment of a condition or disorder comprising administering to a patient in need thereof at least one compound according to claim 1 wherein said condition, disorder, or disease is obesity, dyslipidemia, diabetes, or atherosclerosis.
9. The method according to claim 8 wherein said disorder is obesity.
10. Use of a compound according to claim 1 in the manufacture of a medicament that is useful for the treatment of obesity, dyslipidemail, diabetes or atherosclerosis.
11. Use of a compound according to claim 1 in the manufacture of a medicament that is useful for the treatment of obesity,
PCT/US2009/039770 2008-04-11 2009-04-07 Triazolo compounds useful as dgat1 inhibitors WO2009126624A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/936,969 US8324241B2 (en) 2008-04-11 2009-04-07 Triazolo compounds useful as DGAT1 inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4414508P 2008-04-11 2008-04-11
US61/044,145 2008-04-11

Publications (1)

Publication Number Publication Date
WO2009126624A1 true WO2009126624A1 (en) 2009-10-15

Family

ID=41162217

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/039770 WO2009126624A1 (en) 2008-04-11 2009-04-07 Triazolo compounds useful as dgat1 inhibitors

Country Status (2)

Country Link
US (1) US8324241B2 (en)
WO (1) WO2009126624A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8299061B2 (en) 2009-12-10 2012-10-30 Eli Lilly And Company Inhibitors of diacylglycerol acyltransferase
RU2534804C1 (en) * 2013-08-05 2014-12-10 Общество С Ограниченной Ответственностью "Ньювак" (Ооо "Ньювак") SUBSTITUTED [1,2,4]TRIAZOLO[4,3-a]PYRIDINES, DEMONSTRATING PROPERTIES OF ANTAGONISTS OF ADENOSINE A2A RECEPTORS, AND THEIR APPLICATION
JP2016040321A (en) * 2009-07-17 2016-03-24 日本たばこ産業株式会社 Triazolopyridine compound and effect thereof as prolyl hydroxylase inhibitor and erythropoietin production inducer
RU2677697C1 (en) * 2013-12-09 2019-01-21 Юсб Байофарма Спрл Triazolopyridine derivatives as modulators of tnf activity
WO2023085931A1 (en) 2021-11-11 2023-05-19 Koninklijke Nederlandse Akademie Van Wetenschappen Hepatic organoids

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101981035B (en) 2008-02-05 2013-08-28 赛诺菲-安万特 Triazolium salts as PAR1 inhibitors, production thereof, and use as medicaments
ES2379447T3 (en) * 2008-02-05 2012-04-26 Sanofi Triazolopyridazines as inhibitors of PAR1, their preparation and their use as medicines
SI2890684T1 (en) 2012-07-24 2018-06-29 Bial-Portela & Ca, S.A. Urea compounds and their use as enzyme inhibitors
CN105793247B (en) 2013-07-24 2018-03-16 比亚尔-珀特拉和Ca股份公司 Imidazole carboxamides and its purposes as FAAH inhibitor
IL294032A (en) * 2019-12-24 2022-08-01 Carna Biosciences Inc Diacylglycerol kinase modulating compounds

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7179802B2 (en) * 2002-06-10 2007-02-20 Merck & Co., Inc. 11-beta-hydroxysteroid dehydrogenase 1 inhibitors useful for the treatment of diabetes, obesity and dyslipidemia
EP1785424A2 (en) * 2003-04-11 2007-05-16 Novo Nordisk A/S Fused 1,2,4-triazoles and pharmaceutical uses thereof

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU622330B2 (en) 1989-06-23 1992-04-02 Takeda Chemical Industries Ltd. Condensed heterocyclic compounds having a nitrogen atom in the bridgehead for use as fungicides
CN1227554A (en) 1996-08-28 1999-09-01 辉瑞大药厂 Substituted 6,5-hetero-bicyclic derivatives
WO2001083481A1 (en) 2000-04-27 2001-11-08 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
DE10038019A1 (en) 2000-08-04 2002-02-14 Bayer Ag Substituted triazolopyride (az) ine
WO2004017950A2 (en) 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents
EA200500378A1 (en) 2002-09-18 2005-08-25 Пфайзер Продактс Инк. NEW IMIDAZOL COMPOUNDS AS A TRANSFORMING GROWTH FACTOR INHIBITORS (TGF) INHIBITORS
CA2514473C (en) 2002-11-22 2008-05-27 Japan Tobacco Inc. Fused bicyclic nitrogen-containing heterocycles
EP1619948A4 (en) 2003-05-05 2007-02-14 Neurogen Corp Sustituted imidazolopyrazine and triazolopyrazine derivatives: gabaa receptor ligands
AR044152A1 (en) 2003-05-09 2005-08-24 Bayer Corp RENTAL DERIVATIVES, METHOD OF PREPARATION AND USE FOR THE TREATMENT OF OBESITY
EP1653969A4 (en) 2003-08-07 2006-12-20 Japan Tobacco Inc Pyrrolo 1,2-b pyridazine derivatives
KR20060114376A (en) 2004-01-30 2006-11-06 니뽄 다바코 산교 가부시키가이샤 Anorectic compounds
AU2005212096B2 (en) 2004-02-18 2010-12-16 Msd K.K. Nitrogenous fused heteroaromatic ring derivative
WO2006019020A1 (en) 2004-08-16 2006-02-23 Sankyo Company, Limited Substituted ureas
US20090215817A1 (en) 2004-08-18 2009-08-27 Pfizer Inc Novel Triazolopyridine Compounds for the Treatment of Inflammation
US7572807B2 (en) 2005-06-09 2009-08-11 Bristol-Myers Squibb Company Heteroaryl 11-beta-hydroxysteroid dehydrogenase type I inhibitors
US7632837B2 (en) 2005-06-17 2009-12-15 Bristol-Myers Squibb Company Bicyclic heterocycles as cannabinoid-1 receptor modulators
CN101410397A (en) 2006-03-31 2009-04-15 诺瓦提斯公司 Organic compounds
US20080064717A1 (en) 2006-05-19 2008-03-13 Rajesh Iyengar Inhibitors of diacylglycerol O-acyltransferase type 1 enzyme
CA2651979A1 (en) 2006-05-30 2007-12-06 Pfizer Products Inc. Triazolopyridazine derivatives
AU2007272009A1 (en) 2006-07-12 2008-01-17 Syngenta Limited Triazolopyridine derivatives as herbicides
EA200970931A1 (en) * 2007-04-10 2010-04-30 Х. Лундбекк А/С Analogs of Heteroarylamides
US8394823B2 (en) * 2008-04-11 2013-03-12 Bristol-Myers Squibb Company Triazolopyridine compounds useful as DGAT1 inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7179802B2 (en) * 2002-06-10 2007-02-20 Merck & Co., Inc. 11-beta-hydroxysteroid dehydrogenase 1 inhibitors useful for the treatment of diabetes, obesity and dyslipidemia
EP1785424A2 (en) * 2003-04-11 2007-05-16 Novo Nordisk A/S Fused 1,2,4-triazoles and pharmaceutical uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ARAMPATZIS ET AL.: "Comparative enzymology of 11-hydroxysteroid dehydrogenase type 1 from six species", JOURNAL OF MOLECULAR ENDOCRINOLOGY, vol. 35, 2005, pages 89 - 101 *

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2016040321A (en) * 2009-07-17 2016-03-24 日本たばこ産業株式会社 Triazolopyridine compound and effect thereof as prolyl hydroxylase inhibitor and erythropoietin production inducer
JP2019019144A (en) * 2009-07-17 2019-02-07 日本たばこ産業株式会社 Triazolopyridine compound and effect thereof as prolyl hydroxylase inhibitor and erythropoietin production inducer
US8299061B2 (en) 2009-12-10 2012-10-30 Eli Lilly And Company Inhibitors of diacylglycerol acyltransferase
RU2534804C1 (en) * 2013-08-05 2014-12-10 Общество С Ограниченной Ответственностью "Ньювак" (Ооо "Ньювак") SUBSTITUTED [1,2,4]TRIAZOLO[4,3-a]PYRIDINES, DEMONSTRATING PROPERTIES OF ANTAGONISTS OF ADENOSINE A2A RECEPTORS, AND THEIR APPLICATION
WO2015020565A1 (en) * 2013-08-05 2015-02-12 Общество С Ограниченной Ответственностью "Ньювак" (Ооо "Ньювак") Substituted [1,2,4]triazolo[4,3-a]pyridines exhibiting antagonist properties to adenosine a2a receptors, and use thereof
RU2677697C1 (en) * 2013-12-09 2019-01-21 Юсб Байофарма Спрл Triazolopyridine derivatives as modulators of tnf activity
WO2023085931A1 (en) 2021-11-11 2023-05-19 Koninklijke Nederlandse Akademie Van Wetenschappen Hepatic organoids

Also Published As

Publication number Publication date
US8324241B2 (en) 2012-12-04
US20110034468A1 (en) 2011-02-10

Similar Documents

Publication Publication Date Title
US8324241B2 (en) Triazolo compounds useful as DGAT1 inhibitors
US8394823B2 (en) Triazolopyridine compounds useful as DGAT1 inhibitors
US7037910B2 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
EP1697370B1 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
US7629342B2 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
WO2006138657A1 (en) Bicyclic heterocycles as cannabinoid-1 receptor modulators
CA2617102A1 (en) Substituted tetrahydro-1h-pyrido[4,3,b]indoles as serotonin receptor agonists and antagonists
PT1697371E (en) Azabicyclic heterocycles as cannabinoid receptor modulators
WO2005016286A2 (en) Pyrazine modulators of cannabinoid receptors
EP1644370A2 (en) Tetrahydroquinoline derivatives as cannabinoid receptor modulators
WO2006044362A2 (en) N-sulfonylpiperidine cannabinoid receptor 1 antagonists
EP1891068A1 (en) Triazolopyridine derivatives as cannabinoid receptor 1 antagonists
EP1675859A1 (en) Modulators of serotonin receptors
US7709647B2 (en) Tetrahydroquinoline cannabinoid receptor modulators
WO2006138656A2 (en) Bicyclic heterocycles as cannabinoid-1 receptor modulators
US7314882B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators
US7361766B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators
US8030306B2 (en) Azabicyclic heterocycles as cannabinoid receptor modulators
US7368458B2 (en) Bicyclic heterocycles as cannabinoid receptor modulators

Legal Events

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

Ref document number: 09730053

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 12936969

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09730053

Country of ref document: EP

Kind code of ref document: A1